Page last updated: 2024-11-08

ritonavir

Description Research Excerpts Clinical Trials Roles Classes Pathways Study Profile Bioassays Related Drugs Related Conditions Protein Interactions Research Growth Market Indicators

Description

Ritonavir: An HIV protease inhibitor that works by interfering with the reproductive cycle of HIV. It also inhibits CYTOCHROME P-450 CYP3A. [Medical Subject Headings (MeSH), National Library of Medicine, extracted Dec-2023]

ritonavir : An L-valine derivative that is L-valinamide in which alpha-amino group has been acylated by a [(2-isopropyl-1,3-thiazol-4-yl)methyl]methylcarbamoyl group and in which a hydrogen of the carboxamide amino group has been replaced by a (2R,4S,5S)-4-hydroxy-1,6-diphenyl-5-{[(1,3-thiazol-5-ylmethoxy)carbonyl]amino}hexan-2-yl group. A CYP3A inhibitor and antiretroviral drug from the protease inhibitor class used to treat HIV infection and AIDS, it is often used as a fixed-dose combination with another protease inhibitor, lopinavir. Also used in combination with dasabuvir sodium hydrate, ombitasvir and paritaprevir (under the trade name Viekira Pak) for treatment of chronic hepatitis C virus genotype 1 infection as well as cirrhosis of the liver. [Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Cross-References

ID SourceID
PubMed CID392622
CHEMBL ID163
CHEBI ID45409
SCHEMBL ID6679
MeSH IDM0028918

Synonyms (163)

Synonym
BIDD:PXR0023
BIDD:GT0387
AC-733
chembl163 ,
1,3-thiazol-5-ylmethyl n-[(2s,3s,5s)-3-hydroxy-5-[(2s)-3-methyl-2-{[methyl({[2-(propan-2-yl)-1,3-thiazol-4-yl]methyl})carbamoyl]amino}butanamido]-1,6-diphenylhexan-2-yl]carbamate
bdbm520
HY-90001
norvir
abbott 84538
5-thiazolylmethyl ((alphas)-alpha-((1s,3s-1-hydroxy-3-((2s)-2-(3-((2-isopropyl-4-thiazolyl)methyl)-3-methylureido)-3-methylbutyramido)-4-phenylbutyl)phenethyl)carbamate
hsdb 7160
5-thiazolylmethyl ((alphas)-alpha-((1s,3s)-1-hydroxy-3-((2s)-2-(3-((2-isopropyl-4-thiazolyl)methyl)-3-methylureido)-3-methylbutyramido)-4-phenylbutyl)phenethyl)carbamate
ritonavir [usan]
c37h48n6o5s2
2,4,7,12-tetraazatridecan-13-oic acid, 10-hydroxy-2-methyl-5-(1-methylethyl)-1-(2-(1-methylethyl)-4-thiazolyl)-3,6-dioxo-8,11-bis(phenylmethyl)-, 5-thiazolylmethyl ester, (5s-(5r*,8r*,10r*,11r*))-
drg-0244
RIT ,
abt-538
a-84538
2,4,7,12-tetraazatridecan-13-oic acid, 10-hydroxy-2-methyl-5-(1-methylethyl)-1-[2-(1-methylethyl)-4-thiazolyl]-3,6-dioxo-8,11-bis(phenylmethyl)-, 5-thiazolylmethyl ester, [5s-(5r*,8r*,10r*,11r*)]-
norvir (tm)
abt538
thiazol-5-ylmethyl n-[(1s,2s,4s)-1-benzyl-2-hydroxy-4-[[(2s)-2-[[(2-isopropylthiazol-4-yl)methyl-methyl-carbamoyl]amino]-3-methyl-butanoyl]amino]-5-phenyl-pentyl]carbamate
nsc693184
rtv
MLS001424063
155213-67-5
C07240
ritonavir
MLS000759541
smr000466395
NCGC00159462-02
DB00503
1SH9
1HXW
nsc-693184
ritonavir (jan/usp/inn)
D00427
norvir (tn)
abt 538
HMS2051B08
bdbm50088504
AKOS000280930
viriton
ritovir
chebi:45409 ,
viekirax
ritonavirum
empetus
ritomune
abbott-84538
1,3-thiazol-5-ylmethyl n-[(2s,3s,5s)-3-hydroxy-5-[[(2s)-3-methyl-2-[[methyl-[(2-propan-2-yl-1,3-thiazol-4-yl)methyl]carbamoyl]amino]butanoyl]amino]-1,6-diphenylhexan-2-yl]carbamate
n-[(2s,4s,5s)-4-hydroxy-1,6-diphenyl-5-{[(1,3-thiazol-5-ylmethoxy)carbonyl]amino}hexan-2-yl]-n~2~-(methyl{[2-(propan-2-yl)-1,3-thiazol-4-yl]methyl}carbamoyl)-l-valinamide
NCGC00183130-01
NCGC00159462-03
unii-o3j8g9o825
tmc 114r
ritonavir [usan:usp:inn:ban]
nsc 693184
o3j8g9o825 ,
nsc 760369
cas-155213-67-5
tox21_113431
dtxcid8028553
tox21_112969
dtxsid1048627 ,
nsc760369
nsc-760369
pharmakon1600-01502391
norvir softgel
HMS2235O10
STK634209
CCG-101007
MLS000759541-02
viekirax component ritonavir
2,4,7,12-tetraazatridecan-13-oic acid, 10-hydroxy-2-methyl-5-(1-methylethyl)-1-(2-(1-methylethyl)-4-thiazolyl)-3,6-dioxo-8,11-bis(phenylmethyl)-5-thiazolylmethyl ester (5s-(5r*,8r*,10r*,11r*))-
ritonavirum [who-ip latin]
ritonavir [hsdb]
ritonavir [ema epar]
ritonavir component of paxlovid
ritonavir [usp monograph]
2,7,10,12-tetraazatridecanoic acid, 4-hydroxy-12-methyl-9-(1-methylethyl)-13-(2-(1-methylethyl)-4-thiazolyl)-8,11-dioxo-3,6-bis(phenylmethyl)-, 5-thiazolylmethyl ester, (3s,4s,6s,9s)-
ritonavir [usp impurity]
ritonavir [who-dd]
ritonavir [orange book]
ritonavir [jan]
ritonavir [usp-rs]
5-thiazolylmethyl [(alphas)-alpha-[(1s,3s)-1-hydroxy-3-[(2s)-2-[3-[(2-isopropyl-4-thiazolyl)methyl]-3-methylureido]-3-methylbutyramido]-4-phenylbutyl]phenethyl]carbamate
paxlovid component ritonavir
ritonavir [mart.]
ritonavir component of viekirax
ritonavir component of kaletra
kaletra component ritonavir
ritonavir [mi]
ritonavir [vandf]
BRD-K51485625-001-07-6
ritonavir [who-ip]
ritonavir [inn]
4EYR
CS-0432
S1185
AB00639991-08
gtpl8804
1,3-thiazol-5-ylmethyl n-[(2s,3s,5s)-3-hydroxy-5-[[(2s)-3-methyl-2-[[methyl-[(2-propan-2-yl-1,3-thiazol-4-yl)methyl]carbamoyl]amino]butanoyl]amino]-1,6-di(phenyl)hexan-2-yl]carbamate
AB00639991-06
1,3-thiazol-5-ylmethyl n-[(2s,3s,5r)-3-hydroxy-5-[[(2s)-3-methyl-2-[[methyl-[(2-propan-2-yl-1,3-thiazol-4-yl)methyl]carbamoyl]amino]butanoyl]amino]-1,6-diphenyl-hexan-2-yl]carbamate
ritonavir & plga
NC00257
SCHEMBL6679
NCGC00159462-04
tox21_112969_1
3PRS
KS-5017
(2s,3s,5s)-5-(n-(n-((n-methyl-n-((2-isopropyl-4-thiazolyl)methyl)amino)carbonyl)valinyl)amino)-2-(n-((5-thiazolyl)methoxycarbonyl)amino)-1 .6-diphenyl-3-hydroxyhexane
(2s,3s,5s)-5-(n-(n-((n-methyl-n-((2-isopropyl-4-thiazolyl)methyl)amino)carbonyl)valinyl)amino )-2-(n-((5-thiazolyl)methoxycarbonyl)amino)-1,6-diphenyl-3-hydroxyhexane
n1-((1s,3s,4s)-1-benzyl-3-hydroxy-5-phenyl-4-{[(1,3-thiazol-5-ylmethoxy)carbonyl]amino}pentyl)-n2-{[[(2-isopropyl-1,3-thiazol-4-yl)methyl](methyl)amino]carbonyl}-l-valinamide
(2s, 3s, 5s)-5-(n-(n-((n-methyl-n-((2-isopropyl-4-thiazolyl)methyl)amino)carbonyl)valinvl)amino)-2-(n-((5-thiazolyl)methoxycarbonyl)amino)-1,6-diphenyl-3-hydroxyhexane
(2s,3s,5s)-5-(n-(n-((n-methyl-n-((2-isopropyl-4-thiazolyl)methyl)amino)carbonyl)valinyl)amino)-2-(n-((5-thiazolyl)methoxycarbonyl)amino)-1.6-diphenyl-3-hydroxyhexane
(2s,3s,5s)-5-(n-(n-((n-methyl-n-((2-isopropyl-4-thiazolyl) methyl)amino)carbonyl)valinyl)amino)-2-(n-((5-thiazolyl)methoxycarbonyl)amino)-1,6-diphenyl-3-hydroxyhexane
(2s,3s,5s )-5-(n-(n-((n-methyl-n-((2-isopropyl-4-thiazolyl) methyl)amino)carbonyl)valinyl)amino)-2-(n-((5-thiazolyl) methoxycarbonyl)amino)-1,6-diphenyl-3-hydroxyhexane
(2s,3s,5s)-5-(n-(n-((n-methyl-n-((2-isopropyl-4-thiazolyl)methyl)amino)carbonyl)valinyl)amino)-2-(n-((5-thiazolyl)methoxycarbonyl)amino)-1,6-diphenyl-3-hydroxyhexane
(2s,3s,5s)-5-(n-(n-((n-methyi-n-((2-isopropyl-4-thiazolyl)methyl)amino)carbonyl)valinyl)amino)-2-(n-((5-thiazolyl)methoxycarbonyl)amino)-1.6-diphenyl-3-hydroxyhexane
NCDNCNXCDXHOMX-XGKFQTDJSA-N
(2s,3s,5s)-5-(n-(n-((n-methyl-n-((2-isopropyl-4-thiazolyl)methyl)amino)carbonyl)valinyl)amino)-2-(n-((5-thiazolyl)methoxycarbonyl)amino)-1,6-diphenyl-3hydroxyhexane
(2s,3s,5s)-5-(n-(n((n-methyl-n-((2-isopropyl-4-thiazolyl)methyl)amino)carbonyl)valinyl)amino)-2-(n-((5-thiazolyl)methoxycarbonyl)amino)-1,6-diphenyl-3-hydroxyhexane
n-[(2s,4s,5s)-4-hydroxy-1,6-diphenyl-5-{[(1,3-thiazol-5-ylmethoxy)carbonyl]amino}hexan-2-yl]-n(2)-(methyl{[2-(propan-2-yl)-1,3-thiazol-4-yl]methyl}carbamoyl)-l-valinamide
MLS006011764
thiazol-5-ylmethyl ((2s,3s,5s)-3-hydroxy-5-((s)-2-(3-((2-isopropylthiazol-4-yl)methyl)-3-methylureido)-3-methylbutanamido)-1,6-diphenylhexan-2-yl)carbamate
R0116
AB00639991_10
AB00639991_09
3TNE
a 84538
abbot 84538
5-thiazolylmethyl (3s,4s,6s,9s)-4-hydroxy-12-methyl-9-(1-methylethyl)-13-[2-(1-methylethyl)-4-thiazolyl]-8,11-dioxo-3,6-bis(phenylmethyl)-2,7,10,12-tetraazatridecanoate
ritonavir, >=98% (hplc)
J-009178
MRF-0000287
HMS3715L22
NCGC00159462-07
SW197637-2
thiazol-5-ylmethyl (2s,3s,5s)-3-hydroxy-5-((s)-2-(3-((2-isopropylthiazol-4-yl)methyl)-3-methylureido)-3-methylbutanamido)-1,6-diphenylhexan-2-ylcarbamate
2,7,10,12-tetraazatridecanoic acid, 4-hydroxy-12-methyl-9-(1-methylethyl)-13-[2-(1-methylethyl)-4-thiazolyl]-8,11-dioxo-3,6-bis(phenylmethyl)-, 5-thiazolylmethyl ester, (3s,4s,6s,9s)-
Q422618
ritonavir 100 microg/ml in acetonitrile
norvir, norvir softgel
NCGC00159462-20
A904691
thiazol-5-ylmethyl((2s,3s,5s)-3-hydroxy-5-((s)-2-(3-((2-isopropylthiazol-4-yl)methyl)-3-methylureido)-3-methylbutanamido)-1,6-diphenylhexan-2-yl)carbamate
BR164353
ritonavir- bio-x
HB6070
5-thiazolylmethyl (3s,4s,6s,9s)-4-hydroxy-12-methyl-9- (1-methylethyl)-13-[2- (1-methylethyl)-4-thiazolyl]-8,11-dioxo-3,6-bis (phenylmethyl)-2,7,10,12-tetraazatridecanoate
EN300-119505
(1,3-thiazol-5-yl)methyl n-[(2s,3s,5s)-3-hydroxy-5-[(2s)-3-methyl-2-{[methyl({[2-(propan-2-yl)-1,3-thiazol-4-yl]methyl})carbamoyl]amino}butanamido]-1,6-diphenylhexan-2-yl]carbamate
j05ae03
ritonavir (mart.)
n-((2s,4s,5s)-4-hydroxy-1,6-diphenyl-5-(((1,3-thiazol-5-ylmethoxy)carbonyl)amino)hexan-2-yl)-n(2)-(methyl((2-(propan-2-yl)-1,3-thiazol-4-yl)methyl)carbamoyl)-l-valinamide
5-thiazolylmethyl((alphas)-alpha-((1s,3s)-1-hydroxy-3-((2s)-2-(3-((2-isopropyl-4-thiazolyl)methyl)-3-methylureido)-3-methylbutyramido)-4-phenylbutyl)phenethyl)carbamate
ritonavir (usp monograph)
ritonavir (usp impurity)
(5s,8s,10s,11s)-10-hydroxy-2-methyl-5-(1-methylethyl)-1-
Z1521553599

Research Excerpts

Overview

Ritonavir is a BCS class II antiretroviral agent which shows poor aqueous solubility and low oral bioavailability. It has been proposed as a suitable alternative to ketoconazole.

ExcerptReferenceRelevance
"Ritonavir (RTV) is a first generation HIV-1 protease inhibitor with rapidly emerging drug resistance. "( Insights into the mechanism of drug resistance: X-ray structure analysis of multi-drug resistant HIV-1 protease ritonavir complex.
Brunzelle, JS; Dewdney, TG; Kovari, IA; Kovari, LC; Liu, Z; Reiter, SJ; Wang, Y; Yedidi, RS, 2013
)
2.04
"Ritonavir is a BCS class II antiretroviral agent which shows poor aqueous solubility and low oral bioavailability. "( Improved pharmaceutical properties of ritonavir through co-crystallization approach with liquid-assisted grinding method.
Chaudhari, KR; Savjani, JK; Savjani, KT; Shah, H, 2021
)
2.34
"Ritonavir-boosting is a promising strategy to reduce erlotinib treatment costs and provides a rationale for other expensive therapies metabolized by CYP3A4."( Ritonavir-Boosted Exposure of Kinase Inhibitors: an Open Label, Cross-over Pharmacokinetic Proof-of-Concept Trial with Erlotinib.
Baas, P; Beijnen, JH; Boosman, RJ; Burgers, JA; de Gooijer, CJ; Groenland, SL; Huitema, ADR; Steeghs, N; van der Noort, V, 2022
)
2.89
"Ritonavir is a strong inhibitor for CYP3A4/5-mediated DDIs and has been proposed as a suitable alternative to ketoconazole."( A Mechanistic Absorption and Disposition Model of Ritonavir to Predict Exposure and Drug-Drug Interaction Potential of CYP3A4/5 and CYP2D6 Substrates.
Arora, S; Gardner, I; Jamei, M; Kilford, PJ; Pansari, A; Turner, DB, 2022
)
1.7
"Ritonavir is a well-known protease inhibitor with low aqueous solubility belonging to BCS class II category."( Recent advances in nanoformulation development of Ritonavir, a key protease inhibitor used in the treatment of HIV-AIDS.
Bhaskaran, NA; Jitta, SR; Kumar, L; Marques, SM, 2022
)
1.7
"Ritonavir (RIT) is a well-known case where the most stable polymorph II emerged after being marketed, leading to a loss of $250 million."( Ritonavir Revisited: Melt Crystallization Can Easily Find the Late-Appearing Polymorph II and Unexpectedly Discover a New Polymorph III.
Chen, Z; Li, S; Liu, B; Lu, M; Ou, X; Rong, H, 2023
)
3.07
"Ritonavir is a well-known CYP3A4 and CYP2D6 enzyme inhibitor, frequently used to assess the drug-drug interaction (DDI) liability of susceptible drugs. "( Biopharmaceutic
Arora, S; Gardner, I; Jamei, M; Kilford, P; Pansari, A; Turner, DB, 2020
)
2
"Ritonavir (RTV) is a weakly basic drug with a pH-dependent solubility. "( In Vitro Characterization of Ritonavir Drug Products and Correlation to Human in Vivo Performance.
Gao, P; Marroum, P; Shi, Y; Vela, S; Xu, H, 2017
)
2.19
"Ritonavir is a powerful inhibitor of the cytochrome P450 3A4 (CYP3A4) isoenzyme. "( Secondary Adrenal Insufficiency Due to the Co-Administration of Ritonavir and Inhaled Fluticasone Propionate: Case report.
Al-Maqbali, A; Al-Qassabi, S; Elgalib, A; Kamble, B, 2017
)
2.14
"Ritonavir (RTV) is a weakly basic drug with a pH-dependent solubility. "( In vitro characterization of ritonavir formulations and correlation to in vivo performance in dogs.
Gao, P; Krakow, S; Rosenberg, J; Shi, Y; Xu, H, 2018
)
2.21
"Ritonavir (RTV) is a potent mechanism-based and reversible CYP3A inhibitor and moderate inducer that is used as a pharmacokinetic enhancer in several antiviral treatment regimens."( Guiding dose adjustment of amlodipine after co-administration with ritonavir containing regimens using a physiologically-based pharmacokinetic/pharmacodynamic model.
Menon, RM; Mukherjee, D; Shebley, M; Zha, J, 2018
)
1.44
"Ritonavir (RIT) is a widely used antiviral drug that acts as an HIV protease inhibitor with emerging potential in anticancer therapies. "( Nanotherapeutics with suitable properties for advanced anticancer therapy based on HPMA copolymer-bound ritonavir via pH-sensitive spacers.
Chytil, P; Etrych, T; Janoušková, O; Koziolová, E; Machová, D; Venclíková, K, 2018
)
2.14
"Ritonavir (RIT) is a human immune deficiency virus (HIV) protease inhibitor (PI) active against HIV-1 and HIV-2. "( Formononetin and biochanin A protects against ritonavir induced hepatotoxicity via modulation of NfκB/pAkt signaling molecules.
Agarwal, NK; Azmi, L; Chaturvedi, S; Malik, MY; Naseem, Z; Rao, C; Shukla, I, 2018
)
2.18
"Ritonavir (RTV) is a human immunodeficiency virus (HIV) protease inhibitor (PI) with activity against HIV, practically insoluble in water and recommended to co-administer as a booster along with other HIV-PI to enhance their bioavailability. "( Solid self-microemulsifying drug delivery system of ritonavir.
Deshmukh, A; Kulkarni, S, 2014
)
2.1
"Ritonavir is a HIV protease inhibitor. "( Cardioprotective effect of ritonavir, an antiviral drug, in isoproterenol induced myocardial necrosis: a new therapeutic implication.
Banerjee, SK; Bulani, Y; Gupta, P; Kanwal, A; Khatua, TN; Kuncha, M; Putcha, UK; Sojitra, B, 2013
)
2.13
"Ritonavir is a human immunodeficiency virus (HIV) protease inhibitor and an inhibitor of cytochrome P450 3A4, the major human hepatic drug-metabolizing enzyme. "( Characterization of ritonavir-mediated inactivation of cytochrome P450 3A4.
Hengel, SM; Kunze, KL; Rock, BM; Rock, DA; Wienkers, LC, 2014
)
2.17
"Ritonavir is a protease inhibitor utilized primarily as a pharmaco-enhancer with concomitantly administered antiviral drugs including other protease inhibitors. "( Augmented Inhibition of CYP3A4 in Human Primary Hepatocytes by Ritonavir Solid Drug Nanoparticles.
Giardiello, M; Martin, P; McDonald, TO; Owen, A; Rannard, SP; Siccardi, M; Smith, D, 2015
)
2.1
"Ritonavir is an antiretroviral drug to treat HIV AIDS and inhibits cytochrome P450 3A4. "( Altered pharmacokinetics and pharmacodynamics of repaglinide by ritonavir in rats with healthy, diabetic and impaired hepatic function.
Goud, T; Maddi, S; Nayakanti, D; Thatipamula, RP, 2016
)
2.12
"Ritonavir is a well-known inhibitor of the metabolism of numerous medications that are substrates of the CYP3A and CYP2D6 pathways. "( Induction effects of ritonavir: implications for drug interactions.
Foisy, MM; Hughes, CA; Yakiwchuk, EM, 2008
)
2.11
"Ritonavir is a powerful inhibitor of cytochrome P450 3A (CYP3A) that metabolizes many antiretrovirals. "( Ritonavir greatly impairs CYP3A activity in HIV infection with chronic viral hepatitis.
Greenblatt, DJ; Kaufman, RC; Knox, TA; Oleson, L; von Moltke, LL; Wanke, CA, 2008
)
3.23
"Ritonavir is a potent CYP3A5 isoenzyme and P-gp inhibitor."( Paralytic ileus possibly associated with interaction between ritonavir/lopinavir and vincristine.
Bergerat, JP; Herbrecht, R; Levêque, D; Pavillet, J; Santucci, R, 2009
)
1.32
"Ritonavir is an antiretroviral drug characterized by low solubility and high permeability which corresponds to BCS class II drug. "( Solid dispersion as an approach for bioavailability enhancement of poorly water-soluble drug ritonavir.
Ahuja, A; Ali, J; Ali, M; Baboota, S; Kumar, A; Sinha, S, 2010
)
2.02
"Ritonavir is a protease inhibitor associated with metabolic abnormalities and cardiovascular disease. "( Investigation of low-dose ritonavir on human peripheral blood mononuclear cells using gene expression whole genome microarrays.
Back, D; Boffito, M; Collot-Teixeira, S; De Lorenzo, F; Fletcher, C; Gazzard, B; McGregor, JL; Pozniak, A; Waters, L; Yilmaz, S, 2010
)
2.1
"Ritonavir (RTV) is a commonly used antiretroviral associated with bone loss. "( Effects of HIV infection and antiretroviral therapy with ritonavir on induction of osteoclast-like cells in postmenopausal women.
Cremers, S; Ferris, DC; Laurence, J; McMahon, DJ; Modarresi, R; Santiago, F; Shane, E; Yin, MT; Zhang, CA, 2011
)
2.06
"Ritonavir is a synthetic peptidomimetic human immunodeficiency virus (HIV) protease inhibitor employed in the treatment of AIDS since 1996. "( Micellar electrokinetic chromatography method development for determination of impurities in ritonavir.
Carvalho, AZ; Duppen, JV; El-Attug, MN; Hoogmartens, J; Hove, EV; Schepdael, AV; Zayed, SE, 2010
)
2.02
"Ritonavir is a HIV protease inhibitor routinely prescribed to HIV patients that also potently inactivates cytochrome P4503A4 (CYP3A4), the major human drug-metabolizing enzyme. "( Structure and mechanism of the complex between cytochrome P4503A4 and ritonavir.
Poulos, TL; Sevrioukova, IF, 2010
)
2.04
"Ritonavir is a potential therapeutic agent in lung cancer, but its targets in lung adenocarcinoma are unknown, as are candidate biomarkers for its activity."( The human immunodeficiency virus protease inhibitor ritonavir inhibits lung cancer cells, in part, by inhibition of survivin.
Fukuda, S; Guo, Z; Kathuria, H; Milani, M; Mitra, R; Pelus, LM; Potter, DA; Rizzardi, A; Rodriguez, M; Schmechel, S; Skalnik, DG; Srirangam, A, 2011
)
2.06
"Ritonavir (RTV), which is a human immunodeficiency virus (HIV) protease inhibitor, can induce atherosclerosis in a PKC-dependent manner."( Ritonavir stimulates foam cell formation by activating PKC.
Ding, H; Liu, H; Liu, Y; Mu, Y; Sun, G; Xiang, J; Xu, J; Yang, F, 2011
)
2.53
"Ritonavir (RTV) is a known inhibitor of both P-gp and CYP3A and is co-administered with LPV in anti-HIV therapy."( Effect of grapefruit juice and ritonavir on pharmacokinetics of lopinavir in Wistar rats.
Aditya, N; Ravi, PR; Srivani, S; Thakur, R; Vats, R, 2012
)
1.39
"Ritonavir is a HIV protease inhibitor that also potently inactivates cytochrome P450 3A4 (CYP3A4), a major human drug-metabolizing enzyme. "( Interaction of human cytochrome P4503A4 with ritonavir analogs.
Poulos, TL; Sevrioukova, IF, 2012
)
2.08
"Ritonavir is a protease inhibitor (PI) frequently prescribed with highly active antiretroviral therapy. "( Ritonavir and epidural triamcinolone as a cause of iatrogenic Cushing's syndrome.
Albert, NE; Dougherty, R; Kazi, S; Santoro, J, 2012
)
3.26
"Ritonavir is a potent CYP3A inhibitor and was shown in vitro as a CYP2B6 inhibitor."( Pharmacokinetic interaction between prasugrel and ritonavir in healthy volunteers.
Ancrenaz, V; Daali, Y; Dayer, P; Déglon, J; Desmeules, J; Samer, C; Staub, C, 2013
)
1.36
"Ritonavir is a protease inhibitor whose potent cytochrome P450 3A4 inhibition is exploited for pharmacologic boosting in human immunodeficiency virus (HIV) infection."( Iatrogenic hypercortisolism complicating triamcinolone acetonide injections in patients with HIV on ritonavir-boosted protease inhibitors.
Beach, J; Fessler, D; Keel, J; Stead, W,
)
1.07
"Ritonavir is an extremely strong inhibitor of P450 cytochrome 3A, which is the main metabolizing enzyme of tacrolimus. "( Pretransplantation pharmacokinetic curves of tacrolimus in HIV-infected patients on ritonavir-containing cART: a pilot study.
Crommelin, HA; Mudrikova, T; van den Broek, MP; van Maarseveen, EM; van Zuilen, AD, 2013
)
2.06
"Ritonavir is an HIV protease inhibitor used in the therapy of HIV infection. "( Antitumor effect of the human immunodeficiency virus protease inhibitor ritonavir: induction of tumor-cell apoptosis associated with perturbation of proteasomal proteolysis.
Constantiniu, O; Firat-Geier, E; Freudenberg, M; Gaedicke, S; Galanos, C; Lucchiari-Hartz, M; Niedermann, G, 2002
)
1.99
"Ritonavir is a protease inhibitor, used in combination therapy (often as a pharmacoenhancer) to treat HIV."( The distribution of the HIV protease inhibitor, ritonavir, to the brain, cerebrospinal fluid, and choroid plexuses of the guinea pig.
Anthonypillai, C; Gibbs, JE; Sanderson, RN; Thomas, SA, 2004
)
1.3
"Ritonavir is a large, lipophilic molecule that is practically insoluble in aqueous media and exhibits an exceedingly slow intrinsic dissolution rate. "( Ritonavir-PEG 8000 amorphous solid dispersions: in vitro and in vivo evaluations.
Everitt, EA; Fort, JJ; Krill, SL; Law, D; Marsh, KC; Qiu, Y; Schmitt, EA; Wang, W, 2004
)
3.21
"Ritonavir is a potent in vitro inhibitor of several cytochrome P450 isozymes and ABC transporters including the efflux pump P-glycoprotein (P-gp). "( Substantial pharmacokinetic interaction between digoxin and ritonavir in healthy volunteers.
Burhenne, J; Ding, R; Haefeli, WE; Mikus, G; Riedel, KD; Tayrouz, Y; Weiss, J, 2004
)
2.01
"Ritonavir is an inhibitor of HIV-1 protease with potent in vitro anti-HIV properties and good oral bioavailability."( A short-term study of the safety, pharmacokinetics, and efficacy of ritonavir, an inhibitor of HIV-1 protease. European-Australian Collaborative Ritonavir Study Group.
Bouza, E; Carr, A; Danner, SA; Gonzales, J; Gudiol, F; Lehman, LM; Leonard, JM; Pintado, V; Raventos, A; Rubio, R, 1995
)
1.25
"Ritonavir is a potent inhibitor in vitro of human immunodeficiency virus type 1 (HIV-1) protease, which is needed for virions to mature and become infective. "( A preliminary study of ritonavir, an inhibitor of HIV-1 protease, to treat HIV-1 infection.
Henry, D; Hsu, A; Hurley, AM; La Marca, A; Leonard, JM; Markowitz, M; Powderly, WG; Saag, M; Sattler, F; Valdes, JM, 1995
)
2.04
"Ritonavir was found to be a potent inhibitor of CYP3A-mediated biotransformations (nifedipine oxidation, IC50) = 0.07 microM; 17alpha-ethynylestradiol 2-hydroxylation, IC50 = 2 microM; terfenadine hydroxylation, IC50 = 0.14 microM)."( Cytochrome P450-mediated metabolism of the HIV-1 protease inhibitor ritonavir (ABT-538) in human liver microsomes.
Buko, AM; Denissen, JF; Kumar, GN; Rodrigues, AD, 1996
)
1.25
"Ritonavir is a protease inhibitor with an HIV-1 resistance profile similar to that of indinavir, but different from that of saquinavir. "( Ritonavir.
Faulds, D; Lea, AP, 1996
)
3.18
"Ritonavir (RIT) is a potent inhibitor of CYP3A4 and inhibits saquinavir (SQV) metabolism in healthy volunteers."( Saquinavir pharmacokinetics alone and in combination with ritonavir in HIV-infected patients.
Back, DJ; Barry, MG; Breckenridge, AM; Gibbons, SE; Heavey, J; Merry, C; Mulcahy, F; Ryan, M; Tjia, JF, 1997
)
1.26
"Ritonavir is a potent, orally bioavailable inhibitor of HIV-1 protease. "( Randomised placebo-controlled trial of ritonavir in advanced HIV-1 disease. The Advanced HIV Disease Ritonavir Study Group.
Cameron, DW; Cohen, C; Danner, S; Heath-Chiozzi, M; Henry, D; Kravcik, S; Leonard, J; Maurath, C; Potthoff, A; Rode, R; Sun, E, 1998
)
2.01
"Ritonavir is a potent, orally bioavailable HIV protease inhibitor with demonstrated impact on surrogate endpoints, AIDS-defining disease, and mortality."( A randomized trial of the effect of ritonavir in maintaining quality of life in advanced HIV disease. Advanced HIV Disease Ritonavir Study Group.
Cohen, C; Jiang, P; Nabulsi, A; Revicki, DA; Sarocco, PW, 1998
)
1.3
"Ritonavir is a potent inhibitor of cytochrome P4503A4 that strongly increases saquinavir bioavailability. "( Safety and efficacy of ritonavir and saquinavir in combination with zidovudine and lamivudine.
Arvieux, C; Bazin, C; Bellissant, E; Cartier, F; Chauvin, JP; Delfraissy, JF; Dohin, E; Michelet, C; Raffi, F; Renard, I; Ruffault, A; Sébille, V, 1999
)
2.06
"Ritonavir is a reversible effector of proteasome activity that protected the subunits MB-1 (X) and/or LMP7 from covalent active site modification with the vinyl sulfone inhibitor(125)I-NLVS, suggesting that they are the prime targets for competitive inhibition by Ritonavir."( How an inhibitor of the HIV-I protease modulates proteasome activity.
Bogyo, M; de Giuli, R; Emch, S; Groettrup, M; Groll, M; Holzhütter, HG; Kairies, N; Schmidtke, G, 1999
)
1.02
"Ritonavir is a potent inhibitor of hepatic cytochrome P450, mainly the CYP3A4 isoform."( Protease inhibitor-induced carbamazepine toxicity.
Berbel Garcia, A; Latorre Ibarra, A; Martinez Salio, A; Perez Martinez, D; Porta Etessam, J; Siaz Diaz, R; Toledo Heras, M,
)
0.85
"Ritonavir (RTV) is a powerful inhibitor of P450 3A4 cytochorme. "( [Indinavir-ritonavir combination: pharmacologic results and tolerance in patients infected by HIV].
Bani-Sadr, F; Bernard, L; de Truchis, P; Melchior, JC; Perré, P; Perronne, C; Peytavin, G, 2001
)
2.14
"Ritonavir is an HIV-1 protease inhibitor that is often used to improve the systemic availability of concurrently administered protease inhibitors by impairing their metabolism through cytochrome P450 (CYP) 3A4. "( Rapid and sensitive high-performance liquid chromatographic method for the determination of ritonavir in human plasma.
Gubbins, PO; Lawhorn, WD; Penzak, SR, 2001
)
1.97

Effects

Ritonavir has been shown to inhibit the chymotrypsin-like activity of isolated 20S proteasomes. It inhibits betaPDGFR activation and PDGF-dependent proliferation and migration of VSMCs.

ExcerptReferenceRelevance
"Ritonavir has a double-boosting function for both lopinavir and saquinavir, and in terms of pharmacokinetics, the drug doses selected seemed appropriate for combining these agents in a dual protease inhibitor-based antiretroviral regimen for patients with several prior virologic failures."( Steady-state pharmacokinetics of a double-boosting regimen of saquinavir soft gel plus lopinavir plus minidose ritonavir in human immunodeficiency virus-infected adults.
Azuaje, C; Clotet, B; Crespo, M; Diaz, M; Falcó, V; Lopez, RM; Ocaña, I; Pahissa, A; Pou, L; Ribera, E; Ruiz, I; Ruiz, L, 2004
)
1.26
"Ritonavir and metformin have been administered in humans for the treatment of diabetes in patients with HIV, demonstrating the tolerance to this combination in humans."( Investigating and targeting chronic lymphocytic leukemia metabolism with the human immunodeficiency virus protease inhibitor ritonavir and metformin.
Adekola, KU; Dalva Aydemir, S; Ma, S; Rosen, ST; Shanmugam, M; Zhou, Z, 2015
)
1.34
"Ritonavir has been evaluated at boosting doses of 50–800 mg daily with seven protease inhibitors: amprenavir, atazanavir, darunavir, indinavir, lopinavir,saquinavir and tipranavir. "( How much ritonavir is needed to boost protease inhibitors? Systematic review of 17 dose-ranging pharmacokinetic trials.
Boffito, M; Hill, A; Sawyer, W; van der Lugt, J, 2009
)
2.21
"Ritonavir has also been shown to inhibit the chymotrypsin-like activity of isolated 20S proteasomes."( Antitumor effect of the human immunodeficiency virus protease inhibitor ritonavir: induction of tumor-cell apoptosis associated with perturbation of proteasomal proteolysis.
Constantiniu, O; Firat-Geier, E; Freudenberg, M; Gaedicke, S; Galanos, C; Lucchiari-Hartz, M; Niedermann, G, 2002
)
1.27
"Ritonavir has direct effects on VSMCs at clinically relevant concentrations in vitro, as it inhibits betaPDGFR activation and PDGF-dependent proliferation and migration of VSMCs. "( Ritonavir exhibits anti-atherogenic properties on vascular smooth muscle cells.
Bäumer, AT; Caglayan, E; Fätkenheuer, G; Kappert, K; Rosenkranz, S; Südkamp, M, 2004
)
3.21
"Ritonavir has a double-boosting function for both lopinavir and saquinavir, and in terms of pharmacokinetics, the drug doses selected seemed appropriate for combining these agents in a dual protease inhibitor-based antiretroviral regimen for patients with several prior virologic failures."( Steady-state pharmacokinetics of a double-boosting regimen of saquinavir soft gel plus lopinavir plus minidose ritonavir in human immunodeficiency virus-infected adults.
Azuaje, C; Clotet, B; Crespo, M; Diaz, M; Falcó, V; Lopez, RM; Ocaña, I; Pahissa, A; Pou, L; Ribera, E; Ruiz, I; Ruiz, L, 2004
)
1.26
"Ritonavir has been repeatedly shown to be the most common protease inhibitor to induce these metabolic abnormalities."( Tuberous and tendinous xanthomata secondary to ritonavir-associated hyperlipidemia.
Brown, CA; Lesher, JL; Peterson, CM, 2005
)
1.31
"Ritonavir has good oral bioavailability, and may increase the bioavailability of other protease inhibitors including saquinavir, nelfinavir, indinavir and VX-478."( Ritonavir.
Faulds, D; Lea, AP, 1996
)
2.46
"Ritonavir has been shown to be a potent inhibitor of CYP3A4, an enzyme responsible for warfarin metabolism."( Potential interaction involving warfarin and ritonavir.
Cousins, ES; Knoell, KR; Young, TM, 1998
)
1.28
"Ritonavir has the severest side effects, including nausea, diarrhea, and initially, tingling feeling of the mouth, arms, or legs."( A patient's guide to protease inhibitors.
Elperin, A; Sax, P, 1996
)
1.02

Actions

Ritonavir was found to inhibit in a concentration dependent manner the intrinsic clearance of docetaxel, which was described by an inhibition constant of 0.028 microg ml(-1) (CV 36%). RitonavIR is known to inhibit several drug transporters.

ExcerptReferenceRelevance
"Ritonavir is known to inhibit several drug transporters; therefore, we have examined these compounds together, in relevant concentrations and ratios."( Interactions of the Anti-SARS-CoV-2 Agents Molnupiravir and Nirmatrelvir/Paxlovid with Human Drug Transporters.
Bakos, É; Gáborik, Z; Özvegy-Laczka, C; Sarkadi, B; Telbisz, Á; Temesszentandrási-Ambrus, C, 2023
)
1.63
"Ritonavir was found to inhibit in a concentration dependent manner the intrinsic clearance of docetaxel, which was described by an inhibition constant of 0.028 microg ml(-1) (CV 36%)."( Population pharmacokinetics of intravenously and orally administered docetaxel with or without co-administration of ritonavir in patients with advanced cancer.
Beijnen, JH; Huitema, AD; Koolen, SL; Oostendorp, RL; Schellens, JH, 2010
)
1.29
"Ritonavir can inhibit the metabolism of fentanyl significantly, so caution should be exercised if fentanyl is given to patients receiving ritonavir medication."( Ritonavir's role in reducing fentanyl clearance and prolonging its half-life.
Neuvonen, PJ; Olkkola, KT; Palkama, VJ, 1999
)
3.19

Treatment

Ritonavir treatment initially triggered ER stress during the early hours of treatment. Maraviroc downregulated the local expression of vascular cell adhesion molecule-1. The drug significantly reduced calpain activity in the hippocampus, protected hippocampal neurons from death, preserved cognitive performance.

ExcerptReferenceRelevance
"Ritonavir treatment initially triggered ER stress during the early hours of treatment. "( Endoplasmic reticulum stress leads to mitochondria-mediated apoptosis in cells treated with anti-HIV protease inhibitor ritonavir.
Chaubey, B; Ganta, KK, 2019
)
2.16
"In ritonavir-treated mice, maraviroc reduced plaque areas and macrophage infiltration; downregulated the local expression of vascular cell adhesion molecule-1, intercellular adhesion molecule-1, monocyte chemoattractant protein-1, and interleukin-17A; and reduced tumor necrosis factor-α and RANTES (regulated on activation, normal T cell expressed, and secreted)."( Efficacy of the CCR5 antagonist maraviroc in reducing early, ritonavir-induced atherogenesis and advanced plaque progression in mice.
Baldelli, F; Cipriani, S; D'Amore, C; Fiorucci, S; Francisci, D; Mencarelli, A; Renga, B; Schiaroli, E, 2013
)
1.15
"Ritonavir-treated multiple myeloma cells exhibited increased reliance on glutamine metabolism. "( Targeting the metabolic plasticity of multiple myeloma with FDA-approved ritonavir and metformin.
Adekola, KU; Bajpai, R; Dalva-Aydemir, S; Kandela, I; Koblinski, JE; Martinez, M; Raje, NS; Rosen, ST; Shanmugam, M; Singhal, S; Wei, C, 2015
)
2.09
"Ritonavir treatment alone did not significantly affect the median time of survival (14 vs."( Ritonavir inhibits intratumoral docetaxel metabolism and enhances docetaxel antitumor activity in an immunocompetent mouse breast cancer model.
Beijnen, JH; Hendrikx, JJ; Lagas, JS; Rosing, H; Rottenberg, S; Schellens, JH; Schinkel, AH; Song, JY, 2016
)
2.6
"Ritonavir treatment significantly reduced calpain activity in the hippocampus, protected hippocampal neurons from death, preserved cognitive performance, and suppressed seizure escalation, even when therapy was initiated 36 hours after disease onset."( Neuroprotection mediated by inhibition of calpain during acute viral encephalitis.
Buenz, EJ; Howe, CL; LaFrance-Corey, RG; McGovern, RM; Mirchia, K; Reid, JM; Sauer, BM, 2016
)
1.16
"Ritonavir-treated cells had increased FA efflux, uptake, and incorporation into TAG (all P < 0.01)."( Long-term ritonavir exposure increases fatty acid and glycerol recycling in 3T3-L1 adipocytes as compensatory mechanisms for increased triacylglycerol hydrolysis.
Adler-Wailes, DC; Guiney, EL; Wolins, NE; Yanovski, JA, 2010
)
1.48
"Ritonavir treatment was then continued for an additional 13 days."( Renal insufficiency has no effect on the pharmacokinetics of vicriviroc in a ritonavir-containing regimen.
Assaf, M; Kasserra, C; Keung, A; Marbury, T; O'Mara, E; Sansone-Parsons, A; Tetteh, E, 2010
)
1.31
"In ritonavir-treated cells, protein expression of the lipid droplet-protective protein, perilipin, was significantly decreased, whereas there was no change in hormone-sensitive lipase."( Effects of the human immunodeficiency virus-protease inhibitor, ritonavir, on basal and catecholamine-stimulated lipolysis.
Adler-Wailes, DC; Ahmad, F; Feng, N; Liu, H; Londos, C; Manganiello, V; Yanovski, JA, 2005
)
1.08
"Ritonavir treatment significantly decreased baseline insulin release and glucose-stimulated insulin release in a dose-dependent manner (r=-0.861, -0.839, both P<0.01). "( [Effects of various HIV protease inhibitors on function of rat insulinoma cells].
Li, WP; Schutt, M; Xiang, Z; Zhou, JQ, 2006
)
1.78
"Ritonavir treatment resulted in increased kaolin intake or severe pica, the intensity of which was reduced significantly with S."( Scutellaria baicalensis and a constituent flavonoid, baicalein, attenuate ritonavir-induced gastrointestinal side-effects.
Aung, H; Foo, A; Mehendale, S; Tong, R; Wang, CZ; Xie, JT; Yuan, CS, 2007
)
1.29
"Ritonavir treatment alone initially suppressed plasma viremia, but the viremia recurred with the appearance of ritonavir-resistant HIV isolates."( thy/liv-SCID-hu mice: a system for investigating the in vivo effects of multidrug therapy on plasma viremia and human immunodeficiency virus replication in lymphoid tissues.
Goldstein, H; Katopodis, NF; Kim, A; Kollmann, TR; Pettoello-Mantovani, M; Raker, C; Wiltshire, H; Yurasov, S, 1998
)
1.02
"Ritonavir-treated patients reported fewer fever symptoms and neurologic symptoms than the placebo group after 6 months of treatment (P < 0.005)."( A randomized trial of the effect of ritonavir in maintaining quality of life in advanced HIV disease. Advanced HIV Disease Ritonavir Study Group.
Cohen, C; Jiang, P; Nabulsi, A; Revicki, DA; Sarocco, PW, 1998
)
1.3
"In ritonavir-treated samples derived from HIV-infected individuals, the number of cells expressing ICE also decreased."( Protease inhibitors stimulate hematopoiesis and decrease apoptosis and ICE expression in CD34(+) cells.
Chaudhuri, A; Kim, S; Kumar, P; Maciejewski, J; Sloand, EM; Young, N, 2000
)
0.82
"Ritonavir treatment increased plasma triglyceride and cholesterol levels through increased fatty acid and cholesterol biosynthesis in adipose and liver."( HIV protease inhibitor induces fatty acid and sterol biosynthesis in liver and adipose tissues due to the accumulation of activated sterol regulatory element-binding proteins in the nucleus.
Fichtenbaum, CJ; Hui, DY; Kuhel, DG; Riddle, TM; Woollett, LA, 2001
)
1.03
"Treatment of ritonavir marginally declined simulated S-warfarin concentrations, but substantially elevated that of R-warfarin, resulting in a decrease in the international normalized ratio (INR)."( Physiologically-Based Pharmacokinetic Modeling-Guided Dose Management of Oral Anticoagulants when Initiating Nirmatrelvir/Ritonavir (Paxlovid) for COVID-19 Treatment.
Chan, ECY; Wang, Z, 2022
)
1.28
"Treatment with ritonavir-boosted nirmatrelvir or molnupiravir."( Nirmatrelvir or Molnupiravir Use and Severe Outcomes From Omicron Infections.
Abi Fadel, F; Bartley, P; Duggal, A; Huang, S; Lin, DY; Milinovich, AT; Sacha, GL; Wang, X, 2023
)
1.26
"Treatment with ritonavir-boosted protease inhibitors or efavirenz and nucleoside analogues leads to rises in lipids, which might contribute to cardiovascular risk."( Effects of first-line use of nucleoside analogues, efavirenz, and ritonavir-boosted protease inhibitors on lipid levels.
Gazzard, B; Hill, A; Sawyer, W,
)
0.72
"Treatment with ritonavir-boosted fosamprenavir can produce a durable response."( Fosamprenavir calcium plus ritonavir for HIV infection.
Arduino, RC; Torres, HA, 2007
)
0.98
"Treatment with ritonavir produced alterations in the immune system that included changes in T cell subset distribution and increases in CD4 and CD8 lymphocyte numbers and of lymphocyte function."( Alterations in the immune response of human immunodeficiency virus (HIV)-infected subjects treated with an HIV-specific protease inhibitor, ritonavir.
Carr, A; Cooper, DA; Kelleher, AD; Zaunders, J, 1996
)
0.84
"Treatment with ritonavir in the absence of HIV infection or changes in body composition results in hypertriglyceridemia that is apparently not mediated by impaired LpL activity or the defective removal of remnant lipoproteins, but could be caused by enhanced formation of VLDL. "( Effect of ritonavir on lipids and post-heparin lipase activities in normal subjects.
Achari, R; Brunzell, JD; Knopp, RH; Leonard, JM; Locke, C; Pizzuti, DJ; Purnell, JQ; Zambon, A, 2000
)
1.06

Toxicity

Preliminary data suggest that the protease inhibitor combination ritonavir/indinavir plus double nucleoside therapy appears to be effective and safe in short-term treatment. Treatment-related gastrointestinal adverse events were greater in patients taking lopinavir/ritonavir.

ExcerptReferenceRelevance
" Adverse events included nausea, circumoral paresthesia, elevated hepatic aminotransferase levels, and elevated triglyceride levels."( A short-term study of the safety, pharmacokinetics, and efficacy of ritonavir, an inhibitor of HIV-1 protease. European-Australian Collaborative Ritonavir Study Group.
Bouza, E; Carr, A; Danner, SA; Gonzales, J; Gudiol, F; Lehman, LM; Leonard, JM; Pintado, V; Raventos, A; Rubio, R, 1995
)
0.53
"7%) patients because of adverse events and three (3."( Efficacy and safety of twice daily first-line ritonavir/indinavir plus double nucleoside combination therapy in HIV-infected individuals. German Ritonavir/Indinavir Study Group.
Bergmann, F; Carls, H; Fätkenheuer, G; Fenske, S; Knechten, H; Nadler, M; Oette, M; Rieke, A; Rockstroh, JK; Thiesen, A; Wiesel, W, 2000
)
0.57
"Our preliminary data suggest that the protease inhibitor combination ritonavir/indinavir plus double nucleoside therapy appears to be effective and safe in short-term treatment (up to 24 weeks)."( Efficacy and safety of twice daily first-line ritonavir/indinavir plus double nucleoside combination therapy in HIV-infected individuals. German Ritonavir/Indinavir Study Group.
Bergmann, F; Carls, H; Fätkenheuer, G; Fenske, S; Knechten, H; Nadler, M; Oette, M; Rieke, A; Rockstroh, JK; Thiesen, A; Wiesel, W, 2000
)
0.8
" In this study, it appeared safe to continue ARVT during LEE; however, more data from larger studies are required to confirm this finding."( Risk factors for hepatotoxicity in HIV-1-infected patients receiving ritonavir and saquinavir with or without stavudine. Prometheus Study Group.
Danner, SA; Dreezen, C; Gisolf, EH; Weel, JL; Weverling, GJ, 2000
)
0.54
" The side-effects of this new therapeutic family are quite well known but we wanted to evaluate the attitude of the clinician: can these adverse effects be corrected by symptomatic treatment, do they regress spontaneously or do they lead to an alternative PI therapy? We therefore carried out a retrospective survey in the Infectious Diseases Department of Poitiers Hospital consisting in research on files of patients (n = 70) treated in this department (hospitalization and consultation) for any clinical or biological abnormality attributable to the PI."( [Attitude of the clinican toward adverse effects of protease inhibitors].
Breux, JP; Champagne, X; Nicol, B; Pasdeloup, T; Pérault, MC; Remblier, C; Vandel, B,
)
0.13
"Demographics, dosage regimens, genotype data, viral RNA and CD4+ lymphocyte counts, adverse drug events (ADEs), laboratory tests, and compliance were evaluated over 3 years."( The safety and antiviral effect of protease inhibitors in children.
Koranyi, KI; Nahata, MC; Temple, ME, 2001
)
0.31
" Safety was monitored as clinical adverse events (AEs) and laboratory abnormalities."( Pharmacokinetics and safety of amprenavir and ritonavir following multiple-dose, co-administration to healthy volunteers.
Lloyd, PP; Lou, Y; Piliero, PJ; Preston, SL; Sadler, BM; Stein, DS, 2001
)
0.57
" Three patients discontinued therapy for drug-related adverse events."( Safety and antiviral activity at 48 weeks of lopinavir/ritonavir plus nevirapine and 2 nucleoside reverse-transcriptase inhibitors in human immunodeficiency virus type 1-infected protease inhibitor-experienced patients.
Benson, CA; Brun, SC; Deeks, SG; Eron, JJ; Feinberg, J; Gulick, RM; Hicks, C; Hsu, A; Japour, AJ; Kempf, D; Kessler, HA; King, M; Murphy, RL; Real, K; Riddler, S; Sax, PE; Stryker, R; Sun, E; Thompson, M; Wheeler, D, 2002
)
0.56
" Apart from diarrhoea and nausea, serum lipid abnormalities were identified as the most prominent adverse reaction."( Long-term efficacy and safety of ritonavir/indinavir at 400/400 mg twice a day in combination with two nucleoside reverse transcriptase inhibitors as first line antiretroviral therapy.
Bergmann, F; Carls, H; Fätkenheuer, G; Fenske, S; Knechten, H; Lichterfeld, M; Nadler, M; Nischalke, HD; Oette, M; Rieke, A; Rockstroh, JK; Theisen, A; Wasmuth, JC; Wiesel, W, 2002
)
0.6
"Our results demonstrate that quadruple therapy with RTV/IDV and two NRTIs induces potent, durable and safe HIV suppression and might be particularly beneficial as a first line therapy for patients with a high baseline viral load."( Long-term efficacy and safety of ritonavir/indinavir at 400/400 mg twice a day in combination with two nucleoside reverse transcriptase inhibitors as first line antiretroviral therapy.
Bergmann, F; Carls, H; Fätkenheuer, G; Fenske, S; Knechten, H; Lichterfeld, M; Nadler, M; Nischalke, HD; Oette, M; Rieke, A; Rockstroh, JK; Theisen, A; Wasmuth, JC; Wiesel, W, 2002
)
0.6
" Thus short-term low-dose administration of ritonavir impairs oral clearance of trazodone and increases the occurrence of adverse reactions."( Short-term exposure to low-dose ritonavir impairs clearance and enhances adverse effects of trazodone.
Byron, S; Chen, G; Culm, KE; Daily, JP; Fogelman, SM; Graf, JA; Granda, BW; Greenblatt, DJ; Harmatz, JS; Mertzanis, P; Shader, RI; von Moltke, LL, 2003
)
0.86
"The A/S/D regimen had a low efficacy and a high frequency of adverse events and cannot be recommended."( Low efficacy and high frequency of adverse events in a randomized trial of the triple nucleoside regimen abacavir, stavudine and didanosine.
Gerstoft, J; Katzenstein, TL; Kirk, O; Lundgren, JD; Mathiesen, L; Nielsen, H; Obel, N; Pedersen, C, 2003
)
0.32
" This is a potent, safe combination for the treatment of HIV-1."( The safety and efficacy of indinavir and ritonavir (400/400 mg BID) in HIV-1-infected individuals from an inner-city minority population: a pilot study.
Asthana, D; Campo, R; Jayaweera, DT; Kolber, MA; Robinson, M; Rode, R; Rodriguez, A; Scerpella, E; Tanner, T, 2003
)
0.58
" Ten patients discontinued the study treatment before W48: adverse events (eight), patient's will (one) and simplification of therapy (one)."( Efficacy and safety of ritonavir/indinavir 100/400 mg twice daily in combination with two nucleoside analogues in antiretroviral treatment-naive HIV-infected individuals.
Agher, R; Ait-Mohand, H; Bricaire, F; Calvez, V; Costagliola, D; Duvivier, C; Ghosn, J; Katlama, C; Marcelin, AG; Myrto, A; Peytavin, G; Schneider, L, 2003
)
0.63
"IDV/RTV 400/100 mg twice daily is an effective and safe first-line antiretroviral therapy."( Efficacy and safety of ritonavir/indinavir 100/400 mg twice daily in combination with two nucleoside analogues in antiretroviral treatment-naive HIV-infected individuals.
Agher, R; Ait-Mohand, H; Bricaire, F; Calvez, V; Costagliola, D; Duvivier, C; Ghosn, J; Katlama, C; Marcelin, AG; Myrto, A; Peytavin, G; Schneider, L, 2003
)
0.63
" In our clinical setting IDV/r showed to be less effective and more toxic than LPV/RTV as first-line HAART."( Lopinavir/ritonavir vs. indinavir/ritonavir in antiretroviral naive HIV-infected patients: immunovirological outcome and side effects.
Adorni, F; Bini, T; Bongiovanni, M; Chiesa, E; Cicconi, P; Monforte d'Arminio, A, 2004
)
0.73
" Discontinuation due to adverse events is infrequent."( Safety, efficacy and development of resistance under the new protease inhibitor lopinavir/ritonavir: 48-week results.
Kaiser, R; Mauss, S; Rockstroh, JK; Schmutz, G; Vogel, M; Voigt, E; Wasmuth, JC, 2004
)
0.54
" Adverse events were consistent with prior data for each of the separate agents."( Pharmacokinetics and safety of GW433908 and ritonavir, with and without efavirenz, in healthy volunteers.
Ballow, C; Hendrix, CW; Lou, Y; Piliero, PJ; Preston, SL; Stein, DS; Wire, MB, 2004
)
0.58
" Twenty-eight patients discontinued therapy prior to week 204 because of adverse events (n = 10), lost to follow-up (n = 9), or other reasons (n = 9)."( Long-term safety and durable antiretroviral activity of lopinavir/ritonavir in treatment-naive patients: 4 year follow-up study.
Benson, C; Brun, SC; Eron, JJ; Gulick, RM; Hicks, C; Kessler, HA; King, KR; King, MS; Murphy, RL; White, AC, 2004
)
0.56
" There were no significant differences across treatment arms with regard to drug-related adverse events."( A 14-day dose-response study of the efficacy, safety, and pharmacokinetics of the nonpeptidic protease inhibitor tipranavir in treatment-naive HIV-1-infected patients.
Borin, M; Curry, K; Freimuth, W; MacGregor, T; Mayers, DL; McCallister, S; Valdez, H; Wang, Y, 2004
)
0.32
"ATV treatment of HIV-infected patients with or without a RTV booster was safe and effective in clinical routine."( Atazanavir for treatment of HIV infection in clinical routine: efficacy, pharmacokinetics and safety.
Feldt, T; Göbels, K; Häussinger, D; Kroidl, A; Kuschak, D; Leidel, R; Oette, M; Sagir, A, 2005
)
0.33
" Parameters studied were HIV RNA, CD4+ cell counts, metabolic parameters and drug-related adverse events."( Predictive factors of lopinavir/ritonavir discontinuation for drug-related toxicity: results from a cohort of 416 multi-experienced HIV-infected individuals.
Bini, T; Bongiovanni, M; Chiesa, E; Cicconi, P; Di Biagio, A; Landonio, S; Meraviglia, P; Monforte, Ad; Testa, L; Tordato, F, 2005
)
0.61
"The SCOLTA project (Surveillance Cohort Long-term Toxicity Antiretrovirals) is a system for online surveying of adverse reactions to recently commercialized antiretroviral drugs and a "sentinel" for unexpected and late adverse reactions arising during any antiretroviral treatment (available at: http://www."( An Italian approach to postmarketing monitoring: preliminary results from the SCOLTA (Surveillance Cohort Long-Term Toxicity Antiretrovirals) project on the safety of lopinavir/ritonavir.
Armignacco, O; Bonfanti, P; Carradori, S; Magnani, C; Martinelli, C; Parruti, G; Quirino, T; Ricci, E, 2005
)
0.52
" No significant changes or differences in the concentration of fasting total cholesterol, low-density lipoprotein (LDL) cholesterol or total triglycerides or in the occurrence of adverse events were observed within or between the two groups."( The long-term pharmacokinetics and safety of adding low-dose ritonavir to a nelfinavir 1,250 mg twice-daily regimen in HIV-infected patients.
Andersen, AB; Black, FT; Gerstoft, J; Hansen, IM; Justesen, US; Klitgaard, NA; Mathiesen, LR; Pedersen, C, 2005
)
0.57
" The combination seems to be safe and the nelfinavir/ritonavir regimen could be an option in patients with low nelfinavir+M 8 concentrations."( The long-term pharmacokinetics and safety of adding low-dose ritonavir to a nelfinavir 1,250 mg twice-daily regimen in HIV-infected patients.
Andersen, AB; Black, FT; Gerstoft, J; Hansen, IM; Justesen, US; Klitgaard, NA; Mathiesen, LR; Pedersen, C, 2005
)
0.82
" Adverse events were described in 18 children."( Safety and antiviral response at 12 months of lopinavir/ritonavir therapy in human immunodeficiency virus-1-infected children experienced with three classes of antiretrovirals.
Cabrero, E; De José, MI; Dueñas, J; Fortuny, C; González-Montero, R; Mellado, MJ; Muñoz-Fernández, MA; Mur, A; Navarro, M; Otero, C; Pocheville, I; Ramos, JT, 2005
)
0.57
" Treatment was stopped in 18 patients; two from intolerance, two switched therapy, four as a result of serious adverse event-related death, and ten were lost to follow-up."( Efficacy and safety of indinavir/ritonavir 400/100 mg twice daily plus two nucleoside analogues in treatment-naive HIV-1-infected patients with CD4+ T-cell counts <200 cells/mm3: 96-week outcomes.
Anunnatsiri, S; Boonyaprawit, P; Chetchotisakd, P; Mootsikapun, P, 2005
)
0.61
"Our study demonstrates that indinavir/ritonavir 400/100 mg plus stavudine and lamivudine twice daily, the least expensive boosted protease inhibitor, appears to be effective and safe up to 96 weeks despite high baseline viraemia and low CD4+ cell count in antiretroviral-naive patients."( Efficacy and safety of indinavir/ritonavir 400/100 mg twice daily plus two nucleoside analogues in treatment-naive HIV-1-infected patients with CD4+ T-cell counts <200 cells/mm3: 96-week outcomes.
Anunnatsiri, S; Boonyaprawit, P; Chetchotisakd, P; Mootsikapun, P, 2005
)
0.88
"5 million adverse drug reaction (ADR) reports for 8620 drugs/biologics that are listed for 1191 Coding Symbols for Thesaurus of Adverse Reaction (COSTAR) terms of adverse effects."( Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
Benz, RD; Contrera, JF; Kruhlak, NL; Matthews, EJ; Weaver, JL, 2004
)
0.32
" Decrease in viral load of HIV (VC) has been assessed as primary endpoint and as secondary one, the increase of the numbers of CD4 lymphocytes, percentage of disease progression, adverse reactions and adherence."( [Clinical trial comparing efficacy and safety of four highly active antiretroviral therapy (HAART) in antiretroviral-naive treatment with advanced HIV infection].
Barberá Farré, JR; Beato Pérez, JL; Cuadra García-Tenorio, F; Geijo Martínez, MP; Maciá Martínez, MA; Marcos Sánchez, F; Martínez Alfaro, E; Moreno Mendaña, JM; Rodríguez Zapata, M; Sanz Moreno, J; Sanz Sanz, J; Solera Santos, J, 2006
)
0.33
" Treatment was discontinued due to adverse reactions: 24% in regimen 1, 48% in regimen 2, 26% in regimen 3 and 32% in regimen 4, without significant difference."( [Clinical trial comparing efficacy and safety of four highly active antiretroviral therapy (HAART) in antiretroviral-naive treatment with advanced HIV infection].
Barberá Farré, JR; Beato Pérez, JL; Cuadra García-Tenorio, F; Geijo Martínez, MP; Maciá Martínez, MA; Marcos Sánchez, F; Martínez Alfaro, E; Moreno Mendaña, JM; Rodríguez Zapata, M; Sanz Moreno, J; Sanz Sanz, J; Solera Santos, J, 2006
)
0.33
"In the HIV positive patients with advanced infection, efficacy between the four regimens of HAART is similar, but there is a tendency to require more withdrawal due to adverse effects in the RTV group than in those of IDV, the two used as single PI."( [Clinical trial comparing efficacy and safety of four highly active antiretroviral therapy (HAART) in antiretroviral-naive treatment with advanced HIV infection].
Barberá Farré, JR; Beato Pérez, JL; Cuadra García-Tenorio, F; Geijo Martínez, MP; Maciá Martínez, MA; Marcos Sánchez, F; Martínez Alfaro, E; Moreno Mendaña, JM; Rodríguez Zapata, M; Sanz Moreno, J; Sanz Sanz, J; Solera Santos, J, 2006
)
0.33
" Treatment was not discontinued in any patient because of adverse effects."( Atazanavir and lopinavir/ritonavir: pharmacokinetics, safety and efficacy of a promising double-boosted protease inhibitor regimen.
Azuaje, C; Crespo, M; Curran, A; Diaz, M; Feijoo, M; Lopez, RM; Ocaña, I; Pahissa, A; Pou, L; Ribera, E, 2006
)
0.64
" Subsequently, the patient developed a possible adverse skin reaction to his antiretrovirals and was hospitalized."( Carbamazepine toxicity induced by lopinavir/ritonavir and nelfinavir.
Bates, DE; Herman, RJ, 2006
)
0.59
" There were no severe adverse events (SAEs), and no subject was withdrawn due to BCV."( Single-dose safety and pharmacokinetics of brecanavir, a novel human immunodeficiency virus protease inhibitor.
Anderson, MT; Fernandez, P; Ford, SL; Johnson, MA; Murray, SC; Reddy, YS; Stein, DS, 2006
)
0.33
" Both regimens were reasonably well tolerated, although more gastrointestinal adverse events were reported with saquinavir-SGC/ritonavir."( Efficacy, safety and pharmacokinetics of once-daily saquinavir soft-gelatin capsule/ritonavir in antiretroviral-naive, HIV-infected patients.
Burnside, AF; Jayaweera, DT; Montaner, JS; Saag, MS; Schutz, M; Schwartz, R; Walmsley, S, 2006
)
0.76
" Gastrointestinal adverse effects were commonly associated with treatment failure in the saquinavir-SGC/ritonavir arm of the study."( Efficacy, safety and pharmacokinetics of once-daily saquinavir soft-gelatin capsule/ritonavir in antiretroviral-naive, HIV-infected patients.
Burnside, AF; Jayaweera, DT; Montaner, JS; Saag, MS; Schutz, M; Schwartz, R; Walmsley, S, 2006
)
0.77
" Together, these in vitro results indicate that combination with other antiretrovirals does not significantly increase the toxic potential of TFV in RPTECs."( In vitro cytotoxicity and mitochondrial toxicity of tenofovir alone and in combination with other antiretrovirals in human renal proximal tubule cells.
Alvarez, ML; Cihlar, T; Cordobilla, B; Domingo, JC; Domingo, P; Giralt, M; Guallar, J; López-Dupla, M; Sánchez de la Rosa, R; Saumoy, M; Torres, F; Vidal, F; Villarroya, F, 2006
)
0.33
" Secondary endpoints were presence of grade 3 and serious adverse events, clinical improvement, CD4 count and genotypic resistance to ritonavir/saquinavir."( Safety, efficacy and pharmacokinetics of ritonavir 400mg/saquinavir 400mg twice daily plus rifampicin combined therapy in HIV patients with tuberculosis.
da Silva Vieira, MA; de Jesus, Cda S; Ferreira Filho, M; Gonçalves Morgado, M; Lourenço, MC; Pereira Pinto, D; Rolla, VC; Werneck-Barroso, E, 2006
)
0.8
" During the antiretroviral therapy, 15 patients dropped out, 14 because of adverse events."( Safety, efficacy and pharmacokinetics of ritonavir 400mg/saquinavir 400mg twice daily plus rifampicin combined therapy in HIV patients with tuberculosis.
da Silva Vieira, MA; de Jesus, Cda S; Ferreira Filho, M; Gonçalves Morgado, M; Lourenço, MC; Pereira Pinto, D; Rolla, VC; Werneck-Barroso, E, 2006
)
0.6
"Therapeutic concentrations of the studied drugs and reduction of viral load were achieved; adverse events are the main limitation of use of a ritonavir/saquinavir regimen in treatment-naive patients, but its clinical benefits were evident."( Safety, efficacy and pharmacokinetics of ritonavir 400mg/saquinavir 400mg twice daily plus rifampicin combined therapy in HIV patients with tuberculosis.
da Silva Vieira, MA; de Jesus, Cda S; Ferreira Filho, M; Gonçalves Morgado, M; Lourenço, MC; Pereira Pinto, D; Rolla, VC; Werneck-Barroso, E, 2006
)
0.8
" BCV alone or in combination with RTV was well tolerated, with no serious adverse events reported."( Safety and pharmacokinetics of brecanavir, a novel human immunodeficiency virus type 1 protease inhibitor, following repeat administration with and without ritonavir in healthy adult subjects.
Anderson, MT; Ford, SL; Johnson, MA; Murray, SC; Ng-Cashin, J; Reddy, YS, 2007
)
0.54
" The most frequently reported adverse events (AEs) were diarrhea, nausea, vomiting, fatigue, and headache."( Efficacy and safety of three doses of tipranavir boosted with ritonavir in treatment-experienced HIV type-1 infected patients.
Arasteh, K; Cooper, D; Dohnanyi, C; Gathe, JC; Kohlbrenner, VM; Lalonde, RG; Lazzarin, A; Mayers, D; Pierone, G; Piliero, P; Rubio, R; Sabo, J, 2007
)
0.58
" Adverse event incidence was similar between treatments; headache and diarrhoea were more common with CPI(s)."( Efficacy and safety of TMC114/ritonavir in treatment-experienced HIV patients: 24-week results of POWER 1.
Esposito, R; Gatell, JM; Goffard, JC; Grinsztejn, B; Katlama, C; Parys, W; Pozniak, A; Rockstroh, J; Stoehr, A; Vangeneugden, T; Vetter, N; Yeni, P, 2007
)
0.63
" Although delayed autoinduction is not well established as a reason for adverse events in saquinavir therapy, this observation may be confirmed in the near future by increased use."( Dose reduction effective in alleviating symptoms of saquinavir toxicity.
Carlebach, A; Dauer, B; Haberl, A; Kurowski, M; Rottmann, C; Staszewski, S; Stephan, C; von Hentig, N, 2007
)
0.34
" Overall adverse event rates were similar in both arms, without significant differences among TMC114/r groups."( Week 24 efficacy and safety of TMC114/ritonavir in treatment-experienced HIV patients.
Berger, D; Chiliade, P; Colson, A; Conant, M; Gallant, J; Haubrich, R; Lefebvre, E; Nadler, J; Pierone, G; Saag, M; van Baelen, B; Wilkin, T, 2007
)
0.61
" In the darunavir-ritonavir group, rates of adverse events were mostly lower than or similar to those in the control group when corrected for treatment exposure."( Efficacy and safety of darunavir-ritonavir at week 48 in treatment-experienced patients with HIV-1 infection in POWER 1 and 2: a pooled subgroup analysis of data from two randomised trials.
Bellos, N; Clotet, B; Cohen, C; Cooper, D; Farthing, C; Goffard, JC; Haubrich, R; Jayaweera, D; Katlama, C; Lazzarin, A; Lefebvre, E; Markowitz, M; Molina, JM; Ruane, P; Spinosa-Guzman, S; Wilkin, T; Wöhrmann, A, 2007
)
0.95
" Seven infant adverse events were possibly treatment related (anemia, neutropenia, and hyperbilirubinemia)."( Clinical response and tolerability to and safety of saquinavir with low-dose ritonavir in human immunodeficiency virus type 1-infected mothers and their infants.
Acosta, EP; Bardeguez, A; Heckman, B; Huang, S; Hughes, MD; Jiménez, E; McSherry, G; Mofenson, L; Smith, B; Van Dyke, R; Watts, DH; Zorrilla, CD, 2007
)
0.57
"Of 25 subjects enrolled, scleral icterus was the most common adverse event (3 patients [12."( The safety, efficacy, and pharmacokinetic profile of a switch in antiretroviral therapy to saquinavir, ritonavir, and atazanavir alone for 48 weeks and a switch in the saquinavir formulation.
Cooper, DA; Emery, S; Law, M; MacRae, K; Mallon, PW; Satchell, C; Schutz, M; Williams, KM; Winston, A, 2007
)
0.55
" Brecanavir/ritonavir was well tolerated with no serious adverse events or clinically concerning changes in laboratory parameters."( Preliminary safety and efficacy data of brecanavir, a novel HIV-1 protease inhibitor: 24 week data from study HPR10006.
Garges, HP; Lalezari, JP; Tomkins, SA; Ward, DJ, 2007
)
0.72
" The US Food and Drug Administration's Adverse Event Reporting System was searched for reports of nephrolithiasis in HIV-infected patients taking an atazanavir-based regimen."( Atazanavir-associated nephrolithiasis: cases from the US Food and Drug Administration's Adverse Event Reporting System.
Birnkrant, DB; Chan-Tack, KM; Struble, KA; Truffa, MM, 2007
)
0.34
" Safety was evaluated by adverse events (AEs), grade 3/4 abnormalities, and serious AEs."( Long-term efficacy and safety of tipranavir boosted with ritonavir in HIV-1-infected patients failing multiple protease inhibitor regimens: 80-week data from a phase 2 study.
Goldman, M; Hathaway, B; Kazanjian, PH; Markowitz, M; Mayers, D; McCallister, S; Neubacher, D; Schwartz, R; Slater, LN; Valdez, H; Wheeler, D, 2007
)
0.58
" Grade 3/4 adverse events were similar across groups."( Phase 2 study of the safety and efficacy of vicriviroc, a CCR5 inhibitor, in HIV-1-Infected, treatment-experienced patients: AIDS clinical trials group 5211.
Coakley, E; Flexner, C; Godfrey, C; Greaves, WL; Gross, R; Gulick, RM; Hirsch, M; Hughes, MD; Krambrink, A; Kuritzkes, DR; Reichman, R; Skolnik, PR; Su, Z; Wilkin, TJ; Zolopa, A, 2007
)
0.34
" Safety data were generally similar between the groups; grade 3 or 4 adverse events occurred in 80 (27%) darunavir-ritonavir and 89 (30%) lopinavir-ritonavir patients."( Efficacy and safety of darunavir-ritonavir compared with that of lopinavir-ritonavir at 48 weeks in treatment-experienced, HIV-infected patients in TITAN: a randomised controlled phase III trial.
Banhegyi, D; Berger, D; de Béthune, MP; De Pauw, M; Lefebvre, E; Madruga, JV; McMurchie, M; Norris, D; Ruxrungtham, K; Spinosa-Guzman, S; Suter, F; Tomaka, F; Vangeneugden, T, 2007
)
0.83
" The most common adverse events (AEs) were diarrhea (14%), nasopharyngitis (11%), and nausea (10%)."( Safety and efficacy of darunavir (TMC114) with low-dose ritonavir in treatment-experienced patients: 24-week results of POWER 3.
Cohen, C; Grinsztejn, B; Katlama, C; Lefebvre, E; Meyer, SD; Miralles, D; Molina, JM; Parys, W; Pedro, Rde J; Timerman, A; Vangeneugden, T, 2007
)
0.59
" In this larger set of treatment-experienced patients, darunavir/r at a dose of 600/100 mg twice daily provided substantial virologic and immunologic responses and was generally safe and well tolerated."( Safety and efficacy of darunavir (TMC114) with low-dose ritonavir in treatment-experienced patients: 24-week results of POWER 3.
Cohen, C; Grinsztejn, B; Katlama, C; Lefebvre, E; Meyer, SD; Miralles, D; Molina, JM; Parys, W; Pedro, Rde J; Timerman, A; Vangeneugden, T, 2007
)
0.59
"Treatment with indinavir/ritonavir (IDV/RTV) is very effective but hampered by frequent development of IDV-associated adverse events (mainly nephrotoxicity and skin changes)."( Maintenance of indinavir by dose adjustment in HIV-1-infected patients with indinavir-related toxicity.
Burger, D; Hoffmann, C; Lambertz, I; Rockstroh, JK; Vogel, M; Voigt, E; Wasmuth, JC, 2007
)
0.64
"HIV-infected patients with any IDV/RTV regimen who suffer from IDV-related adverse events were included."( Maintenance of indinavir by dose adjustment in HIV-1-infected patients with indinavir-related toxicity.
Burger, D; Hoffmann, C; Lambertz, I; Rockstroh, JK; Vogel, M; Voigt, E; Wasmuth, JC, 2007
)
0.34
" Overall tolerability improved with respect to incidence and severity of adverse events."( Maintenance of indinavir by dose adjustment in HIV-1-infected patients with indinavir-related toxicity.
Burger, D; Hoffmann, C; Lambertz, I; Rockstroh, JK; Vogel, M; Voigt, E; Wasmuth, JC, 2007
)
0.34
" No statistically significant differences in adverse event incidence occurred between treatment groups, except for a higher vomiting rate in the 400/300 mg dose group."( High-dose lopinavir/ritonavir in highly treatment-experienced HIV-1 patients: efficacy, safety, and predictors of response.
Bernstein, B; Brun, SC; Eron, JJ; Flexner, C; Havlir, DV; Katlama, C; King, MS; Klein, CE; Letendre, SL; Podzamczer, D,
)
0.45
" In both groups, more patients with active co-infection than without co-infection had liver-related adverse events (AEs)."( Safety, tolerability, and efficacy of darunavir (TMC114) with low-dose ritonavir in treatment-experienced, hepatitis B or C co-infected patients in POWER 1 and 3.
Baxter, J; Clotet, B; Lefebvre, E; Murphy, R; Rachlis, A,
)
0.36
" A saquinavir C(min) > 2000 ng/ml was associated with an increased risk of gastrointestinal grade 3 or 4 adverse events and higher total cholesterol."( Pharmacokinetics of two randomized trials evaluating the safety and efficacy of indinavir, saquinavir and lopinavir in combination with low-dose ritonavir: the MaxCmin1 and 2 trials.
Cahn, P; Castagna, A; Clumeck, N; Dragsted, UB; Fox, Z; Gerstoft, J; Justesen, US; Losso, M; Lundgren, JD; Obel, N; Pedersen, C; Peters, B, 2007
)
0.54
" Adverse event-related discontinuations were 8% among ATV300/RTV-treated patients and <1% among ATV400-treated patients."( Efficacy and safety of atazanavir, with or without ritonavir, as part of once-daily highly active antiretroviral therapy regimens in antiretroviral-naive patients.
David, N; Hammond, J; Krantz, E; Malan, DR; McGrath, D; Wirtz, V, 2008
)
0.6
" Three infants (14%) had transient adverse events of grade 3 or more that were possibly related to study treatment but did not require permanent treatment discontinuation."( Pharmacokinetics, safety and efficacy of lopinavir/ritonavir in infants less than 6 months of age: 24 week results.
Capparelli, EV; Chadwick, EG; Chen, J; Hughes, M; Palumbo, P; Pinto, JA; Robbins, B; Rodman, JH; Serchuck, L; Smith, E; Yogev, R, 2008
)
0.6
" Data regarding demographics, clinical disease and antiretroviral treatment history, HIV-1 RNA copies/mL, CD4 T-cell counts [absolute (cells/microL) and percentages (%)], adverse events, clinical laboratory values, reasons for discontinuation of PIs, and concomitant medications were extracted from the database for PI-naive (first-line) and PI-experienced (second- or higher-line) PI use."( Long-term safety and effectiveness of ritonavir, nelfinavir, and lopinavir/ritonavir in antiretroviral-experienced HIV-infected children.
Burri, M; Nadal, D; Rode, R; Rudin, C; Shen, Y, 2008
)
0.62
"Long-term PI-based therapy seems to be safe and to result in durable virologic and immunologic effectiveness in HIV-1-infected antiretroviral-experienced children."( Long-term safety and effectiveness of ritonavir, nelfinavir, and lopinavir/ritonavir in antiretroviral-experienced HIV-infected children.
Burri, M; Nadal, D; Rode, R; Rudin, C; Shen, Y, 2008
)
0.62
"The study showed a high incidence of adverse events when a higher than standard dose of the new lopinavir/ritonavir tablets was combined with rifampicin."( High incidence of adverse events in healthy volunteers receiving rifampicin and adjusted doses of lopinavir/ritonavir tablets.
Aarnoutse, RE; Boeree, MJ; Burger, DM; Koopmans, PP; L'homme, RF; Nijland, HM; Rongen, GA; van Crevel, R; van Uden, P, 2008
)
0.77
" DRV/r had a lower incidence of possibly treatment-related grade 2-4 gastrointestinal-related adverse events (7 versus 14%) and treatment-related moderate-to-severe diarrhea (4 versus 10%) than LPV/r."( Efficacy and safety of once-daily darunavir/ritonavir versus lopinavir/ritonavir in treatment-naive HIV-1-infected patients at week 48.
Andrade-Villanueva, J; De Meyer, S; De Pauw, M; Dejesus, E; Fourie, J; Khanlou, H; Lefebvre, E; Ortiz, R; Spinosa-Guzman, S; van Lunzen, J; Vangeneugden, T; Voronin, E, 2008
)
0.61
" Serious adverse events were noted in 51 (12%) of 441 patients in the atazanavir/ritonavir group and in 42 (10%) of 437 patients in the lopinavir/ritonavir group."( Once-daily atazanavir/ritonavir versus twice-daily lopinavir/ritonavir, each in combination with tenofovir and emtricitabine, for management of antiretroviral-naive HIV-1-infected patients: 48 week efficacy and safety results of the CASTLE study.
Andrade-Villanueva, J; Chetchotisakd, P; Corral, J; David, N; Echevarria, J; Mancini, M; McGrath, D; Molina, JM; Moyle, G; Percival, L; Thiry, A; Yang, R, 2008
)
0.89
"100 patients were evaluated; 17 patients discontinued early including 6 for adverse events."( Evaluation of efficacy, safety, pharmacokinetics, and adherence in HIV-1-infected, antiretroviral-naïve patients treated with ritonavir-boosted atazanavir plus fixed-dose tenofovir DF/emtricitabine given once daily.
Cohen, C; Ebrahimi, R; Elion, R; Fisher, A; Flaherty, J; Kearney, B; McColl, D; Ortiz, R; Reddy, YS; Ruane, P; Ward, D,
)
0.34
" Vomiting, cough and diarrhea were the most frequent adverse events."( Efficacy, safety and tolerability of tipranavir coadministered with ritonavir in HIV-1-infected children and adolescents.
Cahn, P; Castelli-Gattinara, G; Flynn, PM; Fortuny, C; Giaquinto, C; Jelaska, A; Mikl, J; Negra, MD; Ruan, PK; Salazar, JC; Smith, ME; Yogev, R, 2008
)
0.58
" In both studies, BILR 355 appeared to be safe and well tolerated in healthy volunteers when the outcomes in the treated volunteers were compared with those in the placebo group."( Pharmacokinetic and safety evaluation of BILR 355, a second-generation nonnucleoside reverse transcriptase inhibitor, in healthy volunteers.
Hattox, S; Huang, F; Koenen-Bergmann, M; Macgregor, TR; Ring, A; Robinson, P, 2008
)
0.35
" A box warning is a labeling statement about serious adverse events leading to significant injury and/or death."( Intracranial hemorrhage and liver-associated deaths associated with tipranavir/ritonavir: review of cases from the FDA's Adverse Event Reporting System.
Birnkrant, DB; Chan-Tack, KM; Struble, KA, 2008
)
0.57
" Twenty-five patients (62%) experienced at least one adverse event."( Three-year outcome data of second-line antiretroviral therapy in Ugandan adults: good virological response but high rate of toxicity.
Bates, M; Castelnuovo, B; Colebunders, R; John, L; Kamya, MR; Lutwama, F; Ronald, A; Spacek, LA,
)
0.13
" In the studies performed to date, darunavir has been well tolerated, with a better profile than that of the PIs used in control groups in terms of adverse effects such as diarrhea, gastrointestinal tolerability and lipid alterations."( [Safety and tolerability of darunavir].
Antela López, A, 2008
)
0.35
" Common clinical adverse events (AEs) were diarrhoea, nausea, fatigue and headache."( Antiviral activity and safety of aplaviroc, a CCR5 antagonist, in combination with lopinavir/ritonavir in HIV-infected, therapy-naïve patients: results of the EPIC study (CCR100136).
Adkison, KK; Berger, D; Bonny, T; Cimoch, P; Lamarca, A; Lazzarin, A; Madison, SJ; McCarty, D; Millard, J; Nichols, WG; Salvato, P; Smaill, FM; Teofilo, E; Yeni, P, 2009
)
0.57
"Ritonavir-boosted atazanavir is associated with greater virologic control and immune response through 52 weeks compared to non-boosted atazanavir, without greater risk of adverse events except elevated bilirubin."( Efficacy and safety of ritonavir-boosted and unboosted atazanavir among antiretroviral-naïve patients.
Antoniskis, D; Blake, W; Dobrinich, R; Dodge, W; Horberg, M; Hurley, L; Klein, D; Kovach, D; Mogyoros, M; Silverberg, M; Towner, W,
)
1.88
" Adverse events were limited to transient grade 3 neutropenia in 3 subjects."( Early initiation of lopinavir/ritonavir in infants less than 6 weeks of age: pharmacokinetics and 24-week safety and efficacy.
Alvero, CG; Browning, R; Capparelli, E; Chadwick, EG; Hazra, R; Heckman, BE; Hughes, MD; Luzuriaga, K; Palumbo, P; Pinto, J; Purdue, L; Robbins, B; Rodman, J; Serchuck, L; Yogev, R, 2009
)
0.64
" Adverse events (AEs) associated with the study medication occurred in 21% of patients."( Fosamprenavir (GW433908)/ritonavir in HIV-infected patients: efficacy and safety results from the Spanish Expanded Access Program.
Luque, I; Mallolas, J; Pérez-Elias, MJ; Rodríguez-Alcántara, F; Sánchez-Conde, M; Soriano, V, 2009
)
0.66
"Ritonavir-boosted fosamprenavir as part of antiretroviral therapy is a potent, safe treatment in real-life clinical circumstances."( Fosamprenavir (GW433908)/ritonavir in HIV-infected patients: efficacy and safety results from the Spanish Expanded Access Program.
Luque, I; Mallolas, J; Pérez-Elias, MJ; Rodríguez-Alcántara, F; Sánchez-Conde, M; Soriano, V, 2009
)
2.1
" Rates of discontinuation and adverse events, including diarrhea, were similar between arms."( A once-daily lopinavir/ritonavir-based regimen is noninferior to twice-daily dosing and results in similar safety and tolerability in antiretroviral-naive subjects through 48 weeks.
Bernstein, B; Cohen, DE; da Silva, BA; Fredrick, L; Gathe, J; Gibbs, S; Loutfy, MR; Marsh, T; Naylor, C; Podzamczer, D; Rubio, R, 2009
)
0.66
" There is a complex, combined effect of HIV infection plus antiretroviral treatment on the incidence of gastrointestinal symptoms, and, for some trials, the majority of gastrointestinal adverse events may not be related to antiretroviral treatment."( Risk factors for gastrointestinal adverse events in HIV treated and untreated patients.
Balkin, A; Hill, A,
)
0.13
" There were no laboratory or QTc interval changes reportable as adverse events."( A phase I study to explore the activity and safety of SCH532706, a small molecule chemokine receptor-5 antagonist in HIV type-1-infected patients.
Cooper, DA; Emery, S; MacRae, K; McCarthy, MC; Norris, R; Pett, SL; Strizki, JM; Tendolkar, A; Williams, KM, 2009
)
0.35
"Double-boosted SQV+LPV/r was an effective and safe alternative for a second-line regimen in children."( Safety and efficacy of a double-boosted protease inhibitor combination, saquinavir and lopinavir/ritonavir, in pretreated children at 96 weeks.
Ananworanich, J; Boonrak, P; Bunupuradah, T; Burger, D; Engchanil, C; Kosalaraksa, P; Lumbiganon, P; Mahanontharit, A; Mengthaisong, T; Puthanakit, T; Ruxrungtham, K; Tompkins, E; van der Lugt, J, 2009
)
0.57
"Saquinavir exposure in the new tablet formulation generates adequate saquinavir concentrations throughout the course of pregnancy and is safe to use; therefore, no dose adjustment during pregnancy is needed."( The pharmacokinetics, safety and efficacy of boosted saquinavir tablets in HIV type-1-infected pregnant women.
Burger, D; Colbers, A; Hawkins, D; Koopmans, P; Molto, J; Richter, C; Ruxrungtham, K; Schutz, M; van der Ende, M; van der Lugt, J; Vogel, M; Wyen, C, 2009
)
0.35
" Diarrhea was the most frequently reported adverse event."( Long-term efficacy and safety of fosamprenavir plus ritonavir versus lopinavir/ritonavir in combination with abacavir/lamivudine over 144 weeks.
Baril, JG; Duiculescu, D; Estrada, V; Lim, ML; Logue, K; Pharo, C; Plettenberg, A; Pulido, F; Schewe, K; Vavro, C; Yau, L,
)
0.38
" Data on CD4 cell count, HIV viral load, metabolic parameters and adverse events of grade 3-4 are collected through an on-line system every six months."( Efficacy and safety of boosted and unboosted atazanavir-containing antiretroviral regimens in real life: results from a multicentre cohort study.
Abeli, C; Bonfanti, P; Gianelli, E; Giuntini, R; Grosso, C; Madeddu, G; Marconi, P; Martinelli, C; Palvarini, L; Pellicano, G; Penco, G; Quirino, T; Ricci, E; Vichi, F, 2010
)
0.36
"Our results suggest that, in unselected clinical settings, ATV-containing antiretroviral therapy is durable and safe in both its formulations."( Efficacy and safety of boosted and unboosted atazanavir-containing antiretroviral regimens in real life: results from a multicentre cohort study.
Abeli, C; Bonfanti, P; Gianelli, E; Giuntini, R; Grosso, C; Madeddu, G; Marconi, P; Martinelli, C; Palvarini, L; Pellicano, G; Penco, G; Quirino, T; Ricci, E; Vichi, F, 2010
)
0.36
" Grade 2-4 treatment-emergent adverse events at least possibly related to DRV/r (>or=2% incidence, excluding laboratory abnormalities) were diarrhoea (3%), vomiting (3%), nausea (2%) and headache (2%)."( Efficacy and safety of darunavir/ritonavir in treatment-experienced HIV type-1 patients in the POWER 1, 2 and 3 trials at week 96.
Arastéh, K; De Meyer, S; Grinsztejn, B; Hoy, J; Jayaweera, D; Pozniak, A; Roberts, A; Tomaka, F; Vangeneugden, T; Yeni, P, 2009
)
0.63
" We describe two patients who experienced serious quetiapine adverse effects potentially mediated through an interaction with ritonavir-boosted atazanavir."( Clinically significant adverse events from a drug interaction between quetiapine and atazanavir-ritonavir in two patients.
McCoy, C; Pollack, TM; Stead, W, 2009
)
0.78
" Treatment-related gastrointestinal adverse events were greater in patients taking lopinavir/ritonavir."( Once-daily atazanavir/ritonavir compared with twice-daily lopinavir/ritonavir, each in combination with tenofovir and emtricitabine, for management of antiretroviral-naive HIV-1-infected patients: 96-week efficacy and safety results of the CASTLE study.
Absalon, J; Andrade-Villanueva, J; Chetchotisakd, P; Corral, J; David, N; Echevarria, J; Lataillade, M; Mancini, M; McGrath, D; Molina, JM; Moyle, G; Percival, L; Wirtz, V; Yang, R, 2010
)
0.89
" Atazanavir/ritonavir had a better lipid profile and fewer gastrointestinal adverse events than lopinavir/ritonavir."( Once-daily atazanavir/ritonavir compared with twice-daily lopinavir/ritonavir, each in combination with tenofovir and emtricitabine, for management of antiretroviral-naive HIV-1-infected patients: 96-week efficacy and safety results of the CASTLE study.
Absalon, J; Andrade-Villanueva, J; Chetchotisakd, P; Corral, J; David, N; Echevarria, J; Lataillade, M; Mancini, M; McGrath, D; Molina, JM; Moyle, G; Percival, L; Wirtz, V; Yang, R, 2010
)
1.05
" Adverse event-related discontinuations occurred among 8% receiving ATV300/r and 3% receiving ATV400."( 96-week efficacy and safety of atazanavir, with and without ritonavir, in a HAART regimen in treatment-naive patients.
David, N; Iloeje, UH; Krantz, E; Malan, DR; Mathew, M; McGrath, D,
)
0.37
" The occurrence of treatment-related moderate/severe adverse events was comparable for all events except nausea, which was reported more frequently among BID-treated subjects."( Similar safety and efficacy of once- and twice-daily lopinavir/ritonavir tablets in treatment-experienced HIV-1-infected subjects at 48 weeks.
Andrade-Villanueva, J; Badal-Faesen, S; Bernstein, BM; Fredrick, LM; Gathe, J; Gaultier, IA; Mingrone, H; Podsadecki, TJ; Woodward, WC; Zajdenverg, R, 2010
)
0.6
" Furthermore, etravirine was shown to be safe and well-tolerated over this period."( Etravirine in combination with darunavir/ritonavir and optimized background regimen results in suppression of HIV replication in treatment-experienced patients. Evaluation of Katlama C, Haubrich R, Lalezari J, et al. Efficacy and safety of etravirine in t
Hull, MW; Montaner, JS, 2010
)
0.63
"Long-term treatment with LPV/r-based cART is safe and effective in HIV-1-infected children."( Long-term safety and effectiveness of lopinavir/ritonavir in antiretroviral-experienced HIV-1-infected children.
Bucher, HC; Nadal, D; Rickenbach, M; Rudin, C; Wolbers, M, 2010
)
0.62
" Hyperbilirubinemia (13%), diarrhea (4%), nausea (2%), and rash (2%) were the most frequent drug-related Grade 2-4 adverse events."( Safety and efficacy of a 36-week induction regimen of abacavir/lamivudine and ritonavir-boosted atazanavir in HIV-infected patients.
Bellos, N; DeJesus, E; Murphy, D; Patel, LG; Ross, LL; Shaefer, MS; Squires, KE; Sutherland-Phillips, DH; Wannamaker, PG; Young, B; Zhao, HH,
)
0.36
" An understanding of structure-activity relationships (SARs) of chemicals can make a significant contribution to the identification of potential toxic effects early in the drug development process and aid in avoiding such problems."( Developing structure-activity relationships for the prediction of hepatotoxicity.
Fisk, L; Greene, N; Naven, RT; Note, RR; Patel, ML; Pelletier, DJ, 2010
)
0.36
" Through 96 weeks, 77 subjects from each group discontinued prematurely; adverse or HIV-related events contributed to discontinuation of 36 subjects overall, with no significant differences between treatment groups."( Short communication: Comparable safety and efficacy with once-daily versus twice-daily dosing of lopinavir/ritonavir tablets with emtricitabine + tenofovir DF in antiretroviral-naïve, HIV type 1-infected subjects: 96 week final results of the randomized t
Bernstein, B; Cohen, D; da Silva, B; Fredrick, L; González-García, J; Johnson, M; Naylor, C; Sloan, L, 2010
)
0.57
"Darunavir/ritonavir at 900/100 mg once daily is highly effective, safe and well tolerated in treatment-experienced patients with no darunavir resistance, both in early salvage and switch strategies."( Efficacy, safety and pharmacokinetics of 900/100 mg of darunavir/ritonavir once daily in treatment-experienced patients.
Crespo, M; Curran, A; Deig, E; Domingo, P; Gutirerrez, M; Imaz, A; Lopez, RM; Mateo, G; Ocaña, I; Ribera, E, 2010
)
1
"Of 256 subjects enrolled, clinician-reported grade 1-4 adverse events of the nervous system (all cause) were seen in 16% of patients in each treatment arm."( Neuropsychiatric adverse events with ritonavir-boosted darunavir monotherapy in HIV-infected individuals: a randomised prospective study.
Arribas, J; Fätkenheuer, G; Hill, A; Moecklinghoff, C; van Delft, Y; Winston, A,
)
0.4
"In this exploratory analysis, no differences in the evolution of neuropsychiatric adverse events over 48 weeks are observed in HIV-infected subjects randomised to switch antiretroviral therapy to darunavir/ritonavir with or without nucleoside reverse transcriptase inhibitors."( Neuropsychiatric adverse events with ritonavir-boosted darunavir monotherapy in HIV-infected individuals: a randomised prospective study.
Arribas, J; Fätkenheuer, G; Hill, A; Moecklinghoff, C; van Delft, Y; Winston, A,
)
0.59
"Ritonavir-related adverse events have been reported in patients taking tipranavir/ritonavir at the licensed dosage of 500/200 mg twice daily (bid)."( Efficacy and safety of ritonavir dose reduction based on the tipranavir inhibitory quotient in HIV-infected patients on salvage antiretroviral therapy with tipranavir/ritonavir.
Cedeño, S; Clotet, B; Miranda, C; Moltó, J; Negredo, E; Santos, JR; Valle, M; Videla, S; Yritia, M, 2010
)
2.11
" Atazanavir (ATV) was well tolerated with no unanticipated adverse events."( Safety and exposure of once-daily ritonavir-boosted atazanavir in HIV-infected pregnant women.
Bertz, R; Botes, M; Child, M; Conradie, F; Eley, T; Hu, W; Josipovic, D; McGrath, D; Osiyemi, O; Vandeloise, E; Wirtz, V; Zorrilla, C, 2011
)
0.65
" The most frequently reported adverse event was increased bilirubin (16."( Treatment durability, effectiveness, and safety with atazanavir/ritonavir-based HAART regimen in treatment-naïve HIV-infected patients.
Baril, JG; Boulerice, F; Lalonde, R; Loutfy, M; Rachlis, A; Sampalis, JS; Tremblay, C; Trottier, B,
)
0.37
" Thus, ART regimens containing FPV/r QD may be considered safe in HIV/HCV-coinfected patients, including those with cirrhosis."( Liver toxicity of antiretroviral combinations including fosamprenavir plus ritonavir 1400/100 mg once daily in HIV/hepatitis C virus-coinfected patients.
Delgado-Fernández, M; García-Figueras, C; López-Cortés, LF; Macías, J; Márquez-Solero, M; Martínez-Pérez, MA; Mata, R; Merchante, N; Merino, D; Omar, M; Pasquau, J; Pineda, JA; Ríos-Villegas, MJ; Rivero, A, 2011
)
0.6
" Safety was assessed with the incidence of treatment-emergent adverse events (AE)."( Pilot, randomized study assessing safety, tolerability and efficacy of simplified LPV/r maintenance therapy in HIV patients on the 1 PI-based regimen.
Ackad, N; Andrade-Villanueva, J; Bortolozzi, R; Cahn, P; Casiró, AD; Cassetti, I; Junod, P; Krolewiecki, A; Longo, N; Lupo, SH; Montaner, J; Patterson, P; Rampakakis, E; Sampalis, JS; Sierra-Madero, J, 2011
)
0.37
"5%) subjects discontinued ART due to this adverse event."( Liver toxicity associated with antiretroviral therapy including efavirenz or ritonavir-boosted protease inhibitors in a cohort of HIV/hepatitis C virus co-infected patients.
Camacho, A; Collado, A; de Los Santos-Gil, I; González-Serrano, M; Macías, J; Merino, D; Mira, JA; Neukam, K; Parra-García, G; Pineda, JA; Rivero, A; Ruiz-Morales, J; Torres-Cornejo, A, 2011
)
0.6
" Frequency of adverse events (AEs) was similar between arms, with 88."( A randomised comparison of safety and efficacy of nevirapine vs. atazanavir/ritonavir combined with tenofovir/emtricitabine in treatment-naïve patients.
Bhatti, L; Conner, C; Dejesus, E; Mills, A; Storfer, S, 2011
)
0.6
" Three patients allocated elvitegravir had serious adverse events related to study drugs compared with seven assigned raltegravir; two and eight patients died, respectively."( Efficacy and safety of once daily elvitegravir versus twice daily raltegravir in treatment-experienced patients with HIV-1 receiving a ritonavir-boosted protease inhibitor: randomised, double-blind, phase 3, non-inferiority study.
Andrade-Villanueva, J; Cheng, AK; Chuck, SL; Clotet, B; Clumeck, N; Lamarca, A; Liu, YP; Margot, N; Molina, JM; Zhong, L, 2012
)
0.58
" The occurrence of treatment-related, moderate/severe adverse events was similar between treatment groups through 48 weeks of treatment."( Examination of noninferiority, safety, and tolerability of lopinavir/ritonavir and raltegravir compared with lopinavir/ritonavir and tenofovir/ emtricitabine in antiretroviral-naïve subjects: the progress study, 48-week results.
Fredrick, LM; Gathe, J; Lawal, A; Nilius, AM; Podsadecki, TJ; Pulido, F; Reynes, J; Soto-Malave, R; Tian, M,
)
0.37
" The incidence of treatment-related adverse drug reactions (ADRs) and laboratory abnormalities were comparable across gender, age, and race subgroups."( Effect of baseline characteristics on the efficacy and safety of once-daily darunavir/ ritonavir in HIV-1-infected, treatment-naïve ARTEMIS patients at week 96.
Ballesteros, J; DeMasi, R; Domingo, P; Flamm, J; Fourie, J; Kilby, D; Lavreys, L; Lazzarin, A; Rodriguez-French, A; Sosa, N; Spinosa-Guzman, S; Van De Casteele, T,
)
0.35
"0%) men discontinued ETR due to adverse events."( Efficacy and safety outcomes among treatment-experienced women and men treated with etravirine in gender, race and clinical experience.
Hodder, S; Jayaweera, D; Mrus, J; Ryan, R; Witek, J, 2012
)
0.38
" Three patients discontinued antiretroviral therapy due to mild adverse events."( Efficacy and safety of once-daily ritonavir-boosted darunavir and abacavir/lamivudine for treatment-naïve patients: a pilot study.
Gatanaga, H; Kikuchi, Y; Nishijima, T; Oka, S; Teruya, K; Tsukada, K, 2012
)
0.66
" There were no serious adverse events (AEs) in either arm."( A switch in therapy to a reverse transcriptase inhibitor sparing combination of lopinavir/ritonavir and raltegravir in virologically suppressed HIV-infected patients: a pilot randomized trial to assess efficacy and safety profile: the KITE study.
Del Rio, C; Easley, KA; Eaton, ME; Lennox, JL; Ofotokun, I; Sanford, SE; Shenvi, N; Sheth, AN; White, K, 2012
)
0.6
"Regimens including EFV, NVP, or PI/r are generally safe in treatment-naïve HIV/HCV-coinfected patients."( Liver toxicity of initial antiretroviral drug regimens including two nucleoside analogs plus one non-nucleoside analog or one ritonavir-boosted protease inhibitor in HIV/HCV-coinfected patients.
Cartón, JA; Clotet, B; Crespo, M; de Los Santos, I; Domingo, P; Iribarren, JA; Jiménez-Expósito, MJ; Knobel, H; López-Cortés, LF; Macías, J; Mallolas, J; Moreno, S; Neukam, K; Ortega, E; Pineda, JA,
)
0.34
"Treatment simplification with DRV/r monotherapy seems safe and effective in routine clinical practice."( Antiretroviral simplification with darunavir/ritonavir monotherapy in routine clinical practice: safety, effectiveness, and impact on lipid profile.
Bravo, I; Clotet, B; Llibre, JM; Moltó, J; Negredo, E; Ornelas, A; Paredes, R; Santos, JR, 2012
)
0.64
" None of 9 severe adverse events were LPV/r- or liver-related."( A pilot, prospective, open-label simplification study to evaluate the safety, efficacy, and pharmacokinetics of once-daily lopinavir-ritonavir monotherapy in HIV-HCV coinfected patients: the MONOCO study.
Ackad, N; Conway, B; Cooper, C; la Porte, C; Sampalis, J; Tossonian, H,
)
0.34
"Once-daily LPV/r monotherapy in HIV-HCV coinfected individuals offers a safe and effective approach to the management of the HIV infection, with a predictable pharmacokinetic profile."( A pilot, prospective, open-label simplification study to evaluate the safety, efficacy, and pharmacokinetics of once-daily lopinavir-ritonavir monotherapy in HIV-HCV coinfected patients: the MONOCO study.
Ackad, N; Conway, B; Cooper, C; la Porte, C; Sampalis, J; Tossonian, H,
)
0.34
"During the study no serious adverse events were reported."( Efficacy and safety of a dual boosted protease inhibitor-based regimen, atazanavir and fosamprenavir/ritonavir, against HIV: experience in a pediatric population.
Adorni, F; Galli, M; Giacomet, V; Mameli, C; Rusconi, S; Viganò, A; Viganò, O; Zuccotti, GV, 2012
)
0.59
" Four grade 3 and no grade 4 adverse events were reported."( Pharmacokinetics and 48-week safety and efficacy of generic lopinavir/ritonavir in Thai HIV-infected patients.
Ananworanich, J; Avihingsanon, A; Burger, DM; Gorowara, M; Lange, JM; Phanuphak, P; Ramautarsing, RA; Ruxthungtham, K; Sophonphan, J; van der Lugt, J, 2013
)
0.62
" Only one severe adverse event occurred due to saquinavir (maternal grade 3 hepatotoxicity)."( Effectiveness and safety of saquinavir/ritonavir in HIV-infected pregnant women: INEMA cohort.
Brunet, C; Bui, E; De Saint Martin, L; Jovelin, T; Le Moal, G; Perfezou, P; Raffi, F; Reliquet, V; Venisse, N; Winer, N, 2012
)
0.65
"SQV/r 1000/100mg twice daily seems to be effective and safe in HIV-infected pregnant women with adequate saquinavir C(min)."( Effectiveness and safety of saquinavir/ritonavir in HIV-infected pregnant women: INEMA cohort.
Brunet, C; Bui, E; De Saint Martin, L; Jovelin, T; Le Moal, G; Perfezou, P; Raffi, F; Reliquet, V; Venisse, N; Winer, N, 2012
)
0.65
" Although adverse events (AEs) were the main known reason for discontinuation, no unexpected AEs were observed."( Long-term efficacy and safety of atazanavir/ritonavir treatment in a real-life cohort of treatment-experienced patients with HIV type 1 infection.
Brockmeyer, N; Dupke, S; Eychenne, JL; Jansen, K; Jimenez-Exposito, MJ; Nakonz, T; Pugliese, P; Sönnerborg, A; Svedhem, V; Thalme, A, 2013
)
0.65
" Significantly fewer discontinuations because of adverse events were observed with DRV/r (4."( Final 192-week efficacy and safety of once-daily darunavir/ritonavir compared with lopinavir/ritonavir in HIV-1-infected treatment-naïve patients in the ARTEMIS trial.
DeJesus, E; Khanlou, H; Lathouwers, E; Lefebvre, E; Opsomer, M; Orkin, C; Stoehr, A; Supparatpinyo, K; Tomaka, F; Van de Casteele, T, 2013
)
0.63
" Efficacy (HIV-1 RNA levels, CD4+ T-cell counts) and safety and tolerability (treatment discontinuation, treatment-related adverse events [AE], and clinical laboratory abnormalities) at 48 weeks were assessed for total women, women by age (≥50, <50 years) and body mass index (BMI; <25, ≥25 to <30, ≥30 kg/m2), and sex."( Meta-analysis of the safety, tolerability, and efficacy of lopinavir/ritonavir-containing antiretroviral therapy in HIV-1-infected women.
Fredrick, L; Hermes, A; Martinez, M; Norton, M; Pasley, M; Squires, K; Trinh, R,
)
0.37
" The drug combination appears to be generally safe and well tolerated."( Abacavir/lamivudine fixed-dose combination with ritonavir-boosted darunavir: a safe and efficacious regimen for HIV therapy.
Boissonnault, M; Dion, H; Gallant, S; Lavoie, S; Legault, D; Longpré, D; Machouf, N; Nguyen, VK; Thomas, R; Trottier, B; Vézina, S,
)
0.39
" Seven moderate to severe adverse events were recorded (including four renal colics, possibly treatment-related) in six patients."( Safety and feasibility of treatment simplification to atazanavir/ritonavir + lamivudine in HIV-infected patients on stable treatment with two nucleos(t)ide reverse transcriptase inhibitors + atazanavir/ritonavir with virological suppression (Atazanavir an
Cauda, R; Ciccarelli, N; Cingolani, A; Colafigli, M; D'Avino, A; De Luca, A; Di Giambenedetto, S; Fabbiani, M; Farina, S; Mondi, A; Murri, R; Navarra, P; Sidella, L; Tamburrini, E, 2013
)
0.63
"Simplification to atazanavir/ritonavir + lamivudine was apparently safe and associated with rare virological failure, without resistance selection."( Safety and feasibility of treatment simplification to atazanavir/ritonavir + lamivudine in HIV-infected patients on stable treatment with two nucleos(t)ide reverse transcriptase inhibitors + atazanavir/ritonavir with virological suppression (Atazanavir an
Cauda, R; Ciccarelli, N; Cingolani, A; Colafigli, M; D'Avino, A; De Luca, A; Di Giambenedetto, S; Fabbiani, M; Farina, S; Mondi, A; Murri, R; Navarra, P; Sidella, L; Tamburrini, E, 2013
)
0.92
"The Surveillance Cohort Long-Term Toxicity Antiretrovirals (SCOLTA) project is a prospective, observational, multicenter cohort created to assess the incidence of adverse events in patients receiving new antiretroviral drugs."( Safety and durability in a cohort of HIV-1 positive patients treated with once and twice daily darunavir-based therapy (SCOLTA Project).
Bonfanti, P; Carenzi, L; Celesia, BM; Corsi, P; De Socio, G; Di Biagio, A; Franzetti, M; Grosso, C; Guastavigna, M; Madeddu, G; Maggi, P; Martinelli, C; Menzaghi, B; Molteni, C; Orofino, G; Parruti, G; Penco, G; Quirino, T; Ricci, E; Vichi, F, 2013
)
0.39
"Our study showed that DRV/r administrated both once daily or twice daily was safe and well tolerated with few discontinuations due to adverse events."( Safety and durability in a cohort of HIV-1 positive patients treated with once and twice daily darunavir-based therapy (SCOLTA Project).
Bonfanti, P; Carenzi, L; Celesia, BM; Corsi, P; De Socio, G; Di Biagio, A; Franzetti, M; Grosso, C; Guastavigna, M; Madeddu, G; Maggi, P; Martinelli, C; Menzaghi, B; Molteni, C; Orofino, G; Parruti, G; Penco, G; Quirino, T; Ricci, E; Vichi, F, 2013
)
0.39
"Once-daily DRV/r (900/100 mg) was efficacious in pretreated patients, with safe responses."( Efficacy, safety, and pharmacokinetics of once-daily boosted darunavir in pretreated HIV-infected patients.
Benalycherif, A; De Truchis, P; Delassus, JL; Descamps, D; Duvivier, C; Landman, R; Peytavin, G; Tegna, L; Weiss, L; Zucman, D, 2013
)
0.39
" These findings suggest that HIV-infected cancer patients receiving nelfinavir might experience both enhanced antitumor efficacy and unexpected adverse toxicity given the role of MRP4/ABCC4 in exporting nucleoside-based antiretroviral medications and cancer chemotherapeutics."( Human immunodeficiency virus protease inhibitors interact with ATP binding cassette transporter 4/multidrug resistance protein 4: a basis for unanticipated enhanced cytotoxicity.
Ambudkar, SV; Cheepala, SB; Ekins, S; Fukuda, Y; Schuetz, JD; Sparreboom, A; Takenaka, K; Wu, CP, 2013
)
0.39
" The most common adverse events were diarrhea, upper respiratory tract infection, gastroenteritis and otitis media."( Pharmacokinetics, safety and antiviral activity of fosamprenavir/ritonavir-containing regimens in HIV-infected children aged 4 weeks to 2 years-48-week study data.
Cassim, H; Cheng, K; Cotton, M; Ford, SL; Garges, HP; Givens, N; Lou, Y; Pavía-Ruz, N; Perger, T; Ross, LL; Sievers, J; Wire, MB, 2014
)
0.64
" The most common adverse events were vomiting, cough, and diarrhea; 18 subjects experienced serious adverse events, including 9 with suspected abacavir hypersensitivity."( Pharmacokinetics and 48-week safety and antiviral activity of fosamprenavir-containing regimens in HIV-infected 2- to 18-year-old children.
Cheng, K; Cotton, M; Duiculescu, D; Ford, SL; Fortuny, C; Garges, HP; Givens, N; Lou, Y; Perger, T; Ross, LL; Sievers, J; Tamarirt, DP; Wire, MB, 2014
)
0.4
" Drug-related adverse events (investigator defined) were similar across all age groups (55-65%)."( Efficacy and safety of tipranavir coadministered with ritonavir in HIV-1-infected children and adolescents: 5 years of experience.
Cahn, P; De Aquino, MZ; Della Negra, M; Jelaska, A; Mikl, J; Robinson, PA; Salazar, JC, 2014
)
0.65
" The most common (≥ 10%) treatment-emergent adverse events were fatigue (27."( Antiviral activity, pharmacokinetics, and safety of the HIV-1 protease inhibitor TMC310911, coadministered with ritonavir, in treatment-naive HIV-1-infected patients.
Arastéh, K; Dierynck, I; Hoetelmans, RM; Jacquemyn, B; Mariën, K; Meyvisch, P; Schürmann, D; Simmen, K; Smyej, I; Stellbrink, HJ; Stephan, C; Truyers, C; Verloes, R, 2014
)
0.61
"5 log10 copies/mL decrease in plasma HIV-1 RNA) at all evaluated doses, and treatment was generally safe and well tolerated."( Antiviral activity, pharmacokinetics, and safety of the HIV-1 protease inhibitor TMC310911, coadministered with ritonavir, in treatment-naive HIV-1-infected patients.
Arastéh, K; Dierynck, I; Hoetelmans, RM; Jacquemyn, B; Mariën, K; Meyvisch, P; Schürmann, D; Simmen, K; Smyej, I; Stellbrink, HJ; Stephan, C; Truyers, C; Verloes, R, 2014
)
0.61
" In both studies, most treatment-emergent adverse events were related to gastrointestinal system."( Safety and pharmacokinetics of the HIV-1 protease inhibitor TMC310911 coadministered with ritonavir in healthy participants: results from 2 phase 1 studies.
Dierynck, I; Hoetelmans, RM; Jacquemyn, B; Marien, K; Meyvisch, P; Simmen, K; Smyej, I; Verloes, R, 2014
)
0.62
" TMC310911 was generally safe and tolerable when administered with or without ritonavir."( Safety and pharmacokinetics of the HIV-1 protease inhibitor TMC310911 coadministered with ritonavir in healthy participants: results from 2 phase 1 studies.
Dierynck, I; Hoetelmans, RM; Jacquemyn, B; Marien, K; Meyvisch, P; Simmen, K; Smyej, I; Verloes, R, 2014
)
0.85
"2% receiving the increased dose, discontinued treatment because of adverse events (p=0."( A randomized controlled trial to assess safety, tolerability, and antepartum viral load with increased lopinavir/ritonavir dosage in pregnancy.
Bonafe, SM; Castelo, A; Costa, DA; Machado, RH; Pott-Junior, H; Senise, JF; Vaz, MJ, 2013
)
0.6
" Two patients withdrew prematurely from study medications due to adverse events."( Efficacy and safety of danoprevir-ritonavir plus peginterferon alfa-2a-ribavirin in hepatitis C virus genotype 1 prior null responders.
Brennan, BJ; Gane, EJ; Kulkarni, R; Larrey, DG; Le Pogam, S; Morcos, PN; Nájera, I; Petric, R; Rouzier, R; Shulman, NS; Smith, P; Tran, JQ; Wiercinska-Drapalo, A; Yetzer, ES; Zhang, Y, 2014
)
0.68
" The frequency of serious adverse effects was investigated."( Efficacy and safety of once-daily maraviroc plus ritonavir-boosted darunavir in pretreated HIV-infected patients in a real-life setting.
Bancalero, P; Chueca, N; García, C; Jiménez, P; Macías, J; Márquez, M; Merino, D; Muñoz, L; Ojeda, G; Pasquau, J; Pineda, JA; Recio, E, 2014
)
0.66
" Reasons for starting MVC plus DRV/r were: adverse effects in 38 individuals (63%), simplification in 15 (25%) and virological failure in seven (12%)."( Efficacy and safety of once-daily maraviroc plus ritonavir-boosted darunavir in pretreated HIV-infected patients in a real-life setting.
Bancalero, P; Chueca, N; García, C; Jiménez, P; Macías, J; Márquez, M; Merino, D; Muñoz, L; Ojeda, G; Pasquau, J; Pineda, JA; Recio, E, 2014
)
0.66
"Most individuals starting MVC plus DRV/r qd because of simplification or adverse effects maintained HIV suppression after 48 weeks of follow-up."( Efficacy and safety of once-daily maraviroc plus ritonavir-boosted darunavir in pretreated HIV-infected patients in a real-life setting.
Bancalero, P; Chueca, N; García, C; Jiménez, P; Macías, J; Márquez, M; Merino, D; Muñoz, L; Ojeda, G; Pasquau, J; Pineda, JA; Recio, E, 2014
)
0.66
" Both ACT regimens were safe and well tolerated."( Artemisinin-based combination therapies are efficacious and safe for treatment of uncomplicated malaria in HIV-infected Ugandan children.
Achan, J; Arinaitwe, E; Charlebois, E; Clark, TD; Dorsey, G; Havlir, D; Ikilezi, G; Kakuru, A; Kamya, MR; Muhindo, MK; Mwangwa, F; Rosenthal, PJ; Ruel, T; Tappero, JW, 2014
)
0.4
"Treatment of uncomplicated malaria with AL or DP was efficacious and safe in HIV-infected children taking ART."( Artemisinin-based combination therapies are efficacious and safe for treatment of uncomplicated malaria in HIV-infected Ugandan children.
Achan, J; Arinaitwe, E; Charlebois, E; Clark, TD; Dorsey, G; Havlir, D; Ikilezi, G; Kakuru, A; Kamya, MR; Muhindo, MK; Mwangwa, F; Rosenthal, PJ; Ruel, T; Tappero, JW, 2014
)
0.4
"Protease inhibitor (PI) monotherapy for treatment could avoid the adverse events, drug resistance, and additional costs associated with other antiretrovirals that are normally used, particularly the nucleoside analogues."( Is there a higher risk of CNS adverse events for PI monotherapy versus triple therapy? A review of results from randomized clinical trials.
Hill, A; Moecklinghoff, C; Powderly, W,
)
0.13
"There was no clear difference in the risk of central nervous system (CNS) adverse events between PI monotherapy (either DRV/r or LPV/r) and standard triple drug treatment."( Is there a higher risk of CNS adverse events for PI monotherapy versus triple therapy? A review of results from randomized clinical trials.
Hill, A; Moecklinghoff, C; Powderly, W,
)
0.13
" However, the information on CNS adverse events has not been reported using standardized definitions in the studies."( Is there a higher risk of CNS adverse events for PI monotherapy versus triple therapy? A review of results from randomized clinical trials.
Hill, A; Moecklinghoff, C; Powderly, W,
)
0.13
"There is an urgent need for new antituberculosis (anti-TB) drugs, including agents that are safe and effective with concomitant antiretrovirals (ARV) and first-line TB drugs."( Phase I safety, pharmacokinetics, and pharmacogenetics study of the antituberculosis drug PA-824 with concomitant lopinavir-ritonavir, efavirenz, or rifampin.
Allen, R; Aweeka, F; Bao, J; Cramer, Y; Dooley, KE; Haas, DW; Koletar, SL; Luetkemeyer, AF; Marzan, F; Murray, S; Park, JG; Savic, R; Sutherland, D, 2014
)
0.61
"The most frequent clinical side effect was fatigue (in 23 cases, 88."( Side effects and tolerability of post-exposure prophylaxis with zidovudine, lamivudine, and lopinavir/ritonavir: a comparative study with HIV/AIDS patients.
Cai, J; Xiao, J; Zhang, Q, 2014
)
0.62
"027) adverse events were less frequent in ATV/r arm."( Simplification to atazanavir/ritonavir monotherapy for HIV-1 treated individuals on virological suppression: 48-week efficacy and safety results.
Antinori, A; Carini, E; Castagna, A; D'Arminio Monforte, A; Di Biagio, A; Galli, L; Lazzarin, A; Montella, F; Nozza, S; Rusconi, S; Spagnuolo, V; Vinci, C, 2014
)
0.69
" Bayesian fixed-effect network meta-analysis models adjusting for the type of nucleoside reverse transcriptase inhibitor backbone (tenofovir disoproxil fumarate/emtricitabine [TDF/FTC] or abacavir/lamivudine [ABC/3TC]) were used to evaluate week 48 efficacy (HIV-RNA suppression to <50 copies/mL and change in CD4+ cells/µL) and safety (lipid changes, adverse events, and discontinuations due to adverse events) of DTG relative to all other treatments."( 48-week efficacy and safety of dolutegravir relative to commonly used third agents in treatment-naive HIV-1-infected patients: a systematic review and network meta-analysis.
Camejo, RR; Cuffe, R; Gilchrist, KA; Lim, JW; Nichols, G; Patel, DA; Pulgar, S; Snedecor, SJ; Stephens, J; Sudharshan, L; Tang, WY, 2014
)
0.4
" Dolutegravir had better or equivalent changes in total cholesterol, LDL, triglycerides, and lower odds of adverse events and discontinuation due to adverse events compared to all treatments."( 48-week efficacy and safety of dolutegravir relative to commonly used third agents in treatment-naive HIV-1-infected patients: a systematic review and network meta-analysis.
Camejo, RR; Cuffe, R; Gilchrist, KA; Lim, JW; Nichols, G; Patel, DA; Pulgar, S; Snedecor, SJ; Stephens, J; Sudharshan, L; Tang, WY, 2014
)
0.4
" Overall, 12 (31%) patients stopped dual therapy: 7 patients because of adverse events, mostly clinical myositis (n = 3)."( Efficacy and safety of switching to raltegravir plus atazanavir dual therapy in pretreated HIV-1-infected patients over 144 weeks: a cohort study.
Batard, ML; Bernard-Henry, C; Cheneau, C; De Mautort, E; Fafi-Kremer, S; Gantner, P; Koeppel, C; Muret, P; Partisani, M; Priester, M; Rey, D; Sueur, C, 2014
)
0.4
" The Division of AIDS 2004, clarification 2009, table for grading severity of adverse events was used to classify drug toxicities."( Pharmacokinetics and safety of rifabutin in young HIV-infected children receiving rifabutin and lopinavir/ritonavir.
Gous, H; Kellermann, T; Kindra, G; McIlleron, H; Moultrie, H; Sawry, S; Van Rie, A; Wiesner, L, 2015
)
0.63
" It remains unclear whether a safe and effective rifabutin dose exists for treatment of TB in children receiving lopinavir/ritonavir."( Pharmacokinetics and safety of rifabutin in young HIV-infected children receiving rifabutin and lopinavir/ritonavir.
Gous, H; Kellermann, T; Kindra, G; McIlleron, H; Moultrie, H; Sawry, S; Van Rie, A; Wiesner, L, 2015
)
0.84
" Eleven patients (92%) reported ≥1 adverse event (AE), considered in 2 patients to be at least possibly related to darunavir (gastrointestinal-related events and dizziness)."( Efficacy and safety of darunavir/ritonavir at 48 weeks in treatment-naïve, HIV-1-infected adolescents: results from a phase 2 open-label trial (DIONE).
Blanche, S; Flynn, P; Giaquinto, C; Kakuda, TN; Komar, S; Lathouwers, E; Noguera-Julian, A; Opsomer, M; Van de Casteele, T; Welch, S, 2014
)
0.68
" Grade 1 or 2 gastrointestinal adverse events were higher among women on lopinavir/ritonavir versus efavirenz."( Efficacy and safety of lopinavir/ritonavir versus efavirenz-based antiretroviral therapy in HIV-infected pregnant Ugandan women.
Achan, J; Ades, V; Charlebois, ED; Clark, TD; Cohan, D; Gandhi, M; Havlir, DV; Kamya, MR; Koss, CA; Luwedde, F; Mwesigwa, J; Natureeba, P; Nzarubara, B; Plenty, A; Ruel, T, 2015
)
0.92
" Tolerance was good with one case of maternal grade 3 hyperbilirubinaemia, no cases in neonates at delivery and no clinically relevant adverse event."( Pharmacokinetics, safety and efficacy of ritonavir-boosted atazanavir (300/100 mg once daily) in HIV-1-infected pregnant women.
Bourgeois-Moine, A; Bourse, P; Calvez, V; Damond, F; Descamps, D; Dommergues, M; Duro, D; Faucher, P; Ichou, H; Landman, R; Lariven, S; Lê, MP; Legac, S; Mandelbrot, L; Matheron, S; Meier, F; Mortier, E; Peytavin, G; Soulié, C; Tubiana, R; Valantin, MA, 2015
)
0.68
" Concerning safety, 10 moderate to severe adverse events were recorded in eight patients; overall seven cases of renal colic (possibly treatment related) were observed, leading to a discontinuation of treatment in two patients."( Efficacy and safety of treatment simplification to atazanavir/ritonavir + lamivudine in HIV-infected patients with virological suppression: 144 week follow-up of the AtLaS pilot study.
Borghetti, A; Cauda, R; Ciccarelli, N; Colafigli, M; D'Avino, A; De Luca, A; Di Giambenedetto, S; Fabbiani, M; Gagliardini, R; Mondi, A, 2015
)
0.66
"9%) discontinued due to adverse events (AEs); and 11 patients (19."( PRINCE-1: safety and efficacy of atazanavir powder and ritonavir liquid in HIV-1-infected antiretroviral-naïve and -experienced infants and children aged ≥3 months to <6 years.
Arce, PM; Biguenet, S; Cambilargiu, D; Correll, T; Donati, AP; Hardy, H; Lissens, J; Strehlau, R; Yang, R, 2015
)
0.66
" Adverse events included eye stye, insomnia, and pain from an infiltrated intravenous line."( Pharmacokinetics and safety of co-administered paritaprevir plus ritonavir, ombitasvir, and dasabuvir in hepatic impairment.
Awni, WM; Bernstein, BM; Ding, B; Dutta, S; Khatri, A; Lawitz, EJ; Marbury, TC; Menon, RM; Mullally, VM; Podsadecki, TJ, 2015
)
0.65
" Common adverse events included headache, asthenia, pruritus, and diarrhea."( Efficacy and Safety of Ombitasvir, Paritaprevir, and Ritonavir in an Open-Label Study of Patients With Genotype 1b Chronic Hepatitis C Virus Infection With and Without Cirrhosis.
Akarca, US; Hall, C; Hézode, C; Lawitz, E; Makara, M; Mobashery, N; Morillas, RM; Pilot-Matias, T; Preotescu, LL; Redman, R; Thuluvath, PJ; Varunok, P; Vilchez, RA, 2015
)
0.67
" We also found no significant differences between the two groups for adverse events and death."( Efficacy and safety of abacavir-containing combination antiretroviral therapy as first-line treatment of HIV infected children and adolescents: a systematic review and meta-analysis.
Adetokunboh, OO; Balogun, TA; Schoonees, A; Wiysonge, CS, 2015
)
0.42
"0% discontinued treatment due to adverse events."( Short Communication: Efficacy and Safety of Treatment Simplification to Lopinavir/Ritonavir or Darunavir/Ritonavir Monotherapy: A Randomized Clinical Trial.
Bravo, I; Cañadas, MP; Clotet, B; García-Rosado, D; Llibre, JM; Moltó, J; Paredes, R; Pérez-Álvarez, N; Santos, JR, 2016
)
0.66
"To review adverse events (AEs) uniquely associated with DAA therapy across a broad spectrum of patient populations."( Review article: safety and tolerability of direct-acting anti-viral agents in the new era of hepatitis C therapy.
Banerjee, D; Reddy, KR, 2016
)
0.43
" Principal reasons for discontinuation were adverse events (15."( Long-Term Efficacy, Tolerability, and Renal Safety of Atazanavir/Ritonavir-based Antiretroviral Therapy in a Cohort of Treatment-Naïve Patients with HIV-1 Infection: the REMAIN Study.
Antela, A; Jiménez-Expósito, MJ; Knechten, H; Kuhlmann, B; Rocha-Pereira, N; Santos, J; Teófilo, E, 2016
)
0.67
" Drug-related adverse events leading to discontinuation were 3 (6%) in the atazanavir/ritonavir monotherapy arm and 11 (21."( Atazanavir/ritonavir monotherapy: 96 week efficacy, safety and bone mineral density from the MODAt randomized trial.
Antinori, A; Bigoloni, A; Borderi, M; Caramatti, G; Castagna, A; D'Arminio Monforte, A; Di Biagio, A; Di Giambenedetto, S; Galli, L; Gibellini, D; Guaraldi, G; Lazzarin, A; Montella, F; Rusconi, S; Spagnuolo, V, 2016
)
1.05
" In the atazanavir/ritonavir monotherapy arm, reintroduction of nucleosides, as needed, was always effective with no new resistance mutation; monotherapy was also associated with a lower incidence of adverse events and improvement in femur BMD."( Atazanavir/ritonavir monotherapy: 96 week efficacy, safety and bone mineral density from the MODAt randomized trial.
Antinori, A; Bigoloni, A; Borderi, M; Caramatti, G; Castagna, A; D'Arminio Monforte, A; Di Biagio, A; Di Giambenedetto, S; Galli, L; Gibellini, D; Guaraldi, G; Lazzarin, A; Montella, F; Rusconi, S; Spagnuolo, V, 2016
)
1.15
" Regarding safety, adverse events and serious adverse events were more frequently reported in patients taking concomitant ARAs, though baseline population differences may have played a role."( Safety and Efficacy of Ombitasvir/Paritaprevir/Ritonavir Plus Dasabuvir With or Without Ribavirin in HCV-Infected Patients Taking Concomitant Acid-Reducing Agents.
Asselah, T; Bennett, M; Forns, X; Liu, L; Moller, J; Pedrosa, M; Planas Vila, R; Reau, N; Rustgi, V; Shiffman, ML, 2016
)
0.69
" Adverse events occurred in 23 and 18 patients, respectively."( Efficacy and safety of once-daily ritonavir-boosted atazanavir or darunavir in combination with a dual nucleos(t)ide analogue backbone in HIV-1-infected combined ART (cART)-naive patients with severe immunosuppression: a 48 week, non-comparative, randomiz
Assoumou, L; Benalycherif, A; Cabié, A; Costagliola, D; Duvivier, C; Girard, PM; Joly, V; Lambert-Niclot, S; Landman, R; Marcelin, AG; Peytavin, G; Pialoux, G; Samri, A; Slama, L; Valin, N, 2016
)
0.71
" To investigate safety, we compared adverse events (AE) among infants exposed to different maternal cART regimens."( Maternal Lopinavir/Ritonavir Is Associated with Fewer Adverse Events in Infants than Nelfinavir or Atazanavir.
Barr, E; Davies, J; Forster, JE; Kinzie, K; Levin, MJ; McFarland, EJ; Pappas, J; Paul, S; Smith, C; Weinberg, A, 2016
)
0.76
" DRV/r was well tolerated, with few discontinuations due to study-emergent nonfatal adverse events (3."( Effectiveness, durability, and safety of darunavir/ritonavir in HIV-1-infected patients in routine clinical practice in Italy: a postauthorization noninterventional study.
Airoldi, G; Antinori, A; Bini, T; Castagna, A; Colella, E; Gianotti, N; Mancusi, D; Meraviglia, P; Monforte, Ad; Mussini, C; Rusconi, S; Termini, R, 2016
)
0.69
" The frequency of adverse events was similar in the two arms; however, one patient in the monotherapy arm was hospitalized with HIV encephalitis and elevated cerebrospinal fluid HIV-1 RNA."( Week 96 efficacy and safety of darunavir/ritonavir monotherapy vs. darunavir/ritonavir with two nucleoside reverse transcriptase inhibitors in the PROTEA trial.
Antinori, A; Arribas, JR; Bicer, C; Girard, PM; Hadacek, B; Moecklinghoff, C; Netzle-Sveine, B; Ripamonti, D, 2017
)
0.72
"Many adverse drug reactions are caused by the cytochrome P450 (CYP)-dependent activation of drugs into reactive metabolites."( Development of a cell viability assay to assess drug metabolite structure-toxicity relationships.
Jones, LH; Nadanaciva, S; Rana, P; Will, Y, 2016
)
0.43
" Safety outcomes were presented by the incidence of adverse events."( Effectiveness and Safety of Ombitasvir-Paritaprevir/Ritonavir and Dasabuvir With or Without Ribavirin for HCV Genotype 1 Infection for 12 Weeks Under Routine Clinical Practice.
Chamorro-de-Vega, E; Collado Borrell, R; Escudero-Vilaplana, V; Fernandez-Llamazares, CM; Gimenez-Manzorro, A; Herranz, A; Ibañez-Garcia, S; Lallana Sainz, E; Lobato Matilla, E; Lorenzo-Pinto, A; Manrique-Rodriguez, S; Marzal-Alfaro, M; Ribed, A; Rodriguez-Gonzalez, CG; Romero Jimenez, RM; Sanjurjo, M; Sarobe Gonzalez, C, 2016
)
0.68
" Adverse events occurred in 91."( Effectiveness and Safety of Ombitasvir-Paritaprevir/Ritonavir and Dasabuvir With or Without Ribavirin for HCV Genotype 1 Infection for 12 Weeks Under Routine Clinical Practice.
Chamorro-de-Vega, E; Collado Borrell, R; Escudero-Vilaplana, V; Fernandez-Llamazares, CM; Gimenez-Manzorro, A; Herranz, A; Ibañez-Garcia, S; Lallana Sainz, E; Lobato Matilla, E; Lorenzo-Pinto, A; Manrique-Rodriguez, S; Marzal-Alfaro, M; Ribed, A; Rodriguez-Gonzalez, CG; Romero Jimenez, RM; Sanjurjo, M; Sarobe Gonzalez, C, 2016
)
0.68
" Adverse events occurred in 151 (72."( Real-world effectiveness and safety of ombitasvir/paritaprevir/ritonavir ± dasabuvir ± ribavirin in hepatitis C: AMBER study.
Berak, H; Bialkowska, J; Bolewska, B; Fleischer-Stępniewska, K; Flisiak, R; Garlicki, A; Halota, W; Horban, A; Jabłkowski, M; Janczewska, E; Jaroszewicz, J; Karpińska, E; Karwowska, K; Knysz, B; Kryczka, W; Lucejko, M; Madej, G; Mozer-Lisewska, I; Nazzal, K; Piekarska, A; Pisula, A; Rostkowska, K; Simon, K; Tomasiewicz, K; Tronina, O; Tudrujek, M; Wawrzynowicz-Syczewska, M; Wiercińska-Drapało, A; Zarębska-Michaluk, D, 2016
)
0.67
" Before week 96, dual therapy was discontinued in 44 patients (16%) because of various adverse events, with no difference between the two groups."( Switch to Ritonavir-Boosted versus Unboosted Atazanavir plus Raltegravir Dual-Drug Therapy Leads to Similar Efficacy and Safety Outcomes in Clinical Practice.
Bani-Sadr, F; Gantner, P; Garraffo, R; Jovelin, T; Pugliese, P; Rey, D; Roger, PM; Treger, M, 2016
)
0.84
"Emerging RAL-resistance and discontinuations for adverse events resulted in moderate efficacy rates of ATV and RAL dual therapy in heavily pretreated patients."( Switch to Ritonavir-Boosted versus Unboosted Atazanavir plus Raltegravir Dual-Drug Therapy Leads to Similar Efficacy and Safety Outcomes in Clinical Practice.
Bani-Sadr, F; Gantner, P; Garraffo, R; Jovelin, T; Pugliese, P; Rey, D; Roger, PM; Treger, M, 2016
)
0.84
"Paritaprevir/ritonavir, ombitasvir, and dasabuvir with or without ribavirin is effective and safe in patients with genotype 1 HCV infection in real-life clinical setting in Hong Kong."( Real-life efficacy and safety of paritaprevir/ritonavir, ombitasvir, and dasabuvir in chronic hepatitis C patients in Hong Kong.
But, DY; Chan, CK; Chan, HL; Chan, KH; Fung, J; Hui, YT; Lai, CL; Lai, MS; Lam, YS; Lao, WC; Li, C; Lui, GC; Seto, WK; Tsang, OT; Wong, GL; Wong, VW; Yuen, MF, 2017
)
1.08
" No deaths, serious adverse events (AEs), Grade 4 AEs or AEs leading to treatment discontinuation occurred."( Efficacy, safety and pharmacokinetics of simeprevir and TMC647055/ritonavir with or without ribavirin and JNJ-56914845 in HCV genotype 1 infection.
Arastéh, K; Bourgeois, S; Buggisch, P; Francque, S; Hoeben, E; Horsmans, Y; Jacquemyn, B; Kakuda, TN; Luo, D; Moreno, C; Nevens, F; Orlent, H; Schattenberg, JM; Van Remoortere, P; Van Vlierberghe, H; Vandebosch, A; Verloes, R; Vijgen, L, 2017
)
0.69
" Details of serious adverse events (SAEs) were recorded."( Effectiveness, safety and clinical outcomes of direct-acting antiviral therapy in HCV genotype 1 infection: Results from a Spanish real-world cohort.
Albillos, A; Ampuero, J; Arenas, J; Bañares, R; Calleja, JL; Crespo, J; Diago, M; Fernandez, I; García-Eliz, M; García-Samaniego, J; Gea, F; Jorquera, F; Lens, S; Llaneras, J; Llerena, S; Mariño, Z; Morillas, RM; Muñoz, R; Navascues, CA; Pascasio, JM; Perelló, C; Rincón, D; Rodriguez, CF; Ruiz-Antorán, B; Sacristán, B; Serra, MA; Simon, MA; Torras, X; Turnes, J, 2017
)
0.46
" There were no deaths, adverse events leading to discontinuation, or serious adverse events."( Antiviral Activity, Safety, and Exposure-Response Relationships of GSK3532795, a Second-Generation Human Immunodeficiency Virus Type 1 Maturation Inhibitor, Administered as Monotherapy or in Combination With Atazanavir With or Without Ritonavir in a Phase
Boffito, M; Dicker, IB; Grasela, D; Hüser, A; Hwang, C; Keicher, C; Krystal, M; Lataillade, M; Ravindran, P; Ray, N; Schürmann, D; Sevinsky, H; Sobotha, C; Xiao, H, 2017
)
0.64
" Other correlates with adverse events of clinical importance included concomitant ribavirin treatment, sex, race, and presence of cirrhosis, consistent with previous observations."( Exposure-Safety Response Relationship for Ombitasvir, Paritaprevir/Ritonavir, Dasabuvir, and Ribavirin in Patients with Chronic Hepatitis C Virus Genotype 1 Infection: Analysis of Data from Five Phase II and Six Phase III Studies.
Awni, W; DaSilva-Tillmann, B; Dutta, S; Lin, CW; Liu, W; Menon, R; Podsadecki, T; Shulman, N, 2017
)
0.69
" Clinical and laboratory adverse events (AEs) were recorded from baseline to FU12."( Real-World Safety and Efficacy of Ombitasvir/Paritaprevir/Ritonavir/+Dasabuvir±Ribavirin (OBV/PTV/r/+DSV±RBV) Therapy in Recurrent Hepatitis C Virus (HCV) Genotype 1 Infection Post-Liver Transplant: AMBER-CEE Study.
Bolewska, B; Buivydiene, A; Durlik, M; Flisiak, R; Jabłkowski, M; Jakutiene, J; Karpińska, E; Karwowska, KM; Katzarov, K; Kupcinskas, L; Pisula, A; Rostkowska, K; Simonova, M; Tolmane, I; Tronina, O; Wawrzynowicz-Syczewska, M, 2017
)
0.7
" The aim of this study was to compare the efficacy and safety of PrOD-based therapy in hepatitis C genotype 1 patients with and without cirrhosis, and to explore pre-treatment factors predictive of sustained viral response (SVR) and serious adverse events (SAEs) on treatment."( Real-world efficacy and safety of ritonavir-boosted paritaprevir, ombitasvir, dasabuvir ± ribavirin for hepatitis C genotype 1 - final results of the REV1TAL study.
Bollipo, S; Cheng, W; Chivers, S; Dore, G; Fragomeli, V; Galhenage, S; Gazzola, A; George, J; Gow, P; Iser, D; Jones, T; Levy, M; Lubel, J; MacQuillan, G; Mitchell, JL; Nazareth, S; Pianko, S; Roberts, SK; Sasadeusz, J; Strasser, S; Stuart, KA; Thompson, A; Tse, E; Wade, A; Weltman, M; Wigg, A; Zekry, A, 2017
)
0.73
" Baseline demographic, clinical and laboratory information, on-treatment biochemical, virological and haematological indices and details on serious adverse events were collected locally."( Real-world efficacy and safety of ritonavir-boosted paritaprevir, ombitasvir, dasabuvir ± ribavirin for hepatitis C genotype 1 - final results of the REV1TAL study.
Bollipo, S; Cheng, W; Chivers, S; Dore, G; Fragomeli, V; Galhenage, S; Gazzola, A; George, J; Gow, P; Iser, D; Jones, T; Levy, M; Lubel, J; MacQuillan, G; Mitchell, JL; Nazareth, S; Pianko, S; Roberts, SK; Sasadeusz, J; Strasser, S; Stuart, KA; Thompson, A; Tse, E; Wade, A; Weltman, M; Wigg, A; Zekry, A, 2017
)
0.73
" The number and percentage of patients with treatment-emergent adverse events (TEAEs), serious TEAEs, and TEAEs consistent with hepatic decompensation were reported."( Safety of the 2D/3D direct-acting antiviral regimen in HCV-induced Child-Pugh A cirrhosis - A pooled analysis.
Cohen, DE; Cohen, E; Feld, JJ; Foster, GR; Fried, MW; Larsen, L; Mobashery, N; Nelson, DR; Poordad, F; Tatsch, F; Wedemeyer, H, 2017
)
0.46
" Data were analyzed to assess the on-treatment and off-therapy HCV viral load and on-treatment adverse events."( Real-world effectiveness and safety of paritaprevir/ritonavir, ombitasvir, and dasabuvir with or without ribavirin for patients with chronic hepatitis C virus genotype 1b infection in Taiwan.
Chen, DS; Chen, PJ; Hong, CM; Kao, JH; Liu, CH; Liu, CJ; Su, TH; Tseng, TC; Yang, HC, 2018
)
0.73
" Outcomes of interest were viral suppression, mortality, AIDS-defining illnesses or WHO stage 3-4 disease, discontinuations, discontinuations due to adverse events, and serious adverse events."( Comparative efficacy and safety of second-line antiretroviral therapy for treatment of HIV/AIDS: a systematic review and network meta-analysis.
Ayers, D; Calmy, A; Chan, K; Cooper, DA; Doherty, M; Ford, N; Kanters, S; Mills, EJ; Nsanzimana, S; Paton, NI; Popoff, E; Socias, ME; Vitoria, M; Wiens, MO, 2017
)
0.46
"02), while it increased the risk of serious adverse events (p = 0."( Safety and Efficacy of Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir with or without Ribavirin for Treatment of Hepatitis C Virus Genotype 1: A Systematic Review and Meta-analysis.
Abdel-Daim, MM; Abushouk, AI; Ahmed, H; Loutfy, SA; Menshawy, A; Mohamed, A; Negida, A, 2017
)
0.71
" No patients from the G/P Arm prematurely discontinued the study drug or experienced a treatment-emergent serious adverse event (TESAE)."( Efficacy and safety of glecaprevir/pibrentasvir in Japanese patients with chronic genotype 1 hepatitis C virus infection with and without cirrhosis.
Alves, K; Atarashi, T; Burroughs, M; Chayama, K; Eguchi, Y; Karino, Y; Kato, K; Kawakami, Y; Krishnan, P; Kumada, H; Naganuma, A; Oberoi, RK; Pilot-Matias, TJ; Pugatch, DL; Redman, R; Sato, K; Seike, M; Suzuki, F; Takei, Y; Watanabe, T; Xie, W; Yoshiji, H, 2018
)
0.48
" Through 48 weeks, the most common adverse events were upper respiratory tract infections (33."( Safety and Efficacy of Atazanavir Powder and Ritonavir in HIV-1-Infected Infants and Children From 3 Months to <11 Years of Age: The PRINCE-2 Study.
Cambilargiu, D; Correll, TA; Cotton, MF; Gonzalez-Tome, MI; Klauck, I; Liberty, A; Lissens, J; Pikora, C; Torres-Escobar, I; Zaru, L, 2018
)
0.74
" Safety outcomes were based on the incidence of adverse events."( Twelve weeks of ombitasvir/paritaprevir/r and dasabuvir without ribavirin is effective and safe in the treatment of patients with HCV genotype 1b infection and compensated cirrhosis: results from a real-world cohort study.
Chamorro-de-Vega, E; De Lorenzo-Pinto, A; Escudero-Vilaplana, V; Gimenez-Manzorro, A; Herranz-Alonso, A; Iglesias-Peinado, I; Rodriguez-Gonzalez, CG; Sanjurjo Saez, M, 2018
)
0.48
" Adverse events were recorded in 78."( Twelve weeks of ombitasvir/paritaprevir/r and dasabuvir without ribavirin is effective and safe in the treatment of patients with HCV genotype 1b infection and compensated cirrhosis: results from a real-world cohort study.
Chamorro-de-Vega, E; De Lorenzo-Pinto, A; Escudero-Vilaplana, V; Gimenez-Manzorro, A; Herranz-Alonso, A; Iglesias-Peinado, I; Rodriguez-Gonzalez, CG; Sanjurjo Saez, M, 2018
)
0.48
"The simplified regimen of OBV/PTV/r+DSV administered for 12 weeks is effective and safe in patients with chronic HCV genotype 1b infection and compensated cirrhosis."( Twelve weeks of ombitasvir/paritaprevir/r and dasabuvir without ribavirin is effective and safe in the treatment of patients with HCV genotype 1b infection and compensated cirrhosis: results from a real-world cohort study.
Chamorro-de-Vega, E; De Lorenzo-Pinto, A; Escudero-Vilaplana, V; Gimenez-Manzorro, A; Herranz-Alonso, A; Iglesias-Peinado, I; Rodriguez-Gonzalez, CG; Sanjurjo Saez, M, 2018
)
0.48
" Direct-acting antiviral (DAA) drug regimens are safe and highly effective, allowing administration of treatment also in elderly."( Efficacy and safety of paritaprevir/ritonavir, ombitasvir, and dasabuvir with ribavirin for the treatment of HCV genotype 1b compensated cirrhosis in patients aged 70 years or older.
Bataga, S; Brisc, C; Caruntu, FA; Chiriac, S; Cijevschi Prelipcean, C; Curescu, M; Gheorghe, L; Girleanu, I; Goldis, A; Iacob, S; Miftode, E; Mihai, C; Preda, C; Rogoveanu, I; Singeap, AM; Sporea, I; Stanciu, C; Stefanescu, G; Trifan, A, 2017
)
0.73
" Our aim was to analyze the characteristics associated with the presence of adverse events in patients receiving this antiviral regimen, with ribavirin in cirrhotic patients."( Safety assessment in Child A cirrhotic patients treated with Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir with Ribavirin.
Calina, OC; Hristea, A; Jipa, RE; Manea, ED; Olariu, C; Stefan, I,
)
0.36
" We recorded 201 adverse events in 98 (71."( Safety assessment in Child A cirrhotic patients treated with Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir with Ribavirin.
Calina, OC; Hristea, A; Jipa, RE; Manea, ED; Olariu, C; Stefan, I,
)
0.36
"We found a high number of adverse events, but most of them were mild or moderate and only one quarter of them required medical intervention."( Safety assessment in Child A cirrhotic patients treated with Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir with Ribavirin.
Calina, OC; Hristea, A; Jipa, RE; Manea, ED; Olariu, C; Stefan, I,
)
0.36
" Secondary endpoints included treatment adherence, patient satisfaction, incidence of adverse events and differences in plasma HIV-1 RNA viral load (VL) and CD4 cell counts."( High quality of life, treatment tolerability, safety and efficacy in HIV patients switching from triple therapy to lopinavir/ritonavir monotherapy: A randomized clinical trial.
Aguirrebengoa, K; de Jesus, SE; de Los Santos, I; Fariñas, MC; Flores, J; Galindo, MJ; García-Mercé, I; García-Vallecillos, C; Hernández-Quero, J; Hidalgo-Tenorio, C; Imaz, A; Lozano, F; Montes, ML; Orihuela, F; Pasquau, J; Ríos-Villegas, MJ; Romero-Palacios, A; Sanjoaquín, I; Vázquez, P; Vergas, J, 2018
)
0.69
" There were also no differences in the incidence and severity of adverse events, even though 22."( High quality of life, treatment tolerability, safety and efficacy in HIV patients switching from triple therapy to lopinavir/ritonavir monotherapy: A randomized clinical trial.
Aguirrebengoa, K; de Jesus, SE; de Los Santos, I; Fariñas, MC; Flores, J; Galindo, MJ; García-Mercé, I; García-Vallecillos, C; Hernández-Quero, J; Hidalgo-Tenorio, C; Imaz, A; Lozano, F; Montes, ML; Orihuela, F; Pasquau, J; Ríos-Villegas, MJ; Romero-Palacios, A; Sanjoaquín, I; Vázquez, P; Vergas, J, 2018
)
0.69
"Patients who suffered any adverse event (AE) were 74/240 (30."( Safety and efficacy of ombitasvir/paritaprevir/ritonavir/dasabuvir plus ribavirin in patients over 65 years with HCV genotype 1 cirrhosis.
Aghemo, A; Ascione, A; Bruno, S; Craxì, A; De Luca, M; Fontanella, L; Gasbarrini, A; Giannini, EG; Izzi, A; Marzioni, M; Melazzini, M; Messina, V; Montilla, S; Orlandini, A; Petta, S; Puoti, M; Sangiovanni, V; Trotta, MP; Villa, E; Zignego, AL, 2018
)
0.74
"Our findings suggest that OBV/PTV/r + DSV + RBV is safe and effective in real-life use in patients with compensated cirrhosis, HCV-GT1 infection, and age over 65."( Safety and efficacy of ombitasvir/paritaprevir/ritonavir/dasabuvir plus ribavirin in patients over 65 years with HCV genotype 1 cirrhosis.
Aghemo, A; Ascione, A; Bruno, S; Craxì, A; De Luca, M; Fontanella, L; Gasbarrini, A; Giannini, EG; Izzi, A; Marzioni, M; Melazzini, M; Messina, V; Montilla, S; Orlandini, A; Petta, S; Puoti, M; Sangiovanni, V; Trotta, MP; Villa, E; Zignego, AL, 2018
)
0.74
" GSK2838232 administration (up to 250 mg) to 124 subjects across four studies resulted in few mild adverse events (AEs) with similar frequencies to placebo."( The safety, tolerability, and pharmacokinetic profile of GSK2838232, a novel 2nd generation HIV maturation inhibitor, as assessed in healthy subjects.
Baldwin, S; Burke, M; Davies, M; Gould, E; Jeffrey, J; Jewell, RC; Johns, BA; Johnson, M; Lou, Y; Peppercorn, A; Tenorio, AR; Xu, J, 2018
)
0.48
" The other two patients reported only grade 1 adverse effects."( Concomitant use of sorafenib with ombitasvir/paritaprevir/ritonavir and dasabuvir: Effectiveness and safety in clinical practice.
Giménez-Manzorro, A; Herranz-Alonso, A; Matilla-Peña, A; Revuelta-Herrero, JL; Sanjurjo-Sáez, M, 2018
)
0.73
"The concurrent use of OBV/PTV/r+DSV with sorafenib was considered safe and effective."( Concomitant use of sorafenib with ombitasvir/paritaprevir/ritonavir and dasabuvir: Effectiveness and safety in clinical practice.
Giménez-Manzorro, A; Herranz-Alonso, A; Matilla-Peña, A; Revuelta-Herrero, JL; Sanjurjo-Sáez, M, 2018
)
0.73
" However, information about the rate of adverse events (AEs) across different DAA regimens is limited."( Outcome and adverse events in patients with chronic hepatitis C treated with direct-acting antivirals: a clinical randomized study.
Andersen, ES; Bukh, J; Christensen, PB; Fahnøe, U; Gerstoft, J; Kjær, MS; Laursen, AL; Mössner, B; Pedersen, MS; Røge, BT; Schønning, K; Sølund, C; Weis, N, 2018
)
0.48
"BZF961 was safe and well tolerated in the patients studied with no serious adverse events."( The Safety and Antiviral Activity of BZF961 with or without Ritonavir in Patients Infected with Hepatitis C Virus: A Randomized, Multicenter Trial.
Barkan, DT; Barve, A; Bidair, M; Bodendorf, U; Bracken, K; Canino, E; Chen, D; Colvin, RA; Dabovic, K; Heimbach, T; Ison, M; Jones, CL; Jones, CT; Kovacs, SJ; Lakshman, JP; Lawitz, E; Li, B; Magnusson, B; Marbury, T; Raman, P; Steiner-Swiat, R; Thohan, S; Wong, KA; Zhong, W, 2018
)
0.72
" Among various adverse effects of PIs, hepatotoxicity is a very common adverse reaction of RIT which is concentration dependent."( Formononetin and biochanin A protects against ritonavir induced hepatotoxicity via modulation of NfκB/pAkt signaling molecules.
Agarwal, NK; Azmi, L; Chaturvedi, S; Malik, MY; Naseem, Z; Rao, C; Shukla, I, 2018
)
0.74
"Five groups of animals were subjected to treatment as control, toxic control (RIT), third group (RIT + FMN), fourth group (RIT + BCA), the fifth group (RIT + FMN + BCA) and sixth group (FMN + BCA) for 14 days."( Formononetin and biochanin A protects against ritonavir induced hepatotoxicity via modulation of NfκB/pAkt signaling molecules.
Agarwal, NK; Azmi, L; Chaturvedi, S; Malik, MY; Naseem, Z; Rao, C; Shukla, I, 2018
)
0.74
" Sustained virologic response at post-treatment Week 12 (SVR12), adverse events (AEs) and comedication management were assessed for patients initiating treatment before 1 June 2017."( Real-world safety and effectiveness of ombitasvir/paritaprevir/ritonavir ± dasabuvir ± ribavirin in hepatitis C virus genotype 1- and 4-infected patients with diverse comorbidities and comedications: A pooled analysis of post-marketing observational studi
Back, D; Bondin, M; Bourgeois, S; Buggisch, P; Charafeddine, M; Crown, E; Curescu, M; Dorr, P; Ferenci, P; Flisiak, R; Kleine, H; Larrey, D; Marra, F; Norris, S, 2019
)
0.75
" However, ribavirin is associated with adverse events that can limit its use."( Efficacy and safety of ombitasvir/paritaprevir/ritonavir and dasabuvir with low-dose ribavirin in patients with chronic hepatitis C virus genotype 1a infection without cirrhosis.
Bank, L; Bernstein, D; Epstein, M; Krishnan, P; Lucey, MR; Martinez, M; Nelson, DR; Pockros, PJ; Polepally, AR; Poordad, F; Ravendhran, N; Reindollar, R; Sedghi, S; Trinh, R; Unnebrink, K, 2019
)
0.77
" In summary, the current work demonstrated the essential roles of human PXR and CYP3A4 in RTV hepatotoxicity, which can be applied to guide the safe use of RTV-containing regimens in the clinic."( Pregnane X receptor activation potentiates ritonavir hepatotoxicity.
Gonzalez, FJ; Lu, J; Ma, X; McMahon, D; Shehu, AI; Wang, P; Wang, Y; Xie, W; Yang, D; Zhu, J, 2019
)
0.78
"Ritonavir retinal toxicity seems to be an uncommon adverse event that can cause decreased visual function."( Case Report: Retinal Toxicity Secondary to Ritonavir.
Jones, HN; Louie, AK, 2019
)
2.22
" The patients were evaluated in respect of demographic, clinical and virological data, sustained virologic response (SVR) and adverse events."( [EFFICACY AND SAFETY OF OMBITASVIR/PARITAPREVIR/RITONAVIR AND DASABUVIR IN PATIENTS WITH HCV 1B GENOTYPE INFECTION: REAL WORLD DATA].
Skorokhodova, N; Tsarova, O; Zhyvytsia, D, 2019
)
0.77
" Adverse events (grade 1-4) were more common at baseline (27%) than during rifabutin treatment (15%) (P = 0."( Safety and efficacy of rifabutin among HIV/TB-coinfected children on lopinavir/ritonavir-based ART.
Akanmu, S; Brown, B; Chang, C; Darin, KM; David, N; Kanki, PJ; Ogunbosi, B; Okonkwo, P; Oladokun, R; Olaitan, O; Rawizza, HE; Scarsi, KK, 2019
)
0.74
"With clinical and laboratory monitoring, our data suggest that rifabutin is a safe option for TB therapy among children on PI-based ART."( Safety and efficacy of rifabutin among HIV/TB-coinfected children on lopinavir/ritonavir-based ART.
Akanmu, S; Brown, B; Chang, C; Darin, KM; David, N; Kanki, PJ; Ogunbosi, B; Okonkwo, P; Oladokun, R; Olaitan, O; Rawizza, HE; Scarsi, KK, 2019
)
0.74
" We collected data on on-treatment adverse events (AEs), severe AEs, and laboratory abnormalities."( Efficacy and safety of direct-acting antiviral agents for HCV in mild-to-moderate chronic kidney disease.
Aoufi-Rabih, S; Dixit, V; Fabrizi, F; Garcia-Agudo, R; Mendizabal, M; Ridruejo, E; Silva, M,
)
0.13
"7%): 4 due to severe adverse events (including 3 deaths) and 1 patient´s decision."( Effectiveness, safety/tolerability of OBV/PTV/r ± DSV in patients with HCV genotype 1 or 4 with/without HIV-1 co-infection, chronic kidney disease (CKD) stage IIIb-V and dialysis in Spanish clinical practice - Vie-KinD study.
Ahumada, A; Aldamiz-Echevarría, T; Baliellas, C; Barril, G; Benlloch, S; Bonet, L; Carmona, I; Castaño, MA; Castro, Á; de Álvaro, C; de Los Santos, I; Delgado, M; Devesa-Medina, MJ; García-Buey, L; García-Samaniego, J; Gea-Rodríguez, F; González-Parra, E; Gutiérrez, ML; Jiménez-Pérez, M; Laguno, M; Londoño, MC; Losa, JE; Mallolas, J; Manzanares, A; Martín-Granizo, I; Montero-Alonso, M; Montes, ML; Morán-Sánchez, S; Morano, L; Muñoz-Gómez, R; Navascués, CA; Polo-Lorduy, B; Riveiro-Barciela, M; Rivero, A; Roget, M; Serra, MÁ, 2019
)
0.51
" Direct-acting antiviral therapy is efficacious and safe even in patients with advanced liver disease and/ or previous virological failure; Model-for-End-Liver-Disease <10 and alanine aminotransferase reduction during therapy were found to be reliable predicting markers of sustained-virological-response."( Efficacy, Safety, and Predictors of Direct-acting antivirals in Hepatitis C Virus Patients with Heterogeneous Liver Diseases.
Cescon, M; De Pace, V; Galli, S; Maggi, F; Morelli, MC; Pistello, M; Ravaioli, M; Re, MC; Vero, V, 2019
)
0.51
" However, there are several reasons as to why patients may discontinue their antiretroviral therapy, with adverse events being one of the main reasons reported in the literature."( Factors associated to modification of first-line antiretroviral therapy due to adverse events in people living with HIV/AIDS.
Azevedo, LN; Miranda-Filho, DB; Montarroyos, UR; Monteiro, P; Ximenes, RAA,
)
0.13
" Rates of adverse events (AEs) in the patients was 59."( Real-world efficacy, safety, and clinical outcomes of ombitasvir/paritaprevir/ritonavir ± dasabuvir ± ribavirin combination therapy in patients with hepatitis C virus genotype 1 or 4 infection: The Turkey experience experience.
Asan, A; Aygen, B; Barut, Ş; Batırel, A; Bilgin, H; Çelen, MK; Çelik, İ; Demirtürk, N; Ersöz, G; Karakeçili, F; Kınıklı, S; Mıstık, R; Şimşek, F; Türker, N; Turkish Society Of Clinical Microbiology And Infectious Diseases, TSGFVHOT; Ural, O; Yıldız, O; Zararsız, G, 2020
)
0.79
" Adverse events (AEs) were found in less than one half of patients (45 patients, or 42."( [Efficacy and safety of narlaprevir/ritonavir and daclatasvir non interferon combination in population of Russian patients with chronic hepatitis C].
Batskikh, SN; Burnevich, EZ; Chulanov, VP; Gusev, DA; Kizlo, SN; Klimova, EA; Krasavina, EN; Mamonova, NA; Samsonov, MY; Tarkhova, EP; Yushchuk, ND; Znoyko, OO, 2019
)
0.79
" This combination was found to be safe and well - tolerated."( [Efficacy and safety of narlaprevir/ritonavir and daclatasvir non interferon combination in population of Russian patients with chronic hepatitis C].
Batskikh, SN; Burnevich, EZ; Chulanov, VP; Gusev, DA; Kizlo, SN; Klimova, EA; Krasavina, EN; Mamonova, NA; Samsonov, MY; Tarkhova, EP; Yushchuk, ND; Znoyko, OO, 2019
)
0.79
" Secondary outcomes were duration of hospital stay, length of intensive care unit stay, 28-day mortality, effect of early or late administration of IFN on mortality, adverse effects, and complications during the hospitalization."( A Randomized Clinical Trial of the Efficacy and Safety of Interferon β-1a in Treatment of Severe COVID-19.
Abbasian, L; Davoudi-Monfared, E; Hajiabdolbaghi, M; Kazemzadeh, H; Khalili, H; Rahmani, H; Salehi, M; Yekaninejad, MS, 2020
)
0.56
"3%) in the arbidol group experienced adverse events during the follow-up period."( Efficacy and Safety of Lopinavir/Ritonavir or Arbidol in Adult Patients with Mild/Moderate COVID-19: An Exploratory Randomized Controlled Trial.
Cai, W; Chen, X; Deng, X; Guan, Y; Hong, W; Hu, F; Li, F; Li, L; Li, Y; Lin, W; Liu, J; Mo, X; Peng, P; Wang, J; Wang, Y; Wen, C; Xiao, G; Xie, Z; Zhang, F; Zhang, L, 2020
)
0.84
" There were no clinically relevant toxicities nor adverse events in both control and test arms."( Effect of nevirapine, efavirenz and lopinavir/ritonavir on the therapeutic concentration and toxicity of lumefantrine in people living with HIV at Lagos University Teaching Hospital, Nigeria.
Abideen, G; Adeniji, H; Adeuja, O; Agbaje, EO; Akanmu, AS; Akinleye, MO; Akinyede, AA; Busari, AW; Hassan, OO; Ken-Owotor, C; Kogbe, S; Ogunfowokan, T; Onwujuobi, AG; Oreagba, IA; Owolabi, ET; Usman, SO, 2020
)
0.82
"To find effective and safe treatments for COVID-19, the WHO recommended to systemically evaluate experimental therapeutics in collaborative randomised clinical trials."( Protocol for the DisCoVeRy trial: multicentre, adaptive, randomised trial of the safety and efficacy of treatments for COVID-19 in hospitalised adults.
Ader, F, 2020
)
0.56
" In this context, intensive pharmacovigilance allows early detection of adverse events, and thereby infer the safety profile of the indication."( [Adverse effects of lopinavir/ritonavir in critically ill patients with COVID-19].
Carboni Bisso, I; Catanzariti, A; Las Heras, M; Vecchio, G; Zapico, V, 2020
)
0.85
" Its empirical use has been associated with multiple cardiac adverse reactions pertaining to its ancillary multi-channel blocking properties, vaguely characterized until now."( Spontaneous reported cardiotoxicity induced by lopinavir/ritonavir in COVID-19. An alleged past-resolved problem.
Drici, MD; Fresse, A; Gérard, A; Lepelley, M; Rocher, F; Romani, S; Salem, JE; Viard, D, 2021
)
0.87
" Regarding adverse effects, except occurrence of diarrhea (higher in the L/R group), safety was comparable to SOC."( Safety and efficacy of lopinavir/ritonavir combination in COVID-19: A systematic review, meta-analysis, and meta-regression analysis.
Avti, P; Bansal, S; Bhattacharyya, A; Hazarika, M; Kaur, H; Kumar, S; Mahendru, D; Medhi, B; Prajapat, M; Prakash, A; Sarma, P; Sharma, S; Shekhar, N,
)
0.41
" It was associated with high risk IFN-related adverse reactions due to reduced renal clearance of IFN."( Efficacy and safety of ombitasvir/paritaprevir/ritonavir/ribavirin in management of Egyptian chronic hepatitis C virus patients with chronic kidney disease: A real-life experience.
Abd-Elsalam, S; Abo-Amer, YE; Ahmed, R; Badawi, R; El-Abgeegy, M; Elguindy, AMA; Elkadeem, M; Elsergany, HF; Elshweikh, SA; Hawash, N; Mansour, L; Mohmed, AA; Soliman, MY; Soliman, S, 2020
)
0.82
" Secondary Endpoints: All causes mortality, Frequency of respiratory progression (defined as SPO2≤ 94% on room air or PaO2/FiO2 <300mmHg and requirement for supplemental oxygen or more advanced ventilator support), time to defervescence (in those with fever at enrolment), frequency of requirement for supplemental oxygen or non-invasive ventilation, frequency of requirement for mechanical ventilation, frequency of serious adverse events as per DAIDS table grade of severity."( Safety and efficacy of antiviral combination therapy in symptomatic patients of Covid-19 infection - a randomised controlled trial (SEV-COVID Trial): A structured summary of a study protocol for a randomized controlled trial.
Bahurupi, YA; Bandyopadhyay, A; Chikara, G; Moirangthem, B; Panda, PK; Saha, S; Singh, BC, 2020
)
0.56
"Lopinavir plasma concentrations in patients with moderate-to-severe COVID-19 were higher than expected, and they were associated with the occurrence of hepatic or gastrointestinal adverse drug reactions."( Plasma Concentrations and Safety of Lopinavir/Ritonavir in COVID-19 Patients.
Azoulay, C; Batista, R; Benaboud, S; Boujaafar, S; Canouï, E; Carlier, N; Chouchana, L; Gana, I; Kernéis, S; Legendre, P; Preta, LH; Regard, L; Terrier, B; Treluyer, JM; Zerbit, J; Zheng, Y, 2021
)
0.88
" The frequency of grade 3 to 4 adverse events was similar in the two groups; the most common adverse events were diarrhea, upper respiratory tract infections, and grade 3 to 4 increases in triglyceride concentration."( Efficacy and safety of the long-acting fusion inhibitor albuvirtide in antiretroviral-experienced adults with human immunodeficiency virus-1: interim analysis of the randomized, controlled, phase 3, non-inferiority TALENT study.
Cai, WP; Chen, JF; Chen, YY; Dai, LL; He, Y; Hu, JH; Jiang, TY; Li, LH; Liu, L; Lu, HZ; Lu, RJ; Lun, WH; Peng, HY; Shao, YM; Su, B; Sun, LJ; Sun, YT; Wang, H; Wang, M; Wang, MX; Wang, Y; Wu, H; Xia, W; Xie, D; Xing, H; Yao, C; Yu, JH; Zhang, FJ; Zhang, T; Zhao, M; Zhao, QX; Zheng, QS; Zheng, YH; Zhu, B, 2020
)
0.56
" Reportable adverse events (AEs) included AEs considered at least possibly related to treatment with DRV/r, AEs leading to discontinuation or treatment interruption, and serious AEs (SAEs)."( The DIANA Study: Continued Access to Darunavir/Ritonavir (DRV/r) and Long-Term Safety Follow-Up in HIV-1-Infected Pediatric Patients Aged 3 to < 18 Years.
Blanche, S; Chetty, P; Hufkens, V; Masenya, M; Opsomer, M; Vanveggel, S; Violari, A, 2021
)
0.88
" The secondary outcome is the incidence of serious adverse drug reactions within seven days of randomization."( The efficacy and safety of Ivermectin in patients with mild and moderate COVID-19: A structured summary of a study protocol for a randomized controlled trial.
Dadvand, H; Davoodian, P; Fathalipour, M; Ghazizadeh, S; Hassaniazad, M; Hassanipour, S; Hosseini, FS; Kahoori, S; Malektojari, A; Nikoofal-Sahlabadi, S; Nikpoor, AR; Sepandi, M, 2021
)
0.62
" Grade ≥3 adverse events were compared between cohorts."( Pharmacokinetics and Safety of Zidovudine, Lamivudine, and Lopinavir/Ritonavir in HIV-infected Children With Severe Acute Malnutrition in Sub-Saharan Africa: IMPAACT Protocol P1092.
Aweeka, F; Bradford, S; Browning, R; Coletti, A; Costello, D; Graham, B; Hughes, E; Kamthunzi, P; Kawalazira, R; Mmbaga, BT; Moye, J; Musoke, P; Nathoo, K; Norman, J; Owor, M; Purdue, L; Reding, C; Tierney, C; Whalen, ME; Wiesner, L; Ziemba, L, 2021
)
0.86
"For children with mild COVID-19, LPR is inferior to conventional treatment in reducing virus shedding time and hospitalization duration and is associated with increased adverse reactions."( Safety and efficacy of oral lopinavir/ritonavir in pediatric patients with coronavirus disease: a nationwide comparative analysis.
Chen, RJ; Cui, YX; He, YL; Li, XY; Li, ZP; Lu, JM; Ma, SL; Shang, FN; Wang, XF; Xu, H; Ye, QF; Zhai, XW; Zhang, XB; Zhou, AF, 2021
)
0.89
" The most common adverse events were fatigue (12."( Real-world efficacy and safety of Ledipasvir + Sofosbuvir and Ombitasvir/Paritaprevir/Ritonavir ± Dasabuvir combination therapies for chronic hepatitis C: A Turkish experience.
Akarca, US; Akarsu, M; Akbulut, S; Akın, M; Aladağ, M; Albayrak, B; Alkım, H; Atalay, R; Balkan, A; Balkan, Y; Çoban, M; Coşar, AM; Danış, N; Değertekin, B; Demir, M; Demircan, M; Dinçer, D; Doğanay, L; Dursun, H; Erarslan, E; Göktürk, HS; Gündüz, F; Güneş, Ş; Gürel, S; Güzelbulut, F; Harputluoğlu, M; İnci, İ; Irak, K; Kaçar, S; Kani, HT; Kartal, A; Kefeli, A; Koklu, H; Mert, A; Nuriyev, K; Özakyol, A; Özdoğan, O; Öztaşkın, S; Sen, İ; Şimşek, H; Soylu, A; Sümer, H; Temel, T; Üçbilek, E; Uğurlu, ÇB; Uyanıkoğlu, A; Vatansever, S; Yalçın, K; Yaras, S; Yıldırım, E, 2020
)
0.78
" The scientific community is studying and testing numerous compounds that can be effective and safe for treating people with covid-19."( [The praise of uncertainty: a systematic living review to evaluate the efficacy and safety of drug treatments for patients with covid-19.]
Amato, L; Cruciani, F; Davoli, M; De Crescenzo, F; Mitrova, Z; Saulle, R; Vecchi, S, 2021
)
0.62
" No differences for the risk of any adverse events are observed between convalescent plasma and remdesivir compared to standard treatment."( [The praise of uncertainty: a systematic living review to evaluate the efficacy and safety of drug treatments for patients with covid-19.]
Amato, L; Cruciani, F; Davoli, M; De Crescenzo, F; Mitrova, Z; Saulle, R; Vecchi, S, 2021
)
0.62
"08) or incidence of adverse events (P=0."( Comprehensive evaluation of the efficacy and safety of LPV/r drugs in the treatment of SARS and MERS to provide potential treatment options for COVID-19.
Li, X; Liu, J; Luo, R; Wu, D; Wu, L; Xu, P; Zheng, Y, 2021
)
0.62
" The virological cure, radiological improvement and adverse events were pooled as risk ratio (RR) with 95% CI."( Efficacy and safety of lopinavir-ritonavir in COVID-19: A systematic review of randomized controlled trials.
Barvaliya, M; Bhalla, HL; Khosla, PP; Patel, PB; Patel, TK; Saurabh, MK, 2021
)
0.9
" Virological response (VR) was measured, as well as the biochemical parameters related to treatment efficacy and adverse events at baseline and after treatment, at 4 (VR4) and 12 (VR12) weeks post-treatment."( Efficacy and Safety of Ombitasvir plus Paritaprevir, Ritonavir and Ribavirin in Non-cirrhotic Treatment-naïve and Treatment-experienced Egyptians with Chronic HCV Genotype-4 Infection.
Ahmed, M; Gomaa, AA; Mansey, AE; Rabea, HM; Wahsh, EA, 2021
)
0.87
" Methods We conducted a disproportionality analysis of US Food and Drug Administration Adverse Event Reporting System (FAERS) between 2020Q1 and 2021Q1 to evaluate the association between lopinavir-ritonavir and risk of drug-induced liver injury (or severe drug-induced liver injury) and calculated their reporting odds ratios (RORs) with 95% confidence intervals (CIs)."( Drug-induced liver injury associated with lopinavir-ritonavir in patients with COVID-19: a disproportionality analysis of U.S. food and drug administration adverse event reporting system (FAERS) data.
Barnes, EL; Kinlaw, AC; Li, X; Moon, AM; Tang, H; Wang, T; Yang, JY; Zhou, L, 2021
)
1.06
"The aim of the study is to provide an overview of Drug-drug Interactions (DDIs) and adverse effects caused by drugs used in SARS-CoV-2 infection during the first epidemic wave."( [Treatments for SARS-CoV-2 infection: A retrospective study of drug-drug interactions and safety].
Benomar, A; Clou, E; Debrix, I; Dubois, A; Fansi Ndengoue, D; Féral, A; Michot, J; Pain, JB, 2022
)
0.72
" A review of adverse events and DDI-risky drug association on medical record was conducted for each patient."( [Treatments for SARS-CoV-2 infection: A retrospective study of drug-drug interactions and safety].
Benomar, A; Clou, E; Debrix, I; Dubois, A; Fansi Ndengoue, D; Féral, A; Michot, J; Pain, JB, 2022
)
0.72
" Causality assessment by CRPV concluded that 10 (35,7 %) adverse effects were possibly related to SARS-CoV-2 drugs with only 2 (7,1 %) related to DDIs."( [Treatments for SARS-CoV-2 infection: A retrospective study of drug-drug interactions and safety].
Benomar, A; Clou, E; Debrix, I; Dubois, A; Fansi Ndengoue, D; Féral, A; Michot, J; Pain, JB, 2022
)
0.72
"Despite risk of adverse drug reactions and DDIs related to drugs used in SARS-CoV-2 infection, few iatrogenics diseases were found."( [Treatments for SARS-CoV-2 infection: A retrospective study of drug-drug interactions and safety].
Benomar, A; Clou, E; Debrix, I; Dubois, A; Fansi Ndengoue, D; Féral, A; Michot, J; Pain, JB, 2022
)
0.72
" No adverse events were related to study drugs."( Pharmacokinetics and Safety of the Abacavir/Lamivudine/Lopinavir/Ritonavir Fixed-Dose Granule Formulation (4-in-1) in Neonates: PETITE Study.
Andrieux-Meyer, I; Bekker, A; Capparelli, E; Cotton, MF; Cressey, R; Cressey, TR; du Toit, S; Groenewald, M; Kumar, M; Lallemant, M; Nielsen, J; Rabie, H; Salvadori, N; Than-In-At, K, 2022
)
0.96
" In addition to the ongoing research and development of vaccines, there is still a dire need for safe and effective drugs for the control and treatment against the SARS-CoV-2 virus infection."( Safety profile of COVID-19 drugs in a real clinical setting.
Bhardwaj, M; Chiu, MN; Sah, SP, 2022
)
0.72
"Through a literature search conducted on PubMed and Google Scholar database, various adverse events suspected to be induced by eight drugs, including dexamethasone, hydroxychloroquine, chloroquine, remdesivir, favipiravir, lopinavir/ritonavir, ivermectin, and tocilizumab, administered in COVID-19 patients in clinical practice and studies were identified in 30 case reports, 3 case series, and 10 randomized clinical trials."( Safety profile of COVID-19 drugs in a real clinical setting.
Bhardwaj, M; Chiu, MN; Sah, SP, 2022
)
0.9
"Mild, moderate, or severe adverse events of numerous repurposed and investigational drugs caused by various factors and mechanisms were observed."( Safety profile of COVID-19 drugs in a real clinical setting.
Bhardwaj, M; Chiu, MN; Sah, SP, 2022
)
0.72
" We conducted a meta-analysis to investigate the improvement in mortality or hospitalization rates and adverse events among COVID-19 patients with three new oral antivirals (including molnupiravir, fluvoxamine and Paxlovid)."( Efficacy and safety of three new oral antiviral treatment (molnupiravir, fluvoxamine and Paxlovid) for COVID-19:a meta-analysis.
Chen, C; Cheng, Y; Feng, Z; Mao, Q; Tang, J; Wang, C; Wang, M; Wen, W; Wu, Q; Zhang, X; Zhou, M; Zhou, X, 2022
)
0.72
" In addition, the three oral drugs did not increase the occurrence of adverse events, thus exhibiting good overall safety."( Efficacy and safety of three new oral antiviral treatment (molnupiravir, fluvoxamine and Paxlovid) for COVID-19:a meta-analysis.
Chen, C; Cheng, Y; Feng, Z; Mao, Q; Tang, J; Wang, C; Wang, M; Wen, W; Wu, Q; Zhang, X; Zhou, M; Zhou, X, 2022
)
0.72
"To summarise specific adverse effects of remdesivir, hydroxychloroquine and lopinavir/ritonavir in patients with COVID-19."( Adverse effects of remdesivir, hydroxychloroquine and lopinavir/ritonavir when used for COVID-19: systematic review and meta-analysis of randomised trials.
Agoritsas, T; Bartoszko, JJ; Brignardello-Petersen, R; Chu, DK; Ge, L; Izcovich, A; Khamis, AM; Kum, E; McLeod, SL; Mustafa, RA; Qasim, A; Rochwerg, B; Siemieniuk, RA; Vandvik, P; Zeraatkar, D, 2022
)
1.18
" For most interventions and outcomes the certainty of the evidence was very low to low except for gastrointestinal adverse effects from hydroxychloroquine, which was moderate certainty."( Adverse effects of remdesivir, hydroxychloroquine and lopinavir/ritonavir when used for COVID-19: systematic review and meta-analysis of randomised trials.
Agoritsas, T; Bartoszko, JJ; Brignardello-Petersen, R; Chu, DK; Ge, L; Izcovich, A; Khamis, AM; Kum, E; McLeod, SL; Mustafa, RA; Qasim, A; Rochwerg, B; Siemieniuk, RA; Vandvik, P; Zeraatkar, D, 2022
)
0.96
" ICU admission, length of stay (LOS) in hospital, in-hospital mortality, and the incidence of adverse events were also measured."( Efficacy and safety of favipiravir plus interferon-beta versus lopinavir/ritonavir plus interferon-beta in moderately ill patients with COVID-19: A randomized clinical trial.
Bazram, A; Farshidi, H; Fathalipour, M; Gharibzadeh, A; Hassaniazad, M; Khalili, E; Noormandi, A, 2022
)
0.95
" The proportions of participants with any grade 3-4 adverse event were similar between the dolutegravir (26 [11%]) and darunavir (28 [12%]) groups and between the tenofovir (22 [9%]) and zidovudine (32 [14%]) groups."( Efficacy and safety of dolutegravir or darunavir in combination with lamivudine plus either zidovudine or tenofovir for second-line treatment of HIV infection (NADIA): week 96 results from a prospective, multicentre, open-label, factorial, randomised, non
Asienzo, J; Ategeka, G; Balyegisawa, A; Borok, M; Castelnuovo, B; Hoppe, A; Kaimal, A; Kambugu, A; Kiragga, A; Kityo, C; Lugemwa, A; Mirembe, G; Mugerwa, H; Musaazi, J; Odongpiny, ELA; Paton, NI; Siika, A; Walimbwa, S, 2022
)
0.72
" In terms of safety, we monitored the development of adverse reactions, liver cytolysis, cholestasis, and hematologic disorders."( A Real-World Study to Compare the Safety and Efficacy of Paritaprevir/Ombitasvir/Ritonavir and Dasabuvir, with or without Ribavirin, in 587 Patients with Chronic Hepatitis C at the Fundeni Clinical Institute, Bucharest, Romania.
Bacinschi, X; Gales, L; Haineala, B; Iliescu, L; Mercan, A; Popescu, GC; Rodica, A; Serban, D; Toma, L; Zgura, A, 2022
)
0.95
" However, previous studies have shown that RTV has toxic effects on various cell types."( Reproductive toxicity of ritonavir in male: Insight into mouse sperm capacitation.
Bae, JW; Hwang, JM; Jung, EJ; Kwon, WS; Lee, WJ, 2022
)
1.02
" The primary endpoints here considered were any adverse events observed or reported during the treatment cycle with estimates of odds ratio (OR) and 95% confidence interval (CI), until November 6, 2021."( A systematic review and Bayesian network meta-analysis for comparative safety assessment of favipiravir interventions in hospitalized COVID-19 patients.
Chen, M; Dai, W; Yang, F; Yang, K; Zeng, J, 2022
)
0.72
" We extracted data on trials and patient characteristics, and the following primary outcomes: all-cause mortality (ACM), and treatment-emergent adverse events (TEAEs)."( Comparative efficacy and safety of pharmacological interventions for severe COVID-19 patients: An updated network meta-analysis of 48 randomized controlled trials.
Chen, J; Cheng, Q; Fang, Z; Jia, Q; Zhao, G, 2022
)
0.72
" We found that most medications were safe in treating severe COVID-19."( Comparative efficacy and safety of pharmacological interventions for severe COVID-19 patients: An updated network meta-analysis of 48 randomized controlled trials.
Chen, J; Cheng, Q; Fang, Z; Jia, Q; Zhao, G, 2022
)
0.72
"01), and adverse events (OR = 2."( Efficacy and safety of nirmatrelvir/ritonavir (Paxlovid) for COVID-19: A rapid review and meta-analysis.
Amani, B, 2023
)
1.19
" The results of the cytotoxicity assay and drug uptake study showed that PPG4ER was safe and biocompatible up to 12."( Effect of PEGylation on drug uptake, biodistribution, and tissue toxicity of efavirenz-ritonavir loaded PAMAM G4 dendrimers.
Dhobley, A; Kharwade, R; Mahajan, N; Mendhi, S; More, S; Palve, D; Warokar, A, 2023
)
1.13
" Moreover, adverse events were more frequent in the fed state, but all were mild."( Pharmacokinetics, Safety, and Bioequivalence of 2 Lopinavir/Ritonavir (200/50 mg) Tablets in Healthy Chinese Volunteers: Effect of Food on Absorption.
Bao, D; Fu, W; Hu, W; Lin, L; Liu, Y; Zhang, Q; Zhang, W; Zheng, L, 2023
)
1.15
"Molnupiravir (MOL) and nirmatrelvir/ritonavir (NIR) were recently approved for the early treatment of COVID-19, but real-life data on tolerability, safety, and adverse events (AEs) are still scarce."( Molnupiravir and Nirmatrelvir/Ritonavir: Tolerability, Safety, and Adherence in a Retrospective Cohort Study.
Bonadiman, N; Calandrino, L; Cattelan, AM; Cavinato, S; Ferrari, A; Gardin, S; Mazzitelli, M; Mengato, D; Sasset, L; Scaglione, V; Trivellato, S; Venturini, F, 2023
)
1.47
"6%) patients experienced any AEs following antivirals intake: 98/124 (79%) patients reporting adverse events presented grade 1 AEs, 23/124 (18."( Molnupiravir and Nirmatrelvir/Ritonavir: Tolerability, Safety, and Adherence in a Retrospective Cohort Study.
Bonadiman, N; Calandrino, L; Cattelan, AM; Cavinato, S; Ferrari, A; Gardin, S; Mazzitelli, M; Mengato, D; Sasset, L; Scaglione, V; Trivellato, S; Venturini, F, 2023
)
1.2
" However, a comprehensive review of the adverse event (AE) profile of the DAAs is lacking."( Evaluation of the Safety Profile of Direct-Acting Antivirals on Patients with Hepatitis C Virus: A Pharmacovigilance Study.
El-Marakby, MG; Sabri, NA; Solayman, MH, 2023
)
0.91
" In terms of safety, although the incidence of any adverse events was higher in the nirmatrelvir/ritonavir group (OR = 2."( Comparative efficacy and safety of nirmatrelvir/ritonavir and molnupiravir for COVID-19: A systematic review and meta-analysis.
Akbarzadeh, A; Amani, B; Kardanmoghadam, V; Khorramnia, S; Navidi, Z; Rajabkhah, K; Shabestan, R, 2023
)
1.38
" For adverse events (AEs) outcome, the OR was >1 and p < 0."( Efficacy and safety of paxlovid (nirmatrelvir/ritonavir) in the treatment of COVID-19: An updated meta-analysis and trial sequential analysis.
Song, T; Tang, L; Tian, H; Tian, Y; Wen, L; Xu, W; Yang, C; Zhang, X; Zhou, K, 2023
)
1.17
" Dysgeusia with a bitter or metallic taste is a common side effect of nirmatrelvir/ritonavir."( Neurological side effects and drug interactions of antiviral compounds against SARS-CoV-2.
Akhvlediani, T; Bernard-Valnet, R; Dias, SP; Eikeland, R; Pfausler, B; Sellner, J, 2023
)
1.14
" Further studies are required to better evaluate their efficacy and adverse events in patients with concomitant neurological diseases."( Neurological side effects and drug interactions of antiviral compounds against SARS-CoV-2.
Akhvlediani, T; Bernard-Valnet, R; Dias, SP; Eikeland, R; Pfausler, B; Sellner, J, 2023
)
0.91
" The secondary outcome included hospital discharging conditions, severe conversion of symptom, and adverse events."( Efficacy and safety of Huashi Baidu granule plus Nirmatrelvir-Ritonavir combination therapy in patients with high-risk factors infected with Omicron (B.1.1.529): A multi-arm single-center, open-label, randomized controlled trial.
Chen, B; Cong, J; Feng, H; Gao, Y; Li, D; Lv, J; Qv, W; Shang, B; Tao, R; Wu, H; Xv, C; Xv, Y; Yang, Y; Yu, Z; Yuan, W; Zhang, X; Zheng, Y; Zhu, X, 2023
)
1.15
" No severe adverse events occurred among combination therapy and monotherapies in the trial."( Efficacy and safety of Huashi Baidu granule plus Nirmatrelvir-Ritonavir combination therapy in patients with high-risk factors infected with Omicron (B.1.1.529): A multi-arm single-center, open-label, randomized controlled trial.
Chen, B; Cong, J; Feng, H; Gao, Y; Li, D; Lv, J; Qv, W; Shang, B; Tao, R; Wu, H; Xv, C; Xv, Y; Yang, Y; Yu, Z; Yuan, W; Zhang, X; Zheng, Y; Zhu, X, 2023
)
1.15
"For patients with severe COVID-19, HSBD exhibits similar efficacy to Paxlovid, while combination therapy is more likely to increase the curative efficacy of Omicron variant than monotherapies, with few serious adverse events."( Efficacy and safety of Huashi Baidu granule plus Nirmatrelvir-Ritonavir combination therapy in patients with high-risk factors infected with Omicron (B.1.1.529): A multi-arm single-center, open-label, randomized controlled trial.
Chen, B; Cong, J; Feng, H; Gao, Y; Li, D; Lv, J; Qv, W; Shang, B; Tao, R; Wu, H; Xv, C; Xv, Y; Yang, Y; Yu, Z; Yuan, W; Zhang, X; Zheng, Y; Zhu, X, 2023
)
1.15
"Hepatitis C virus (HCV) infection is an independent risk factor associated with adverse outcomes in patients with end-stage renal disease (ESRD)."( The efficacy and safety of direct-acting antiviral regimens for end-stage renal disease patients with HCV infection: a systematic review and network meta-analysis.
Chen, R; Wang, X; Xiao, Y; Xiong, Y; Zeng, Y; Zheng, Y, 2023
)
0.91
" Study data that contained patient characteristics, study design, treatment regimens, intention-to-treat sustained virologic response (SVR), and adverse event (AE) data per regimen were extracted into a structured electronic database and analyzed."( The efficacy and safety of direct-acting antiviral regimens for end-stage renal disease patients with HCV infection: a systematic review and network meta-analysis.
Chen, R; Wang, X; Xiao, Y; Xiong, Y; Zeng, Y; Zheng, Y, 2023
)
0.91

Pharmacokinetics

Lopinavir/ritonavir dose of 500/125 mg bid administered with efavirenz most closely approximates the pharmacokinetic exposure of lopinavIR/rit onavir 400/100 mg alone.

ExcerptReferenceRelevance
" These results indicate that ritonavir can favorably alter the pharmacokinetic profiles of other protease inhibitors."( Pharmacokinetic enhancement of inhibitors of the human immunodeficiency virus protease by coadministration with ritonavir.
Baroldi, PA; Denissen, JF; Granneman, GR; Hsu, A; Kempf, DJ; Kukulka, MJ; Kumar, G; Leonard, JM; Marsh, KC; McDonald, E; Norbeck, DW; Pizzuti, D; Plattner, JJ; Rodrigues, AD; Sun, E, 1997
)
0.8
"For subjects receiving rifabutin and placebo who completed the study (n = 11), there were small but statistically significant differences (< or = 32%) in several rifabutin and 25-O-desacetylrifabutin pharmacokinetic parameters between the regimens of rifabutin alone and rifabutin with placebo."( The effect of multiple doses of ritonavir on the pharmacokinetics of rifabutin.
Cato, A; Cavanaugh, J; Granneman, R; Hsu, A; Leonard, J; Shi, H, 1998
)
0.58
"To assess the pharmacokinetic interaction between ritonavir and saquinavir."( Pharmacokinetic interactions between two human immunodeficiency virus protease inhibitors, ritonavir and saquinavir.
Baroldi, P; Brown, F; Cao, G; Carothers, L; el-Shourbagy, T; Erdman, K; Granneman, GR; Hsu, A; Leonard, JM; Sun, E, 1998
)
0.77
"Coadministration of ritonavir markedly increased the area under the plasma concentration-time curve (AUC) and peak concentration of saquinavir (> 50-fold and 22-fold, respectively)."( Pharmacokinetic interactions between two human immunodeficiency virus protease inhibitors, ritonavir and saquinavir.
Baroldi, P; Brown, F; Cao, G; Carothers, L; el-Shourbagy, T; Erdman, K; Granneman, GR; Hsu, A; Leonard, JM; Sun, E, 1998
)
0.84
"Statistically significant decreases in ethinyl oestradiol mean Cmax (-32%) and mean AUC (-41%), and a statistically significant increase in the mean terminal elimination rate constant (+31%) were observed during concomitant ritonavir."( Effect of ritonavir on the pharmacokinetics of ethinyl oestradiol in healthy female volunteers.
Cavanaugh, JH; Eason, CJ; Granneman, GR; Hsu, A; Leonard, JM; Locke, CS; Ouellet, D; Qian, J, 1998
)
0.89
" After administration of a single dose of OC, serum ethinyl estradiol concentrations peaked at 4 hours and declined thereafter, with a typical half-life of 17 hours."( Effect of ritonavir on the pharmacokinetics of ethinyl oestradiol in healthy female volunteers.
Cavanaugh, JH; Eason, CJ; Granneman, GR; Hsu, A; Leonard, JM; Locke, CS; Ouellet, D; Qian, J, 1998
)
0.7
"The pharmacokinetic interaction between indinavir and ritonavir was evaluated in five groups of healthy adult volunteers to explore the potential for twice-daily (b."( Pharmacokinetic interaction between ritonavir and indinavir in healthy volunteers.
Berg, J; Cao, G; Carothers, L; Dennis, S; El-Shourbagy, T; Erdman, K; Granneman, GR; Hsu, A; Japour, A; Leonard, JM; Sun, E, 1998
)
0.82
"To evaluate the pharmacokinetic effects of concomitant administration of multiple doses of ritonavir and clarithromycin."( Pharmacokinetic interaction between ritonavir and clarithromycin.
Carlson, G; Cavanaugh, J; Granneman, GR; Guenther, H; Hsu, A; Leonard, JM; Ouellet, D, 1998
)
0.8
" The mean area under the plasma concentration-time curve (AUC) for clarithromycin increased by 77% with concomitant ritonavir, and the harmonic mean terminal half-life increased from 5 hours to 14 hours."( Pharmacokinetic interaction between ritonavir and clarithromycin.
Carlson, G; Cavanaugh, J; Granneman, GR; Guenther, H; Hsu, A; Leonard, JM; Ouellet, D, 1998
)
0.78
"Observational pharmacokinetic study."( The steady-state plasma pharmacokinetics of indinavir alone and in combination with a low dose of ritonavir in twice daily dosing regimens in HIV-1-infected individuals.
Beijnen, JH; Hoetelmans, RM; Hsu, A; Lange, JM; Meenhorst, PL; Mulder, JW; Schreij, G; van Heeswijk, RP; Veldkamp, AI, 1999
)
0.52
"028), and for the mean plasma elimination half-life (1."( The steady-state plasma pharmacokinetics of indinavir alone and in combination with a low dose of ritonavir in twice daily dosing regimens in HIV-1-infected individuals.
Beijnen, JH; Hoetelmans, RM; Hsu, A; Lange, JM; Meenhorst, PL; Mulder, JW; Schreij, G; van Heeswijk, RP; Veldkamp, AI, 1999
)
0.52
"Combination of indinavir and 100 mg ritonavir in twice daily dosing regimens significantly affects the pharmacokinetic profile of indinavir."( The steady-state plasma pharmacokinetics of indinavir alone and in combination with a low dose of ritonavir in twice daily dosing regimens in HIV-1-infected individuals.
Beijnen, JH; Hoetelmans, RM; Hsu, A; Lange, JM; Meenhorst, PL; Mulder, JW; Schreij, G; van Heeswijk, RP; Veldkamp, AI, 1999
)
0.8
"Open-label, crossover, pharmacokinetic study."( The effect of ritonavir on the pharmacokinetics of meperidine and normeperidine.
Bertz, RJ; Davey, R; Kress, DR; Pau, A; Piscitelli, SC, 2000
)
0.67
"Our data show that there are no significant differences in the pharmacokinetic parameters of daunorubicin in patients receiving DaunoXome in combination with indinavir and ritonavir compared with those in patients not receiving PIs."( The pharmacokinetics of liposomal encapsulated daunorubicin are not modified by HAART in patients with HIV-associated Kaposi's sarcoma.
Careddu, A; D'Incalci, M; Fumagalli, L; Lazzarin, A; Parisi, I; Viganò, MG; Zecca, B; Zucchetti, M, 2000
)
0.5
"Open-label, multi-dose, pharmacokinetic pilot study."( Once-daily dosing of saquinavir and low-dose ritonavir in HIV-1-infected individuals: a pharmacokinetic pilot study.
Beijnen, JH; Hoetelmans, RM; Lange, JM; Meenhorst, PL; Mulder, JW; van Heeswijk, RP; Veldkamp, AI, 2000
)
0.57
" A non-compartmental pharmacokinetic method was used to calculate the area under the plasma concentration versus time curve (AUC[0-24h]), the maximum and trough plasma concentrations (Cmax and Cmin), the time to reach Cmax (Tmax), the elimination half-life (t1/2), the apparent clearance (Cl/F), and the apparent volume of distribution (V/F)."( Once-daily dosing of saquinavir and low-dose ritonavir in HIV-1-infected individuals: a pharmacokinetic pilot study.
Beijnen, JH; Hoetelmans, RM; Lange, JM; Meenhorst, PL; Mulder, JW; van Heeswijk, RP; Veldkamp, AI, 2000
)
0.57
"This pharmacokinetic study indicates that the combination of 1600 mg of saquinavir (soft gelatin capsules) and 200 mg of ritonavir (liquid formulation) in a once-daily dosing regimen generally results in therapeutic plasma concentrations of saquinavir."( Once-daily dosing of saquinavir and low-dose ritonavir in HIV-1-infected individuals: a pharmacokinetic pilot study.
Beijnen, JH; Hoetelmans, RM; Lange, JM; Meenhorst, PL; Mulder, JW; van Heeswijk, RP; Veldkamp, AI, 2000
)
0.77
"To investigate the effect of the antiretroviral protease inhibitors saquinavir (soft gelatin capsule) and ritonavir on the pharmacokinetic properties and tolerability of sildenafil and to investigate the effect of sildenafil on the steady-state pharmacokinetics of saquinavir and ritonavir."( Pharmacokinetic interactions between sildenafil and saquinavir/ritonavir.
Buss, N; Fielding, A; Kleinermans, D; Muirhead, GJ; Wulff, MB, 2000
)
0.76
"Both protease inhibitors significantly increased Cmax, AUC, tmax and t(1/2) values for both sildenafil and UK-103, 320."( Pharmacokinetic interactions between sildenafil and saquinavir/ritonavir.
Buss, N; Fielding, A; Kleinermans, D; Muirhead, GJ; Wulff, MB, 2000
)
0.55
"005), prolonged elimination half-life (41 versus 3 hours; p < ."( Differential impairment of triazolam and zolpidem clearance by ritonavir.
Daily, JP; Durol, AL; Graf, JA; Greenblatt, DJ; Harmatz, JS; Hoffman, JL; Mertzanis, P; Shader, RI; von Moltke, LL, 2000
)
0.55
" Addition of ritonavir (100 mg) to saquinavir-SGC (1,200 to 1,800 mg/day) increased the area under the concentration-time curve (AUC) for saquinavir severalfold, and the intersubject peak concentration in plasma and AUC variability were reduced compared to those achieved with saquinavir-SGC alone (3,600 mg/day), while trough saquinavir levels (24 h post-dose) were substantially higher than the 90% inhibitory concentration calculated from HIV-1 clinical isolates."( Safety and pharmacokinetics of once-daily regimens of soft-gel capsule saquinavir plus minidose ritonavir in human immunodeficiency virus-negative adults.
Buss, N; Ehrensing, E; Gizzi, N; Kilby, JM; Oo, C; Saag, MS; Sfakianos, G; Siemon-Hryczyk, P, 2000
)
0.89
"Indinavir + ritonavir regimens show improved pharmacokinetic properties, allowing twice-daily dosing with food."( A retrospective, cohort-based survey of patients using twice-daily indinavir + ritonavir combinations: pharmacokinetics, safety, and efficacy.
Aarnoutse, RE; Blok, WL; Burger, DM; Dieleman, JP; Hugen, PW; Lange, JM; Meenhorst, PL; Mulder, JW; Prins, JM; Reiss, P; ten Veen, JH; van der Meer, JT; van der Poll, T, 2001
)
0.92
"To evaluate the safety and pharmacokinetic interaction between amprenavir (APV) and ritonavir (RTV)."( Pharmacokinetics and safety of amprenavir and ritonavir following multiple-dose, co-administration to healthy volunteers.
Lloyd, PP; Lou, Y; Piliero, PJ; Preston, SL; Sadler, BM; Stein, DS, 2001
)
0.79
"Three open-label, randomized, two-sequence, multiple-dose studies having the same design (7 days of APV or RTV alone followed by 7 days of both drugs together) used 450 or 900 mg APV with 100 or 300 mg RTV every 12 h with pharmacokinetic assessments on days 7 and 14."( Pharmacokinetics and safety of amprenavir and ritonavir following multiple-dose, co-administration to healthy volunteers.
Lloyd, PP; Lou, Y; Piliero, PJ; Preston, SL; Sadler, BM; Stein, DS, 2001
)
0.57
"A 24-hour methadone pharmacokinetic study was performed before antiretroviral therapy was begun and after 15 days of therapy."( Effect of ritonavir/saquinavir on stereoselective pharmacokinetics of methadone: results of AIDS Clinical Trials Group (ACTG) 401.
Aberg, J; Aweeka, F; D'Amico, R; Flexner, C; Gal, J; Gerber, JG; Gulick, R; Hsu, A; Hughes, V; Mildvan, D; Rosenkranz, S; Segal, Y, 2001
)
0.71
"To evaluate the pharmacokinetic interaction between ritonavir and mefloquine."( Pharmacokinetic interaction between mefloquine and ritonavir in healthy volunteers.
Carignan, G; Cooper, C; Gallicano, K; Khaliq, Y; McCarthy, A; Tisdale, C, 2001
)
0.81
"Healthy volunteers participated in two separate, nonfasted, three-treatment, three-period, longitudinal pharmacokinetic studies."( Pharmacokinetic interaction between mefloquine and ritonavir in healthy volunteers.
Carignan, G; Cooper, C; Gallicano, K; Khaliq, Y; McCarthy, A; Tisdale, C, 2001
)
0.56
"Study 1: Ritonavir caused less than 7% changes with high precision (90% CIs: -12% to 11%) in overall plasma exposure (AUC(0,168 h)) and peak concentration (Cmax) of mefloquine, its two enantiomers, and carboxylic acid metabolite, and in the metabolite/mefloquine and enantiomeric AUC ratios."( Pharmacokinetic interaction between mefloquine and ritonavir in healthy volunteers.
Carignan, G; Cooper, C; Gallicano, K; Khaliq, Y; McCarthy, A; Tisdale, C, 2001
)
0.98
" In general, this series of heterocyclic inhibitors displayed good activity (in both enzymatic and cellular tests) and low cellular toxicity; furthermore, several analogues exhibited improved pharmacokinetic parameters in animal models."( 4-Hydroxy-5,6-dihydropyrones as inhibitors of HIV protease: the effect of heterocyclic substituents at C-6 on antiviral potency and pharmacokinetic parameters.
Brodfuehrer, J; Domagala, J; Gajda, C; Gracheck, SJ; Hagen, SE; Holler, T; Hupe, D; Lovdahl, M; Lunney, EA; Nouhan, C; Pavlovsky, A; Saunders, J; Tait, BD; Urumov, A; VanderRoest, S; Wise, E; Zeikus, E; Zeikus, G, 2001
)
0.31
"Open-label, crossover, steady-state pharmacokinetic study."( Steady-state pharmacokinetics of twice-daily dosing of saquinavir plus ritonavir in HIV-1-infected individuals.
Beijnen, JH; Hoetelmans, RM; Lange, JM; Meenhorst, PL; Mulder, JW; Schreij, G; van der Geest, S; van Heeswijk, RP; Veldkamp, AI, 2001
)
0.54
" After 14 days, the patients came to the hospital for assessment of a pharmacokinetic profile during 12 hours."( Steady-state pharmacokinetics of twice-daily dosing of saquinavir plus ritonavir in HIV-1-infected individuals.
Beijnen, JH; Hoetelmans, RM; Lange, JM; Meenhorst, PL; Mulder, JW; Schreij, G; van der Geest, S; van Heeswijk, RP; Veldkamp, AI, 2001
)
0.54
"The median values of the pharmacokinetic parameters for SQV SGC (1000 mg twice daily, normal breakfast) were: AUC0-12h, 18."( Steady-state pharmacokinetics of twice-daily dosing of saquinavir plus ritonavir in HIV-1-infected individuals.
Beijnen, JH; Hoetelmans, RM; Lange, JM; Meenhorst, PL; Mulder, JW; Schreij, G; van der Geest, S; van Heeswijk, RP; Veldkamp, AI, 2001
)
0.54
" From a pharmacokinetic perspective, the combination of 1000 mg of SQV SGC twice daily and 100 mg of RTV twice daily seems to be a good option for further clinical evaluation."( Steady-state pharmacokinetics of twice-daily dosing of saquinavir plus ritonavir in HIV-1-infected individuals.
Beijnen, JH; Hoetelmans, RM; Lange, JM; Meenhorst, PL; Mulder, JW; Schreij, G; van der Geest, S; van Heeswijk, RP; Veldkamp, AI, 2001
)
0.54
"The authors assessed the impact of protease and reverse transcription (RT) mutations and individual pharmacokinetic parameters on virologic response to a four-drug regimen including ritonavir/saquinavir."( Pharmacokinetics and resistance mutations affect virologic response to ritonavir/saquinavir-containing regimens.
Birac, V; Breilh, D; Deneyrolles, M; Fleury, HJ; Mercié, P; Neau, D; Pellegrin, I; Pellegrin, JL; Saux, MC; Trylesinski, A, 2001
)
0.74
"Twenty-four patients without mycobacterial infection who were human immunodeficiency virus seropositive and who were receiving 400 mg each of ritonavir and saquinavir twice daily participated in a 3-period, 2-group longitudinal pharmacokinetic study."( A pharmacokinetic study of intermittent rifabutin dosing with a combination of ritonavir and saquinavir in patients infected with human immunodeficiency virus.
Cameron, DW; Carignan, G; Gallicano, K; Khaliq, Y; Tseng, A; Walmsley, S, 2001
)
0.74
"This was a randomized, double-blind, placebo-controlled parallel study in human immunodeficiency virus type 1 (HIV-1)-uninfected healthy subjects to investigate the pharmacokinetic interaction between indinavir (IDV) and ritonavir (RTV)."( Pharmacokinetic profile and tolerability of indinavir-ritonavir combinations in healthy volunteers.
Deutsch, PJ; Nessly, ML; Rhodes, RR; Saah, AJ; Seniuk, MA; Winchell, GA, 2001
)
0.74
" Pharmacokinetic parameters of saquinavir and ritonavir were determined and adverse events, vital signs, and clinical laboratory variables recorded."( Saquinavir and ritonavir pharmacokinetics following combined ritonavir and saquinavir (soft gelatin capsules) administration.
Bock, J; Buss, N; Hsu, A; Jorga, K; Snell, P, 2001
)
0.92
" Pharmacokinetic and safety profiles obtained in the current study indicate that the use of a combination with a lower dose of RTV and a higher dose of SQV than the 400 : 400 mg combination frequently used in clinical practice should be further explored."( Saquinavir and ritonavir pharmacokinetics following combined ritonavir and saquinavir (soft gelatin capsules) administration.
Bock, J; Buss, N; Hsu, A; Jorga, K; Snell, P, 2001
)
0.66
"Data from three pharmacokinetic drug interaction studies of amprenavir and ritonavir were used to develop a pharmacokinetic interaction model using NONMEM (nonlinear mixed-effect model)."( Pharmacokinetic modeling and simulations of interaction of amprenavir and ritonavir.
Sadler, BM; Sale, M; Stein, DS, 2002
)
0.78
"Fifty-six subjects completed both pharmacokinetic study days."( Pharmacokinetic interactions between protease inhibitors and statins in HIV seronegative volunteers: ACTG Study A5047.
Aberg, JA; Alston, B; Aweeka, F; Blaschke, T; Fang, F; Fichtenbaum, CJ; Gerber, JG; Kosel, B; Rosenkranz, SL; Segal, Y, 2002
)
0.31
" This observation may, at least in part, be attributed to the poor pharmacokinetic characteristics of the PIs."( Combination of protease inhibitors for the treatment of HIV-1-infected patients: a review of pharmacokinetics and clinical experience.
Beijnen, JH; Hoetelmans, RM; Lange, JM; Meenhorst, PL; Mulder, JW; van Heeswijk, RP; Veldkamp, A, 2001
)
0.31
"To examine the use of low-dose ritonavir as a pharmacokinetic enhancer for HIV protease inhibitors."( Low-dose ritonavir for protease inhibitor pharmacokinetic enhancement.
Rathbun, RC; Rossi, DR, 2002
)
1.02
"Combined analysis of pharmacokinetic data at steady state obtained from two open-label, randomized, parallel-group, multiple-dose, single-centre studies involving healthy volunteers."( The effect of ritonavir on saquinavir plasma concentration is independent of ritonavir dosage: combined analysis of pharmacokinetic data from 97 subjects.
Buss, N; Hill, A; Kilby, JM, 2002
)
0.68
"RTV enhances SQV concentrations to increase Cmax and Cmin."( The effect of ritonavir on saquinavir plasma concentration is independent of ritonavir dosage: combined analysis of pharmacokinetic data from 97 subjects.
Buss, N; Hill, A; Kilby, JM, 2002
)
0.68
" We compared olanzapine noncompartmental pharmacokinetic parameter values before and after ritonavir with a paired Student t test."( Influence of ritonavir on olanzapine pharmacokinetics in healthy volunteers.
Hon, YY; Jann, MW; Lawhorn, WD; Penzak, SR; Shirley, KL; Spratlin, V, 2002
)
0.9
"The response to regimens including lopinavir-ritonavir (LPV/r) in patients who have received multiple protease (PR) inhibitors (PI) can be analyzed in terms of human immunodeficiency virus type 1 (HIV-1) genotypic and pharmacokinetic (pK) determinants."( Human immunodeficiency virus type 1 genotypic and pharmacokinetic determinants of the virological response to lopinavir-ritonavir-containing therapy in protease inhibitor-experienced patients.
Breilh, D; Dabis, F; Dupon, M; Fleury, H; Lavignolle, V; Lawson-Ayayi, S; Masquelier, B; Morlat, P; Neau, D; Ragnaud, JM, 2002
)
0.78
"Eighteen healthy human immunodeficiency virus-negative subjects participated in an open-label, six-period, incomplete Latin-square crossover pharmacokinetic study."( Model-based analysis of the pharmacokinetic interactions between ritonavir, nelfinavir, and saquinavir after simultaneous and staggered oral administration.
Blaschke, TF; Flexner, C; Lu, JF; Rosenkranz, SL; Sheiner, LB, 2002
)
0.55
"Low doses of ritonavir, a strong inhibitor of cytochrome P450 3A4, enhances the pharmacokinetic profile of indinavir with increased serum levels."( Experience of a combination including indinavir 400 mg plus ritonavir 200 mg twice daily in HIV-infected patients: pharmacokinetic data.
Durand, A; Gastaut, JA; Lacarelle, B; Orticoni, M; Petit, N; Solas, C, 2002
)
0.93
" Pharmacokinetic parameters for nelfinavir and its active metabolite M8 were assessed on study days 15 and 16, after administration of the regimens with a full (610 kcal) or light (271 kcal) breakfast, respectively."( Pharmacokinetics, food intake requirements and tolerability of once-daily combinations of nelfinavir and low-dose ritonavir in healthy volunteers.
Aarnoutse, RE; Burger, DM; Droste, JA; Hekster, YA; Koopmans, PP; Popescu, M; Reiss, P; van Oosterhout, JJ, 2003
)
0.53
"The mechanisms of pharmacokinetic interactions of a novel anti-human immunodeficiency virus (anti-HIV-1) antagonist of chemokine receptor 5 (CCR5) [2-(R)-[N-methyl-N-(1-(R)-3-(S)-((4-(3-benzyl-1-ethyl-(1H)-pyrazol-5-yl)piperidin-1-yl)methyl)-4-(S)-(3-fluorophenyl)cyclopent-1-yl)amino]-3-methylbutanoic acid (MRK-1)] with ritonavir were evaluated in rats and monkeys."( Pharmacokinetics and interactions of a novel antagonist of chemokine receptor 5 (CCR5) with ritonavir in rats and monkeys: role of CYP3A and P-glycoprotein.
Baillie, TA; Chen, Q; Chiu, SH; Didolkar, V; Franklin, RB; Iliff, SA; Kumar, S; Kwei, GY; Lin, JH; Pearson, PG; Poon, GK; Wang, RW; Wang, Y; Yamazaki, M, 2003
)
0.71
"We evaluated 13 randomly selected HIV-1-infected subjects taking once-daily SQV SGC/RTV, 1600/100 mg, plus dual nucleoside reverse transcriptase inhibitors (NRTIs) in this pharmacokinetic (PK) substudy."( Pharmacokinetics of once-daily saquinavir hard-gelatin capsules and saquinavir soft-gelatin capsules boosted with ritonavir in HIV-1-infected subjects.
Burger, D; Cardiello, PG; Cooper, DA; Hill, A; Lange, JM; Mahanontharit, A; Monhaphol, T; Phanuphak, P; Ruxrungtham, K; van Heeswijk, RP, 2003
)
0.53
" Five HIV-infected patients on steady-state nelfinavir-containing therapy were subject to pharmacokinetic sampling for nelfinavir concentration twice: without sildenafil and with sildenafil 25 mg as a single dose."( Sildenafil does not alter nelfinavir pharmacokinetics.
Bratt, G; Ståhle, L, 2003
)
0.32
" Intensive steady-state pharmacokinetic sampling from baseline to 8 hours revealed very low plasma concentrations of nelfinavir: area under the plasma concentration-time curve (AUC(0-24)) <10% of adult population values for 750 mg every 8 hours and nonquantifiable concentrations of nelfinavir's principal metabolite (M8)."( Ritonavir-enhanced pharmacokinetics of nelfinavir/M8 during rifampin use.
Bergshoeff, AS; Burger, DM; Geelen, SP; Wolfs, TF, 2003
)
1.76
" Ritonavir resolved the pharmacokinetic interaction between rifampin and nelfinavir by boosting nelfinavir and, especially, M8 concentrations."( Ritonavir-enhanced pharmacokinetics of nelfinavir/M8 during rifampin use.
Bergshoeff, AS; Burger, DM; Geelen, SP; Wolfs, TF, 2003
)
2.67
"Thirty-six HIV-1-infected patients who participated in HIV-NAT 005 study gave informed consent to record a pharmacokinetic curve 4 weeks after starting a regimen containing either indinavir 800 mg every 8 h (n = 19) or indinavir 800 mg + ritonavir 100 mg every 12 h (n = 17)."( Pharmacokinetics and pharmacodynamics of indinavir with or without low-dose ritonavir in HIV-infected Thai patients.
Boyd, M; Burger, D; Cooper, D; Duncombe, C; Felderhof, M; Lange, J; Mahanontharit, A; Phanupak, P; Reiss, P; Ruxrungtham, K; Stek, M; Ubolyam, S, 2003
)
0.73
" Median and IQR values for indinavir AUC, Cmax and Cmin were 20."( Pharmacokinetics and pharmacodynamics of indinavir with or without low-dose ritonavir in HIV-infected Thai patients.
Boyd, M; Burger, D; Cooper, D; Duncombe, C; Felderhof, M; Lange, J; Mahanontharit, A; Phanupak, P; Reiss, P; Ruxrungtham, K; Stek, M; Ubolyam, S, 2003
)
0.55
" The pharmacokinetic profile of SQV was determined on day 10."( Pharmacokinetic and tolerability profile of twice-daily saquinavir hard gelatin capsules and saquinavir soft gelatin capsules boosted with ritonavir in healthy volunteers.
Hill, A; Kurowski, M; Möcklinghoff, C; Sawyer, A; Sternfeld, T, 2003
)
0.52
"There was a statistically significant increase in the geometric means of all the pharmacokinetic variables evaluated for SQV-HGC/RTV compared with SQV-SGC/RTV."( Pharmacokinetic and tolerability profile of twice-daily saquinavir hard gelatin capsules and saquinavir soft gelatin capsules boosted with ritonavir in healthy volunteers.
Hill, A; Kurowski, M; Möcklinghoff, C; Sawyer, A; Sternfeld, T, 2003
)
0.52
"To evaluate the pharmacokinetic effect of adding delavirdine mesylate to the antiretroviral regimens of human immunodeficiency virus (HIV)-infected patients stabilized on a full dosage of ritonavir (600 mg every 12 h), 12 HIV-1-infected subjects had delavirdine mesylate (400 mg every 8 h) added to their current antiretroviral regimens for 21 days."( Pharmacokinetics of ritonavir and delavirdine in human immunodeficiency virus-infected patients.
Adams, J; Cox, S; Della-Coletta, A; Hewitt, RG; Morse, GD; Shelton, MJ, 2003
)
0.83
"001), prolonged elimination half-life (6."( Short-term exposure to low-dose ritonavir impairs clearance and enhances adverse effects of trazodone.
Byron, S; Chen, G; Culm, KE; Daily, JP; Fogelman, SM; Graf, JA; Granda, BW; Greenblatt, DJ; Harmatz, JS; Mertzanis, P; Shader, RI; von Moltke, LL, 2003
)
0.6
"The aim of this study was to determine whether pharmacokinetic interactions between the protease inhibitors saquinavir soft gel, nelfinavir, and ritonavir are affected by the timing of administration."( Effect of simultaneous versus staggered dosing on pharmacokinetic interactions of protease inhibitors.
Aberg, JA; Blaschke, TF; Flexner, C; Rosenkranz, SL; Segal, Y; Sheiner, LB; Washington, CB, 2003
)
0.52
" We measured protease inhibitor concentrations over a 24-hour period by HPLC and estimated pharmacokinetic parameters by noncompartmental methods."( Effect of simultaneous versus staggered dosing on pharmacokinetic interactions of protease inhibitors.
Aberg, JA; Blaschke, TF; Flexner, C; Rosenkranz, SL; Segal, Y; Sheiner, LB; Washington, CB, 2003
)
0.32
"The aim of this study was to investigate the potential for pharmacokinetic interaction between escitalopram and ritonavir after concomitant administration of a single dose of each in healthy young subjects."( An evaluation of the potential for pharmacokinetic interaction between escitalopram and the cytochrome P450 3A4 inhibitor ritonavir.
Abramowitz, W; Gutierrez, MM; Rosenberg, J, 2003
)
0.74
" Blood was collected and plasma was analyzed for the pharmacokinetic parameters (maximum plasma concentration [C(max)], time to C(max) [T(max)], area under the plasma concentration-time curve, plasma elimination half-life, oral clearance, and apparent volume of distribution) of escitalopram, S-DCT, and ritonavir."( An evaluation of the potential for pharmacokinetic interaction between escitalopram and the cytochrome P450 3A4 inhibitor ritonavir.
Abramowitz, W; Gutierrez, MM; Rosenberg, J, 2003
)
0.7
" The pharmacokinetic parameters of ritonavir were also unaffected by coadministration of escitalopram."( An evaluation of the potential for pharmacokinetic interaction between escitalopram and the cytochrome P450 3A4 inhibitor ritonavir.
Abramowitz, W; Gutierrez, MM; Rosenberg, J, 2003
)
0.8
"In general, no pharmacokinetic interaction was observed between escitalopram and ritonavir in the present study."( An evaluation of the potential for pharmacokinetic interaction between escitalopram and the cytochrome P450 3A4 inhibitor ritonavir.
Abramowitz, W; Gutierrez, MM; Rosenberg, J, 2003
)
0.75
" Pharmacokinetic curves were obtained for lopinavir and saquinavir."( Lopinavir/ritonavir plus saquinavir in salvage therapy; pharmacokinetics, tolerability and efficacy.
Burger, DM; la Porte, CJ; Rockstroh, JK; Schneider, K; Wasmuth, JC, 2003
)
0.72
"The study was retrospective with a prospective pharmacokinetic study at a single centre."( Low-dose indinavir in combination with low-dose ritonavir: steady-state pharmacokinetics and long-term clinical outcome follow-up.
Justesen, US; Levring, AM; Lindberg, JA; Pedersen, C; Tauris, P; Thomsen, A, 2003
)
0.57
" The median IDV morning and evening Cmin were 434 ng/mL and 220 ng/mL, respectively."( Low-dose indinavir in combination with low-dose ritonavir: steady-state pharmacokinetics and long-term clinical outcome follow-up.
Justesen, US; Levring, AM; Lindberg, JA; Pedersen, C; Tauris, P; Thomsen, A, 2003
)
0.57
" However, rather low IDV Cmin suggests that the regimen should be evaluated further before its widespread use and that the regimen probably should be guided by pharmacokinetic evaluation."( Low-dose indinavir in combination with low-dose ritonavir: steady-state pharmacokinetics and long-term clinical outcome follow-up.
Justesen, US; Levring, AM; Lindberg, JA; Pedersen, C; Tauris, P; Thomsen, A, 2003
)
0.57
" High indinavir Cmax values have been associated with indinavir-related nephrotoxicity."( Administration of indinavir and low-dose ritonavir (800/100 mg twice daily) with food reduces nephrotoxic peak plasma levels of indinavir.
Aarnoutse, RE; Burger, DM; de Boo, TM; Fätkenheuer, G; Hekster, YA; Reiss, P; Rockstroh, JK; Schmitz, K; Schneider, K; Wasmuth, JC, 2003
)
0.58
"This was an open-label, randomized, two-treatment, two-period, cross-over pharmacokinetic study performed at steady state."( Administration of indinavir and low-dose ritonavir (800/100 mg twice daily) with food reduces nephrotoxic peak plasma levels of indinavir.
Aarnoutse, RE; Burger, DM; de Boo, TM; Fätkenheuer, G; Hekster, YA; Reiss, P; Rockstroh, JK; Schmitz, K; Schneider, K; Wasmuth, JC, 2003
)
0.58
"Administration of indinavir/ritonavir 800/100 mg on an empty stomach results in a higher indinavir Cmax compared to ingestion with a light meal."( Administration of indinavir and low-dose ritonavir (800/100 mg twice daily) with food reduces nephrotoxic peak plasma levels of indinavir.
Aarnoutse, RE; Burger, DM; de Boo, TM; Fätkenheuer, G; Hekster, YA; Reiss, P; Rockstroh, JK; Schmitz, K; Schneider, K; Wasmuth, JC, 2003
)
0.88
"At week 4 of the study, 12-hour pharmacokinetic profiles for indinavir/ritonavir were obtained for 20 HIV-infected subjects."( Pharmacokinetics of indinavir/ritonavir (800/100 mg) in combination with efavirenz (600 mg) in HIV-1-infected subjects.
Aarnoutse, RE; Boyd, MA; Burger, DM; Cooper, DA; Lange, JM; Phanuphak, P; Ruxrungtham, K; Stek, M; van Heeswijk, RP, 2003
)
0.84
" A >10-fold variation in indinavir Cmin was observed."( Pharmacokinetics of indinavir/ritonavir (800/100 mg) in combination with efavirenz (600 mg) in HIV-1-infected subjects.
Aarnoutse, RE; Boyd, MA; Burger, DM; Cooper, DA; Lange, JM; Phanuphak, P; Ruxrungtham, K; Stek, M; van Heeswijk, RP, 2003
)
0.61
"Despite the known pharmacokinetic interaction between efavirenz and indinavir/ritonavir, the combination of indinavir/ritonavir at 800/100 mg bid and efavirenz at 600 mg qd results in adequate minimum concentrations of both indinavir and efavirenz for treatment-naive patients."( Pharmacokinetics of indinavir/ritonavir (800/100 mg) in combination with efavirenz (600 mg) in HIV-1-infected subjects.
Aarnoutse, RE; Boyd, MA; Burger, DM; Cooper, DA; Lange, JM; Phanuphak, P; Ruxrungtham, K; Stek, M; van Heeswijk, RP, 2003
)
0.84
" This study confirmed the pharmacokinetic profiles of SQV during ethanol intake in rats."( Pharmacokinetic characterization of a human immunodeficiency virus protease inhibitor, saquinavir, during ethanol intake in rats.
Kageyama, M; Kimura, K; Kishida, T; Kuwahara, T; Shibata, N; Shirasaka, T; Takada, K; Toh, J; Yoshikawa, Y, 2003
)
0.32
" A two compartment pharmacokinetic model is employed to determine the time evolution of the intracellular concentrations of the active forms of drugs, and thereby drug efficacy."( Complex patterns of viral load decay under antiretroviral therapy: influence of pharmacokinetics and intracellular delay.
Dixit, NM; Perelson, AS, 2004
)
0.32
" Pharmacokinetic evaluations of steady-state concentrations of indinavir and ritonavir were performed before and after administration of rifampin (300 mg every day for 4 days)."( Pharmacokinetic interaction between rifampin and the combination of indinavir and low-dose ritonavir in HIV-infected patients.
Andersen, AB; Brøsen, K; Gerstoft, J; Justesen, US; Klitgaard, NA; Pedersen, C, 2004
)
0.77
"The pharmacokinetic parameters of lopinavir (LPV) were examined by administering Kaletra (LPV+ritonavir) to 8 healthy Japanese volunteers both in the fasting and postprandial conditions."( Pharmacokinetics of lopinavir after administration of Kaletra in healthy Japanese volunteers.
Ito, H; Kaneda, T; Mamiya, N; Nagaoka, K; Nakai, M; Oki, T; Sagisaka, M; Usami, Y; Utsumi, M; Yamanaka, K, 2004
)
0.54
" Pharmacokinetic evaluations of total and unbound methadone enantiomers (R and S) were conducted before and after SQV/rtv."( The effects of once-daily saquinavir/minidose ritonavir on the pharmacokinetics of methadone.
Berenson, CS; Buggé, CJ; Cloen, D; de Caprariis, PJ; DiFrancesco, R; Esch, A; Espinosa, O; Hewitt, RG; Ljungqvist, A; Palic, B; Schur, JL; Shelton, MJ, 2004
)
0.58
"To evaluate the safety and pharmacokinetic interaction between GW433908, ritonavir (RTV), and efavirenz (EFV)."( Pharmacokinetics and safety of GW433908 and ritonavir, with and without efavirenz, in healthy volunteers.
Ballow, C; Hendrix, CW; Lou, Y; Piliero, PJ; Preston, SL; Stein, DS; Wire, MB, 2004
)
0.82
" Amprenavir (APV) pharmacokinetic sampling and safety assessments were performed on the last day of each period."( Pharmacokinetics and safety of GW433908 and ritonavir, with and without efavirenz, in healthy volunteers.
Ballow, C; Hendrix, CW; Lou, Y; Piliero, PJ; Preston, SL; Stein, DS; Wire, MB, 2004
)
0.58
" Patients receiving the same doses of lopinavir/ritonavir (n = 10) or amprenavir with ritonavir (n = 8) were chosen as controls for pharmacokinetic analyses."( Deep salvage with amprenavir and lopinavir/ritonavir: correlation of pharmacokinetics and drug resistance with pharmacodynamics.
Baldini, F; Cauda, R; Cingolani, A; De Luca, A; Di Giambenedetto, S; Hoetelmans, RM, 2004
)
0.84
" Concentrations of the agents were quantitated at steady state after observed doses, and the pharmacokinetic parameters were determined."( Pharmacokinetic characteristics of ritonavir, zidovudine, lamivudine, and stavudine in children with human immunodeficiency virus infection.
Fletcher, CV; McIntosh, K; Nachman, SA; Pelton, S; Stanley, K; Wiznia, A; Yogev, R, 2004
)
0.6
"Therapy for HIV is complex, and pharmacodynamic data indicate that relationships exist between systemic concentrations of antiretroviral drugs and virologic response."( Pharmacokinetic characteristics of ritonavir, zidovudine, lamivudine, and stavudine in children with human immunodeficiency virus infection.
Fletcher, CV; McIntosh, K; Nachman, SA; Pelton, S; Stanley, K; Wiznia, A; Yogev, R, 2004
)
0.6
"Coadministration of lopinavir-ritonavir, an antiretroviral protease inhibitor, at the standard dose (400/100 mg twice a day [BID]) with the antituberculous agent rifampin is contraindicated because of a significant pharmacokinetic interaction due to induction of cytochrome P450 3A by rifampin."( Pharmacokinetics of adjusted-dose lopinavir-ritonavir combined with rifampin in healthy volunteers.
Bertz, R; Boeree, MJ; Burger, DM; Colbers, EP; Hekster, YA; Koopmans, PP; la Porte, CJ; Voncken, DS; Wikstrom, K, 2004
)
0.87
"Single-center, open-label, pharmacokinetic study."( Sex-related differences in the pharmacokinetics of once-daily saquinavir soft-gelatin capsules boosted with low-dose ritonavir in patients infected with human immunodeficiency virus type 1.
Diaz-Linares, M; Novak, RM; Pai, MP; Rodvold, KA; Schriever, CA, 2004
)
0.53
" Standard noncompartmental methods were used to calculate the pharmacokinetic parameters."( Sex-related differences in the pharmacokinetics of once-daily saquinavir soft-gelatin capsules boosted with low-dose ritonavir in patients infected with human immunodeficiency virus type 1.
Diaz-Linares, M; Novak, RM; Pai, MP; Rodvold, KA; Schriever, CA, 2004
)
0.53
" Digoxin pharmacokinetic parameter values were determined using noncompartmental methods."( Ritonavir decreases the nonrenal clearance of digoxin in healthy volunteers with known MDR1 genotypes.
Alfaro, RM; Falloon, J; Natarajan, V; Penzak, SR; Remaley, AT; Shen, JM, 2004
)
1.77
" After 12-hour pharmacokinetic sampling, the third protease inhibitor (PI) was added, and pharmacokinetics sampling was repeated 14 days later."( The pharmacokinetics, safety, and initial virologic response of a triple-protease inhibitor salvage regimen containing amprenavir, saquinavir, and ritonavir.
Corbett, AH; Eron, JJ; Fiscus, SA; Kashuba, AD; Rezk, NL, 2004
)
0.52
"Eighteen HIV-infected adults treated with the standard twice-daily SQV/RTV 1000/100 mg regimen were enrolled in this open-label, two-phase, crossover pharmacokinetic study."( Pharmacokinetics of once-daily saquinavir/ritonavir in HIV-infected subjects: comparison with the standard twice-daily regimen.
Back, D; Boffito, M; Dickinson, L; Fletcher, C; Gazzard, B; Higgs, C; Hill, A; Mandalia, S; Moyle, G; Nelson, M; Pozniak, A, 2004
)
0.59
"Compared with SQV 1000 mg twice daily, the Cmax of SQV following a 1600 mg and 2000 mg dose increased in a dose-proportional manner [geometric mean (95% CI) 1915 (1656-2850) ng/ml for 1000 mg, 2782 (2249-4330) ng/ml for 1600 mg and 4179 (3429-6105) ng/ml for 2000 mg doses, respectively]."( Pharmacokinetics of once-daily saquinavir/ritonavir in HIV-infected subjects: comparison with the standard twice-daily regimen.
Back, D; Boffito, M; Dickinson, L; Fletcher, C; Gazzard, B; Higgs, C; Hill, A; Mandalia, S; Moyle, G; Nelson, M; Pozniak, A, 2004
)
0.59
"A pharmacokinetic comparison of three dosing regimens of saquinavir/ritonavir was carried out: 1600/100 mg once-daily with 1000/100 mg twice-daily, and 1600/100 mg once-daily with 2000/100 mg once-daily."( Pharmacokinetic study of saquinavir hard gel caps/ritonavir in HIV-1-infected patients: 1600/100 mg once-daily compared with 2000/100 mg once-daily and 1000/100 mg twice-daily.
Ananworanich, J; Apateerapong, W; Autar, RS; Burger, D; Cooper, D; Hill, A; Hirschel, B; Lange, J; Phanuphak, P; Ruxrungtham, K; Sankote, J, 2004
)
0.81
" Two pharmacokinetic curves were plotted, at baseline (day 0) and 7 days after the switch."( Pharmacokinetic study of saquinavir hard gel caps/ritonavir in HIV-1-infected patients: 1600/100 mg once-daily compared with 2000/100 mg once-daily and 1000/100 mg twice-daily.
Ananworanich, J; Apateerapong, W; Autar, RS; Burger, D; Cooper, D; Hill, A; Hirschel, B; Lange, J; Phanuphak, P; Ruxrungtham, K; Sankote, J, 2004
)
0.58
"Compared with saquinavir/ritonavir 1600/100 mg once-daily dosing, the saquinavir AUC and Cmin improved significantly when dosed as 1000/100 mg twice-daily (53% and 299%, respectively), and as 2000/100 mg once-daily (71% and 65%, respectively)."( Pharmacokinetic study of saquinavir hard gel caps/ritonavir in HIV-1-infected patients: 1600/100 mg once-daily compared with 2000/100 mg once-daily and 1000/100 mg twice-daily.
Ananworanich, J; Apateerapong, W; Autar, RS; Burger, D; Cooper, D; Hill, A; Hirschel, B; Lange, J; Phanuphak, P; Ruxrungtham, K; Sankote, J, 2004
)
0.88
"On day 1, 12-hour pharmacokinetic profiles for saquinavir/ritonavir (1000/100 mg bid) were obtained for 18 subjects."( Steady-State pharmacokinetics of saquinavir hard-gel/ritonavir/fosamprenavir in HIV-1-infected patients.
Back, D; Boffito, M; Dickinson, L; Fletcher, C; Gazzard, B; Higgs, C; Hill, A; Moyle, G; Nelson, M; Pozniak, A, 2004
)
0.82
" The median pharmacokinetic parameters of lopinavir were as follows: area under the concentration-time curve from 0 to 12 h (AUC(0-12)), 85."( Steady-state pharmacokinetics of a double-boosting regimen of saquinavir soft gel plus lopinavir plus minidose ritonavir in human immunodeficiency virus-infected adults.
Azuaje, C; Clotet, B; Crespo, M; Diaz, M; Falcó, V; Lopez, RM; Ocaña, I; Pahissa, A; Pou, L; Ribera, E; Ruiz, I; Ruiz, L, 2004
)
0.54
" The main pharmacokinetic parameters were calculated using noncompartmental methods."( Effect of efavirenz treatment on the pharmacokinetics of nelfinavir boosted by ritonavir in healthy volunteers.
Burger, DM; Colbers, EP; de Graaff-Teulen, MJ; Hekster, YA; Ibanez, SM; Koopmans, PP; la Porte, CJ; Voncken, DS, 2004
)
0.55
"On day 1, 12 h pharmacokinetic profiles for saquinavir/ritonavir (1000/100 mg given twice daily) were obtained for 18 subjects."( Pharmacokinetics of saquinavir hard gel/ritonavir (1000/100 mg twice daily) when administered with tenofovir diproxil fumarate in HIV-1-infected subjects.
Back, D; Boffito, M; Di Perri, G; Gazzard, B; Hill, A; Moyle, G; Nelson, M; Pozniak, A; Stainsby-Tron, M; Tomkins, J, 2005
)
0.84
"The aim of this study was to develop and validate a population pharmacokinetic model of ritonavir, used as an antiviral agent or as a booster, in a large patient population and to identify factors influencing its pharmacokinetics."( Development and validation of a population pharmacokinetic model for ritonavir used as a booster or as an antiviral agent in HIV-1-infected patients.
Beijnen, JH; Crommentuyn, KM; Huitema, AD; Kappelhoff, BS; Mairuhu, AT; Meenhorst, PL; Mulder, JW; van Gorp, EC, 2005
)
0.79
" Furthermore, complete pharmacokinetic curves were available in some patients."( Development and validation of a population pharmacokinetic model for ritonavir used as a booster or as an antiviral agent in HIV-1-infected patients.
Beijnen, JH; Crommentuyn, KM; Huitema, AD; Kappelhoff, BS; Mairuhu, AT; Meenhorst, PL; Mulder, JW; van Gorp, EC, 2005
)
0.56
"From 186 patients 505 ritonavir plasma concentrations at a single time-point and 55 full pharmacokinetic profiles were available, resulting in a database of 1228 plasma ritonavir concentrations."( Development and validation of a population pharmacokinetic model for ritonavir used as a booster or as an antiviral agent in HIV-1-infected patients.
Beijnen, JH; Crommentuyn, KM; Huitema, AD; Kappelhoff, BS; Mairuhu, AT; Meenhorst, PL; Mulder, JW; van Gorp, EC, 2005
)
0.88
"The pharmacokinetic parameters of ritonavir were adequately described by our population pharmacokinetic model."( Development and validation of a population pharmacokinetic model for ritonavir used as a booster or as an antiviral agent in HIV-1-infected patients.
Beijnen, JH; Crommentuyn, KM; Huitema, AD; Kappelhoff, BS; Mairuhu, AT; Meenhorst, PL; Mulder, JW; van Gorp, EC, 2005
)
0.84
"To characterize the steady-state pharmacokinetic combination of the nonpeptidic protease inhibitor tipranavir (TPV) with ritonavir (RTV) in 95 healthy adult volunteers, a phase 1, single-center, open-label, randomized, parallel-group trial was conducted."( Pharmacokinetic characterization of different dose combinations of coadministered tipranavir and ritonavir in healthy volunteers.
Galitz, L; Johnson, P; MacGregor, TR; McCallister, S; Norris, SH; Sabo, JP,
)
0.56
" Trough concentration of >80 ng/ml and peak concentration of 2000-6000 ng/ml were regarded as sufficient."( Atazanavir for treatment of HIV infection in clinical routine: efficacy, pharmacokinetics and safety.
Feldt, T; Göbels, K; Häussinger, D; Kroidl, A; Kuschak, D; Leidel, R; Oette, M; Sagir, A, 2005
)
0.33
" Pharmacokinetic data are available for 32 patients, all patients had sufficient trough levels."( Atazanavir for treatment of HIV infection in clinical routine: efficacy, pharmacokinetics and safety.
Feldt, T; Göbels, K; Häussinger, D; Kroidl, A; Kuschak, D; Leidel, R; Oette, M; Sagir, A, 2005
)
0.33
" The variability of pharmacokinetic results in our sample supports therapeutic drug monitoring in patients treated with ATV."( Atazanavir for treatment of HIV infection in clinical routine: efficacy, pharmacokinetics and safety.
Feldt, T; Göbels, K; Häussinger, D; Kroidl, A; Kuschak, D; Leidel, R; Oette, M; Sagir, A, 2005
)
0.33
" LPV/r concentrations were measured by LC-MS/MS, and the data were analyzed by noncompartmental pharmacokinetic analysis."( Absence of circadian variation in the pharmacokinetics of lopinavir/ritonavir given as a once daily dosing regimen in HIV-1-infected patients.
Bourbeau, M; Cameron, DW; Garber, GE; Giguere, P; Seguin, I; van Heeswijk, RP, 2005
)
0.56
"This was a steady-state, open-label pharmacokinetic study of 19 patients."( Pharmacokinetics of reduced-dose indinavir/ritonavir 400/100 mg twice daily in HIV-1-infected Thai patients.
Boyd, M; Bunyaprawit, P; Burger, D; Chuenyam, T; Cooper, D; Horsakulchai, M; Lange, J; Mahanontharit, A; Mootsikapun, P; Phanuphak, P; Ruxrungtham, K; Sangkote, J; Ubolyam, S, 2005
)
0.59
" The median (interquartile ranges) for indinavir AUC, Cmax and Cmin were 18."( Pharmacokinetics of reduced-dose indinavir/ritonavir 400/100 mg twice daily in HIV-1-infected Thai patients.
Boyd, M; Bunyaprawit, P; Burger, D; Chuenyam, T; Cooper, D; Horsakulchai, M; Lange, J; Mahanontharit, A; Mootsikapun, P; Phanuphak, P; Ruxrungtham, K; Sangkote, J; Ubolyam, S, 2005
)
0.59
" Therapeutic Cmin levels of indinavir were achieved in >80% of the subjects and short-term virological response was satisfactory in this cohort of patients starting highly active antiretroviral therapy at an advanced disease stage with high baseline viral loads."( Pharmacokinetics of reduced-dose indinavir/ritonavir 400/100 mg twice daily in HIV-1-infected Thai patients.
Boyd, M; Bunyaprawit, P; Burger, D; Chuenyam, T; Cooper, D; Horsakulchai, M; Lange, J; Mahanontharit, A; Mootsikapun, P; Phanuphak, P; Ruxrungtham, K; Sangkote, J; Ubolyam, S, 2005
)
0.59
" Indinavir plasma levels were determined by HPLC and pharmacokinetic parameters by non-compartmental analysis."( Low-doses of indinavir boosted with ritonavir in HIV-infected Thai patients: pharmacokinetics, efficacy and tolerability.
Cressey, TR; Jourdain, G; Kunkeaw, S; Lallemant, M; Leenasirimakul, P; Puttimit, C; Sukrakanchana, PO; Tod, M, 2005
)
0.6
" The aim of this study was to evaluate the potential of pharmacokinetic interaction between cetirizine and ritonavir, the most potent cytochrome P450 (CYP) inhibitor."( Evaluation of pharmacokinetic interaction between cetirizine and ritonavir, an HIV-1 protease inhibitor, in healthy male volunteers.
Cremieux, AC; Delatour, F; Farinotti, R; Gautran, C; Melac, M; Moulaert, B; Otoul, C; Peytavin, G; Strolin-Benedetti, M, 2005
)
0.78
"An open-label, single-center, one-sequence crossover pharmacokinetic study was conducted in three running periods: cetirizine (CTZ) alone, ritonavir (RTV) alone and then CTZ plus RTV."( Evaluation of pharmacokinetic interaction between cetirizine and ritonavir, an HIV-1 protease inhibitor, in healthy male volunteers.
Cremieux, AC; Delatour, F; Farinotti, R; Gautran, C; Melac, M; Moulaert, B; Otoul, C; Peytavin, G; Strolin-Benedetti, M, 2005
)
0.77
" The RTV pharmacokinetic parameter values were not affected by CTZ co-treatment."( Evaluation of pharmacokinetic interaction between cetirizine and ritonavir, an HIV-1 protease inhibitor, in healthy male volunteers.
Cremieux, AC; Delatour, F; Farinotti, R; Gautran, C; Melac, M; Moulaert, B; Otoul, C; Peytavin, G; Strolin-Benedetti, M, 2005
)
0.57
"CTZ does not significantly affect the pharmacokinetic parameters of RTV, and the association does not, thus, require a modification of the dosage of the protease inhibitor."( Evaluation of pharmacokinetic interaction between cetirizine and ritonavir, an HIV-1 protease inhibitor, in healthy male volunteers.
Cremieux, AC; Delatour, F; Farinotti, R; Gautran, C; Melac, M; Moulaert, B; Otoul, C; Peytavin, G; Strolin-Benedetti, M, 2005
)
0.57
" We evaluated potential bidirectional pharmacokinetic interactions between calcium channel blockers and coadministered indinavir and ritonavir."( Pharmacokinetic interactions between indinavir plus ritonavir and calcium channel blockers.
Aberg, JA; Fichtenbaum, CJ; Gerber, JG; Glesby, MJ; Grosskopf, N; Hafner, R; Hall, S; Kendall, MA; Zolopa, AR, 2005
)
0.78
" Twenty-four-hour pharmacokinetic collection was performed on days 7 and 26, with 12-hour collection on day 19."( Pharmacokinetic interactions between indinavir plus ritonavir and calcium channel blockers.
Aberg, JA; Fichtenbaum, CJ; Gerber, JG; Glesby, MJ; Grosskopf, N; Hafner, R; Hall, S; Kendall, MA; Zolopa, AR, 2005
)
0.58
" Full pharmacokinetic curves were available from 45 patients."( Population pharmacokinetics of indinavir alone and in combination with ritonavir in HIV-1-infected patients.
Beijnen, JH; Huitema, AD; Kappelhoff, BS; Meenhorst, PL; Mulder, JW; Prins, JM; Sankatsing, SU; Van Gorp, EC, 2005
)
0.56
"The pharmacokinetic parameters of indinavir were adequately described by our population model."( Population pharmacokinetics of indinavir alone and in combination with ritonavir in HIV-1-infected patients.
Beijnen, JH; Huitema, AD; Kappelhoff, BS; Meenhorst, PL; Mulder, JW; Prins, JM; Sankatsing, SU; Van Gorp, EC, 2005
)
0.56
" Patients who were randomized to have ritonavir added (n=9) participated in three 12-h pharmacokinetic evaluations at baseline, week 4 and week 24."( The long-term pharmacokinetics and safety of adding low-dose ritonavir to a nelfinavir 1,250 mg twice-daily regimen in HIV-infected patients.
Andersen, AB; Black, FT; Gerstoft, J; Hansen, IM; Justesen, US; Klitgaard, NA; Mathiesen, LR; Pedersen, C, 2005
)
0.84
" There were no differences between the nelfinavir or M 8 pharmacokinetic parameters at weeks 4 and 24."( The long-term pharmacokinetics and safety of adding low-dose ritonavir to a nelfinavir 1,250 mg twice-daily regimen in HIV-infected patients.
Andersen, AB; Black, FT; Gerstoft, J; Hansen, IM; Justesen, US; Klitgaard, NA; Mathiesen, LR; Pedersen, C, 2005
)
0.57
"To develop a population pharmacokinetic model for lopinavir in combination with ritonavir, in which the interaction between both drugs was characterized, and in which relationships between patient characteristics and pharmacokinetics were identified."( Population pharmacokinetics of lopinavir in combination with ritonavir in HIV-1-infected patients.
Beijnen, JH; Crommentuyn, KM; Huitema, AD; Kappelhoff, BS; Mairuhu, AT; Meenhorst, PL; Mulder, JW; van Gorp, EC, 2005
)
0.8
" First, ritonavir data were fitted to a previously developed model to obtain individual Bayesian estimates of pharmacokinetic parameters."( Population pharmacokinetics of lopinavir in combination with ritonavir in HIV-1-infected patients.
Beijnen, JH; Crommentuyn, KM; Huitema, AD; Kappelhoff, BS; Mairuhu, AT; Meenhorst, PL; Mulder, JW; van Gorp, EC, 2005
)
1
"We have developed a model that has defined a time-independent inverse relationship between the exposure to ritonavir and the CL/F of lopinavir, and provided an adequate description of the pharmacokinetic parameters for the latter."( Population pharmacokinetics of lopinavir in combination with ritonavir in HIV-1-infected patients.
Beijnen, JH; Crommentuyn, KM; Huitema, AD; Kappelhoff, BS; Mairuhu, AT; Meenhorst, PL; Mulder, JW; van Gorp, EC, 2005
)
0.78
"Saquinavir/ritonavir data, dosed as 1600/100 mg once daily, from three separate pharmacokinetic studies, in 45 patients from Thailand and the UK, were pooled."( Interindividual variability of once-daily ritonavir boosted saquinavir pharmacokinetics in Thai and UK patients.
Ananworanich, J; Autar, RS; Boffito, M; Burger, DM; Cooper, DA; Hassink, E; Lange, JM; Phanuphak, P; Pozniak, A; Ruxrungtham, K; Siangphoe, U; Wit, FW, 2005
)
0.98
"Samples were collected for a 12-hour pharmacokinetic profile in all children."( Pharmacokinetics and 24-week efficacy/safety of dual boosted saquinavir/lopinavir/ritonavir in nucleoside-pretreated children.
Ananworanich, J; Bergshoeff, A; Burger, D; Engchanil, C; Hill, A; Kosalaraksa, P; Pancharoen, C; Ruxrungtham, K; Siangphoe, U, 2005
)
0.55
" Median area under the concentration curve at 0-12 hours and Cmin were 39."( Pharmacokinetics and 24-week efficacy/safety of dual boosted saquinavir/lopinavir/ritonavir in nucleoside-pretreated children.
Ananworanich, J; Bergshoeff, A; Burger, D; Engchanil, C; Hill, A; Kosalaraksa, P; Pancharoen, C; Ruxrungtham, K; Siangphoe, U, 2005
)
0.55
" Intensive pharmacokinetic sampling occurred after 4 weeks of therapy."( Efficacy, tolerability and pharmacokinetics of two nelfinavir-based regimens in human immunodeficiency virus-infected children and adolescents: pediatric AIDS clinical trials group protocol 403.
Acosta, EP; Aldrovandi, G; Chen, J; Damle, B; Hodge, J; Hughes, MD; King, JR; Nachman, S; Wiznia, A; Yogev, R, 2005
)
0.33
" NFV pharmacokinetic measurements were not statistically different between the treatment groups, yet exposure to the NFV metabolite, M8, was significantly higher in subjects receiving RTV."( Efficacy, tolerability and pharmacokinetics of two nelfinavir-based regimens in human immunodeficiency virus-infected children and adolescents: pediatric AIDS clinical trials group protocol 403.
Acosta, EP; Aldrovandi, G; Chen, J; Damle, B; Hodge, J; Hughes, MD; King, JR; Nachman, S; Wiznia, A; Yogev, R, 2005
)
0.33
"Corticosteroid therapy has been associated with bone toxicities (eg, osteonecrosis) and Cushing syndrome in HIV-infected patients; this may be partially attributable to a pharmacokinetic drug interaction between HIV protease inhibitors and corticosteroids."( Prednisolone pharmacokinetics in the presence and absence of ritonavir after oral prednisone administration to healthy volunteers.
Alfaro, RM; Formentini, E; Kovacs, J; Long, M; Natarajan, V; Penzak, SR, 2005
)
0.57
"The influence of saquinavir hard-gel capsules on lopinavir pharmacokinetic parameters was investigated using a population approach."( No significant influence of saquinavir hard-gel capsule administration on pharmacokinetics of lopinavir in combination with ritonavir: a population approach.
Allavena, C; Dailly, E; Gagnieu, MC; Jolliet, P; Raffi, F, 2005
)
0.54
"In this prospective trial, blood samples were drawn during a 6-hour time interval between two doses at enrolment to assess protease inhibitor (PI) pharmacokinetic parameters, and 4 months later to assess plasma trough and peak concentrations."( High variability of indinavir and nelfinavir pharmacokinetics in HIV-infected patients with a sustained virological response on highly active antiretroviral therapy.
Bonnet, B; Diquet, B; Duval, X; Goujard, C; Katlama, C; Legrand, M; Leport, C; Mentré, F; Panhard, X; Peytavin, G; Salmon-Céron, D; Taburet, AM; Vincent, I, 2005
)
0.33
" Ritonavir significantly increased indinavir elimination half-life and plasma exposure."( High variability of indinavir and nelfinavir pharmacokinetics in HIV-infected patients with a sustained virological response on highly active antiretroviral therapy.
Bonnet, B; Diquet, B; Duval, X; Goujard, C; Katlama, C; Legrand, M; Leport, C; Mentré, F; Panhard, X; Peytavin, G; Salmon-Céron, D; Taburet, AM; Vincent, I, 2005
)
1.24
" When the 2 hepatic impairment groups were combined, lopinavir Cmax and AUC12 were increased 20% to 30% compared to the controls."( Pharmacokinetics of lopinavir/ritonavir in HIV/hepatitis C virus-coinfected subjects with hepatic impairment.
Arribas, J; Brun, SC; Causemaker, SJ; Cepeda, C; DaSilva, B; García Cabanillas, JA; Li, J; Lorenzo, A; Peng, JZ; Pulido, F, 2006
)
0.62
" FPV 1,400 mg BID plus RTV 200 mg BID is not recommended due to an increased rate of marked hepatic transaminase elevations and lack of pharmacokinetic advantage."( Pharmacokinetic and safety evaluation of high-dose combinations of fosamprenavir and ritonavir.
Adamkiewicz, B; Lou, Y; Min, SS; Shelton, MJ; Wire, MB, 2006
)
0.56
"To assess potential pharmacokinetic (PK) interactions between atazanavir (ATV, 300 mg, once daily) and lopinavir (LPV, 400 mg, twice daily), both boosted by ritonavir (RTV, 100 mg)."( Ritonavir-boosted atazanavir-lopinavir combination: a pharmacokinetic interaction study of total, unbound plasma and cellular exposures.
Biollaz, J; Buclin, T; Cavassini, M; Colombo, S; Decosterd, LA; Franc, C; Guignard, N; Khonkarly, M; Rochat, B; Tarr, PE; Telenti, A, 2006
)
1.97
" Steady-state RTV AUCtau and Cmax levels were not significantly altered, whereas Ctau was 23% higher upon coadministration of SQV/RTV and TDF."( Pharmacokinetics of tenofovir disoproxil fumarate and ritonavir-boosted saquinavir mesylate administered alone or in combination at steady state.
Adda, N; Begley, JA; Blum, MR; Chittick, GE; Kearney, BP; Sorbel, JJ; Zong, J, 2006
)
0.58
" An intensive pharmacokinetic (PK) evaluation was performed on day 14 and multiple trough concentrations were subsequently collected."( Pharmacodynamics of antiretroviral agents in HIV-1 infected patients: using viral dynamic models that incorporate drug susceptibility and adherence.
Acosta, EP; Eron, JJ; Gerber, JG; Huang, Y; Kuritzkes, DR; Park, JG; Perelson, AS; Rosenkranz, SL; Wu, H; Yu, S, 2006
)
0.33
"A drug-drug interaction study was conducted to determine whether ritonavir (200 mg; 4 doses over 2 days) alters the pharmacokinetic disposition of bupropion (75 mg; once) coadministered to 7 healthy volunteers in a placebo-controlled 2-way crossover study."( Ritonavir has minimal impact on the pharmacokinetic disposition of a single dose of bupropion administered to human volunteers.
Court, MH; Greenblatt, DJ; Hesse, LM; von Moltke, LL, 2006
)
2.01
" Pharmacokinetic sampling was conducted on the last day of each treatment."( Coadministration of esomeprazole with fosamprenavir has no impact on steady-state plasma amprenavir pharmacokinetics.
Borland, J; Ford, SL; Lou, Y; Min, SS; Shelton, MJ; Wire, MB; Xue, ZG; Yuen, G, 2006
)
0.33
"A population pharmacokinetic analysis was performed in the context of therapeutic drug monitoring (87 patients, 121 samples)."( Influence of tenofovir, nevirapine and efavirenz on ritonavir-boosted atazanavir pharmacokinetics in HIV-infected patients.
Arvieux, C; Dailly, E; Jolliet, P; Perré, P; Raffi, F; Tattevin, P; Tribut, O, 2006
)
0.58
" Serial plasma samples were collected for 12-h pharmacokinetic profiles of saquinavir and ritonavir on days 10 and 15 and safety analysis on days 1, 4, 10, 15 and 29."( Effect of omeprazole on the pharmacokinetics of saquinavir-500 mg formulation with ritonavir in healthy male and female volunteers.
Back, D; Boffito, M; Fletcher, C; Gazzard, B; Pozniak, AL; Robinson, L; Schutz, M; Tolowinska, I; Unsworth, J; Winston, A, 2006
)
0.78
" No significant changes were observed in saquinavir elimination half life, ritonavir pharmacokinetic parameters or in safety laboratory tests."( Effect of omeprazole on the pharmacokinetics of saquinavir-500 mg formulation with ritonavir in healthy male and female volunteers.
Back, D; Boffito, M; Fletcher, C; Gazzard, B; Pozniak, AL; Robinson, L; Schutz, M; Tolowinska, I; Unsworth, J; Winston, A, 2006
)
0.79
" Minimum and maximum concentrations (C subsetmin, C subsetmax), area under the concentration-time curve (AUC subset0-12h), total clearance (CL subsettot) and half-life (t1/2) were calculated."( Pharmacokinetics and tolerability of a combination of indinavir, lopinavir and ritonavir in multiply pretreated HIV-1 infected adults.
Harder, S; Klauke, S; Kurowski, M; Lutz, T; Müller, A; Rottmann, C; Staszewski, S; von Hentig, N, 2006
)
0.56
" 3,467 ng*h/mL; Cmin 220 ng/mL vs."( Pharmacokinetics and tolerability of a combination of indinavir, lopinavir and ritonavir in multiply pretreated HIV-1 infected adults.
Harder, S; Klauke, S; Kurowski, M; Lutz, T; Müller, A; Rottmann, C; Staszewski, S; von Hentig, N, 2006
)
0.56
"To assess the impact of baseline HIV-1 substitutions, individual pharmacokinetic (PK) parameters (Cmin, Cmax, area under the curve [AUC0-->24 h]) and genotype-inhibitory quotient (GIQ) on virological responses (VR) to atazanavir-ritonavir (300 mg/100 mg)-based highly active antiretroviral therapy (HAART) in 71 antiretroviral-experienced, atazanavir-naive patients in virological failure (VF) on HAART."( Virological responses to atazanavir-ritonavir-based regimens: resistance-substitutions score and pharmacokinetic parameters (Reyaphar study).
Boucher, S; Breilh, D; Fleury, H; Lazaro, E; Neau, D; Pellegrin, I; Pellegrin, JL; Ragnaud, JM; Saux, MC; Schrive, MH; Vray, M, 2006
)
0.79
" Respective steady-state Cmin, Cmax and AUC0-->24 h were 300 (200; 700) ng/ml, 620 (430; 750) ng/ml and 78,000 (61,000; 94,000) ng."( Virological responses to atazanavir-ritonavir-based regimens: resistance-substitutions score and pharmacokinetic parameters (Reyaphar study).
Boucher, S; Breilh, D; Fleury, H; Lazaro, E; Neau, D; Pellegrin, I; Pellegrin, JL; Ragnaud, JM; Saux, MC; Schrive, MH; Vray, M, 2006
)
0.61
" The mean (standard deviation) AUC0-24, Cmax and Cmin of lopinavir were 149."( Pharmacokinetics of a once-daily regimen of lopinavir/ritonavir in HIV-1-infected children.
Burger, D; de Groot, R; van der Lee, M; Verweel, G, 2006
)
0.58
"Our findings indicate that 460/115 mg/m2 lopinavir/ritonavir once daily leads to mean pharmacokinetic parameters comparable to data of 800/200 mg lopinavir/ritonavir once daily in adults, although the variability observed in the trough levels is much higher in children."( Pharmacokinetics of a once-daily regimen of lopinavir/ritonavir in HIV-1-infected children.
Burger, D; de Groot, R; van der Lee, M; Verweel, G, 2006
)
0.83
" Pharmacokinetic sampling was performed on day 8 of each treatment, and samples were analyzed for FPV, amprenavir (APV), LPV, and RTV concentrations by high-performance liquid chromatography-tandem mass spectrometry."( Dose separation does not overcome the pharmacokinetic interaction between fosamprenavir and lopinavir/ritonavir.
Corbett, AH; Eron, JJ; Kalvass, LA; Kashuba, AD; Lim, ML; Ngo, LT; Patterson, KB; Tien, HC, 2006
)
0.55
" Atazanavir was increased to 200 mg and pharmacokinetic assessment repeated (day 30)."( Pharmacokinetics of saquinavir hard-gel/ritonavir and atazanavir when combined once daily in HIV Type 1-infected individuals administered different atazanavir doses.
Back, D; Boffito, M; Dickinson, L; Fletcher, C; Gazzard, B; Hill, A; Maitland, D; Moyle, G; Nelson, M; Pozniak, A, 2006
)
0.6
" The pharmacokinetic profile of saquinavir-SGC was analyzed in a subset of randomly selected patients."( Efficacy, safety and pharmacokinetics of once-daily saquinavir soft-gelatin capsule/ritonavir in antiretroviral-naive, HIV-infected patients.
Burnside, AF; Jayaweera, DT; Montaner, JS; Saag, MS; Schutz, M; Schwartz, R; Walmsley, S, 2006
)
0.56
" Pharmacokinetic profiles in 6 patients showed an observed median Cmin at 24 hours of 429 ng/mL (range, 68-1750 ng/mL)."( Efficacy, safety and pharmacokinetics of once-daily saquinavir soft-gelatin capsule/ritonavir in antiretroviral-naive, HIV-infected patients.
Burnside, AF; Jayaweera, DT; Montaner, JS; Saag, MS; Schutz, M; Schwartz, R; Walmsley, S, 2006
)
0.56
" This study aimed to assess the ATV pharmacokinetic profile in a target population of HIV patients, to characterize interpatient and intrapatient variability, and to identify covariates that might influence ATV disposition."( Population pharmacokinetics of atazanavir in patients with human immunodeficiency virus infection.
Biollaz, J; Buclin, T; Cavassini, M; Colombo, S; Csajka, C; Décosterd, LA; Telenti, A, 2006
)
0.33
" When comparing pharmacokinetic values in the nine patients receiving saquinavir/ritonavir with and without atazanavir, the median cellular saquinavir AUC0-24 was significantly increased (34."( Influence of atazanavir 200 mg on the intracellular and plasma pharmacokinetics of saquinavir and ritonavir 1600/100 mg administered once daily in HIV-infected patients.
Back, D; Boffito, M; Ford, J; Gazzard, B; Hill, A; Khoo, S; Maitland, D; Moyle, G; Nelson, M; Pozniak, A, 2006
)
0.78
"The aim of this study is to investigate in vivo the oral bioavailability of ritonavir and to evaluate the pharmacokinetic model that best describes the plasma concentration behavior after oral and intravenous administration."( Bioavailability and pharmacokinetic model for ritonavir in the rat.
Casabó, VG; Lledó-García, R; Merino-Sanjuán, M; Nácher, A; Prats-García, L, 2007
)
0.83
" A pharmacokinetic study in healthy volunteers was conducted to assess the potential for a drug interaction between these agents."( Pharmacokinetics and safety of tenofovir disoproxil fumarate on coadministration with lopinavir/ritonavir.
Cheng, AK; Ebrahimi, R; Kearney, BP; Mathias, A; Mittan, A; Sayre, J, 2006
)
0.55
" Pharmacokinetic assessments were performed over 24 hours on days 7, 21, and 35."( Pharmacokinetics and safety of tenofovir disoproxil fumarate on coadministration with lopinavir/ritonavir.
Cheng, AK; Ebrahimi, R; Kearney, BP; Mathias, A; Mittan, A; Sayre, J, 2006
)
0.55
" This randomized, two 14-day-period, crossover pharmacokinetic study compared the steady-state plasma APV concentrations, safety, and tolerability of FPV at 1,400 mg QD boosted with either 100 mg or 200 mg of RTV QD in 36 healthy volunteers."( Plasma amprenavir pharmacokinetics and tolerability following administration of 1,400 milligrams of fosamprenavir once daily in combination with either 100 or 200 milligrams of ritonavir in healthy volunteers.
Lancaster, CT; Lou, Y; Luber, AD; Pappa, KA; Ruane, PJ; Shelton, MJ; Wire, MB, 2007
)
0.53
" Thus, for ritonavir AUC12, Cmax and Cmin in HCV+/FIB+patients were 63%, 86% and 100% higher, respectively, when compared with those parameters in HCV-patients (p=0."( Lopinavir/ritonavir pharmacokinetics in HIV and hepatitis C virus co-infected patients without liver function impairment: influence of liver fibrosis.
Barbanoj, MJ; Blanco, A; Clotet, B; Costa, J; DelaVarga, M; Domingo, P; Miranda, C; Miranda, J; Moltó, J; Negredo, E; Tural, C; Valle, M; Vilaró, J, 2007
)
1.13
"Darunavir (DRV; TMC114; Prezista) is a human immunodeficiency virus (HIV) protease inhibitor used in combination with low-dose ritonavir (RTV) (DRV/r) as a pharmacokinetic enhancer."( Pharmacokinetic interaction between darunavir boosted with ritonavir and omeprazole or ranitidine in human immunodeficiency virus-negative healthy volunteers.
De Marez, T; De Paepe, E; De Pauw, M; Hoetelmans, RM; Lefebvre, E; Parys, W; Sekar, VJ, 2007
)
0.79
" Treatments were separated by 10 days, and pharmacokinetic analyses were performed on days 11, 32, and 53."( Pharmacokinetics of saquinavir with atazanavir or low-dose ritonavir administered once daily (ASPIRE I) or twice daily (ASPIRE II) in seronegative volunteers.
Acosta, EP; Becker, SL; Kakuda, TN; King, JR; Paul, S; Tse, MM, 2007
)
0.58
"This study evaluated the steady-state pharmacokinetic interaction between ritonavir-boosted saquinavir and nelfinavir."( Saquinavir, nelfinavir and M8 pharmacokinetics following combined saquinavir, ritonavir and nelfinavir administration.
Arastéh, K; Becker, M; Berger, M; Breske, A; Herzmann, C; Hill, A; Kruse, G; Kurowski, M; Schulbin, H; Steinmüller, J; Stocker, H, 2007
)
0.8
" Pharmacokinetic assessments were performed before and 7 days after the start of combined treatment with nelfinavir/saquinavir/ritonavir."( Saquinavir, nelfinavir and M8 pharmacokinetics following combined saquinavir, ritonavir and nelfinavir administration.
Arastéh, K; Becker, M; Berger, M; Breske, A; Herzmann, C; Hill, A; Kruse, G; Kurowski, M; Schulbin, H; Steinmüller, J; Stocker, H, 2007
)
0.77
" The pharmacokinetic and safety profile of BCV-RTV supports continued investigation in HIV-1-infected subjects."( Safety and pharmacokinetics of brecanavir, a novel human immunodeficiency virus type 1 protease inhibitor, following repeat administration with and without ritonavir in healthy adult subjects.
Anderson, MT; Ford, SL; Johnson, MA; Murray, SC; Ng-Cashin, J; Reddy, YS, 2007
)
0.54
" Patients were given a 12/24-h pharmacokinetic assessment at steady state."( Pharmacokinetics of saquinavir, atazanavir, and ritonavir in a twice-daily boosted double-protease inhibitor regimen.
Dauer, B; Harder, S; Klauke, S; Kurowski, M; Lutz, T; Müller, A; Oertel, B; Rottmann, C; Staszewski, S; von Hentig, N; Wolf, T, 2007
)
0.6
" The impact of baseline HIV type 1 (HIV-1) mutations, pharmacokinetic (PK) parameters, and genotype inhibitory quotient (GIQ) on the virological response at week 12 (W12) was assessed."( Interpretation of genotype and pharmacokinetics for resistance to fosamprenavir-ritonavir-based regimens in antiretroviral-experienced patients.
Boucher, S; Breilh, D; Coureau, G; Dabis, F; Fleury, H; Lacoste, D; Lazaro, E; Merel, P; Neau, D; Pellegrin, I; Pellegrin, JL; Saux, MC; Thiébaut, R, 2007
)
0.57
" A complete pharmacokinetic evaluation of the steady-state concentrations of atazanavir and ritonavir was performed."( Pharmacokinetic interaction between rifampicin and ritonavir-boosted atazanavir in HIV-infected patients.
Blanco, JL; Gatell, JM; López-Púa, Y; Mallolas, J; Martínez, E; Nomdedeu, M; Sarasa, M; Soriano, A, 2007
)
0.81
"In all three cases, more than 50% of the time the atazanavir level was below the minimum recommended trough plasma level (150 ng/mL according to current pharmacokinetic guidelines) to inhibit HIV wild-type replication."( Pharmacokinetic interaction between rifampicin and ritonavir-boosted atazanavir in HIV-infected patients.
Blanco, JL; Gatell, JM; López-Púa, Y; Mallolas, J; Martínez, E; Nomdedeu, M; Sarasa, M; Soriano, A, 2007
)
0.59
" In conclusion, given the pharmacokinetic profile, efficacy, and tolerability of this regimen, once-daily low-dose SQVr may be considered a treatment option in treatment-naive or limited PI-experienced HIV-infected patients, with the additional benefit of being currently the least-expensive PI-based regimen available."( Clinical and pharmacokinetic data support once-daily low-dose boosted saquinavir (1,200 milligrams saquinavir with 100 milligrams ritonavir) in treatment-naive or limited protease inhibitor-experienced human immunodeficiency virus-infected patients.
Gutierrez, A; Lopez-Cortes, LF; Marin-Niebla, A; Mata, R; Pascual, R; Rodriguez, M; Ruiz-Valderas, R; Viciana, P, 2007
)
0.54
" Pharmacokinetic parameters for darunavir and ritonavir were determined under fasted conditions and following a standard breakfast, a high-fat breakfast, a nutritional protein-rich drink, or a croissant with coffee."( The effect of different meal types on the pharmacokinetics of darunavir (TMC114)/ritonavir in HIV-negative healthy volunteers.
De Paepe, E; De Pauw, M; Hoetelmans, RM; Kestens, D; Lefebvre, E; Sekar, V; Spinosa-Guzman, S; Vangeneugden, T, 2007
)
0.82
" Pharmacokinetic (PK) blood draws were performed on days 7, 17, and 27."( Pharmacokinetics of emtricitabine, tenofovir, and GS-9137 following coadministration of emtricitabine/tenofovir disoproxil fumarate and ritonavir-boosted GS-9137.
Cheng, A; Kearney, BP; Ramanathan, S; Shen, G, 2007
)
0.54
" Lopinavir and ritonavir pharmacokinetic parameters were evaluated."( Lack of effect of gastric acid-reducing agents on the pharmacokinetics of lopinavir/ritonavir in HIV-infected patients.
Awni, W; Brun, S; Chiu, YL; King, KR; Klein, CE; Naylor, C; Woodward, WC, 2007
)
0.92
" Ritonavir-boosted double-protease inhibitor (PI)-only regimens are such an option but are prone to pharmacokinetic interactions."( The safety, efficacy, and pharmacokinetic profile of a switch in antiretroviral therapy to saquinavir, ritonavir, and atazanavir alone for 48 weeks and a switch in the saquinavir formulation.
Cooper, DA; Emery, S; Law, M; MacRae, K; Mallon, PW; Satchell, C; Schutz, M; Williams, KM; Winston, A, 2007
)
1.46
" The study also assessed the pharmacokinetic profile of a change in the SQV formulation, from 200 mg to 500 mg, in 2 regimens (SQV-RTV twice per day plus NRTIs [arm 1] and SQV-RTV-ATV once per day without NRTIs [arm 2]) in human immunodeficiency virus type 1-infected subjects (plasma human immunodeficiency virus RNA level, <50 copies/mL)."( The safety, efficacy, and pharmacokinetic profile of a switch in antiretroviral therapy to saquinavir, ritonavir, and atazanavir alone for 48 weeks and a switch in the saquinavir formulation.
Cooper, DA; Emery, S; Law, M; MacRae, K; Mallon, PW; Satchell, C; Schutz, M; Williams, KM; Winston, A, 2007
)
0.55
"The protease inhibitor indinavir is characterized by an important interindividual pharmacokinetic variability, which results from the actions of the metabolizing enzymes cytochrome P450 (CYP) 3A and the multidrug efflux pump P-glycoprotein (P-gp), encoded by MDR1."( Minimal effect of MDR1 and CYP3A5 genetic polymorphisms on the pharmacokinetics of indinavir in HIV-infected patients.
Bourgarel-Rey, V; Brunet, C; Drogoul, MP; Durand, A; Frixon-Marin, V; Gastaut, JA; Lacarelle, B; Poizot-Martin, I; Quaranta, S; Simon, N; Solas, C, 2007
)
0.34
"The MDR1 C3435T genotype affects the absorption constant of indinavir suggesting that P-gp may be implicated in its pharmacokinetic variability."( Minimal effect of MDR1 and CYP3A5 genetic polymorphisms on the pharmacokinetics of indinavir in HIV-infected patients.
Bourgarel-Rey, V; Brunet, C; Drogoul, MP; Durand, A; Frixon-Marin, V; Gastaut, JA; Lacarelle, B; Poizot-Martin, I; Quaranta, S; Simon, N; Solas, C, 2007
)
0.34
"The effect of tenofovir disoproxil fumarate (TDF) in combination with two boosted fosamprenavir regimens on amprenavir pharmacokinetic parameters was assessed in this prospective phase I crossover study with 30 healthy volunteers."( Fosamprenavir/ritonavir plus tenofovir does not affect amprenavir pharmacokinetics: no effect of tenofovir.
Banik, N; Breske, A; Kruse, G; Kurowski, M; Mazur, D; Richter, H; Stocker, H; Walli, RK, 2007
)
0.7
"To assess the pharmacokinetic profile, safety and virological response of two novel investigational antiretroviral agents when used in combination in HIV-1-infected subjects with multidrug-resistant virus."( Pharmacokinetics and antiretroviral response to darunavir/ritonavir and etravirine combination in patients with high-level viral resistance.
Boffito, M; Fletcher, C; Gazzard, B; Hoetelmans, R; Jackson, A; Miralles, D; Moyle, G; Nelson, M; Pozniak, A; Tolowinska, I; Winston, A, 2007
)
0.58
" Detailed pharmacokinetic assessments of darunavir and etravirine were determined on days 7 and 28."( Pharmacokinetics and antiretroviral response to darunavir/ritonavir and etravirine combination in patients with high-level viral resistance.
Boffito, M; Fletcher, C; Gazzard, B; Hoetelmans, R; Jackson, A; Miralles, D; Moyle, G; Nelson, M; Pozniak, A; Tolowinska, I; Winston, A, 2007
)
0.58
"This first study to assess the use of etravirine and darunavir in HIV-1-infected subjects with no treatment options showed highly effective virological and immunological responses over 24 weeks of therapy with no new safety concerns or unexpected pharmacokinetic interactions."( Pharmacokinetics and antiretroviral response to darunavir/ritonavir and etravirine combination in patients with high-level viral resistance.
Boffito, M; Fletcher, C; Gazzard, B; Hoetelmans, R; Jackson, A; Miralles, D; Moyle, G; Nelson, M; Pozniak, A; Tolowinska, I; Winston, A, 2007
)
0.58
"TMC114 is a new HIV protease inhibitor, used in combination with low-dose ritonavir (TMC114/r) as a pharmacokinetic enhancer."( Pharmacokinetic interaction between TMC114/ritonavir and tenofovir disoproxil fumarate in healthy volunteers.
De Doncker, P; De Pauw, M; Hill, A; Hoetelmans, RM; Lefebvre, E; Mariën, K; Peeters, M; Sekar, V; Woodfall, B, 2007
)
0.83
" Pharmacokinetic parameters estimated by noncompartmental method were reported as 90% confidence intervals (CIs) about the geometric mean ratio (GMR)."( Lopinavir/Ritonavir pharmacokinetic profile: impact of sex and other covariates following a change from twice-daily to once-daily therapy.
Acosta, EP; Binongo, JN; Chuck, SK; Lennox, JL; Ofotokun, I; Palau, M, 2007
)
0.74
" A human pharmacokinetic model of VX-950 co-administered with low-dose ritonavir suggested that improved efficacy and/or dosing convenience may be feasible by pharmacokinetic enhancement with ritonavir."( Pharmacokinetic enhancement of the hepatitis C virus protease inhibitors VX-950 and SCH 503034 by co-dosing with ritonavir.
Chen, HJ; Chovan, LE; Dandliker, PJ; Guan, Z; Hernandez, L; Kempf, DJ; Klein, C; Klein, LL; Lau, YY; Marsh, KC; Randolph, JT; Turner, TM; Yeung, C, 2007
)
0.78
"Atazanavir/ritonavir plasma concentrations were measured by liquid chromatography tandem mass spectrometry, and the geometric means of minimum and maximum concentrations (C(min), C(max)), the area under the time-concentration curve (AUC), half-life (t(1/2)) and total clearance (CL(tot)) were subject to a matched pairs-analysis."( Tenofovir comedication does not impair the steady-state pharmacokinetics of ritonavir-boosted atazanavir in HIV-1-infected adults.
Dauer, B; Haberl, A; Harder, S; Klauke, S; Lutz, T; Staszewski, S; von Hentig, N, 2007
)
0.96
"The coadministration of tenofovir-DF did not impair the plasma concentrations of ritonavir-boosted atazanavir in a pharmacokinetic analysis of patient pairs matched for gender, ethnicity, weight and CDC status."( Tenofovir comedication does not impair the steady-state pharmacokinetics of ritonavir-boosted atazanavir in HIV-1-infected adults.
Dauer, B; Haberl, A; Harder, S; Klauke, S; Lutz, T; Staszewski, S; von Hentig, N, 2007
)
0.8
"This open-label, crossover study investigated the pharmacokinetic interaction between TMC114 (darunavir [Prezista]), administered with low-dose ritonavir (TMC114/r) and efavirenz (EFV) in HIV-negative, healthy volunteers."( Pharmacokinetic interaction between TMC114/r and efavirenz in healthy volunteers.
De Pauw, M; Hoetelmans, RM; Lefebvre, E; Mariën, K; Peeters, M; Sekar, VJ, 2007
)
0.54
" TMC114/r and EFV coadministration resulted in TMC114 Cmin, Cmax and AUC12h decreases of 31%, 15% and 13%, respectively."( Pharmacokinetic interaction between TMC114/r and efavirenz in healthy volunteers.
De Pauw, M; Hoetelmans, RM; Lefebvre, E; Mariën, K; Peeters, M; Sekar, VJ, 2007
)
0.34
"The clinical significance of the changes in AUC and Cmin seen with TMC114/r and EFV coadministration has not been established; this combination should be used with caution."( Pharmacokinetic interaction between TMC114/r and efavirenz in healthy volunteers.
De Pauw, M; Hoetelmans, RM; Lefebvre, E; Mariën, K; Peeters, M; Sekar, VJ, 2007
)
0.34
" For TMC125 200 mg twice daily coadministered with DRV/r, AUC12h, Cmax and Cmin of TMC125 were 80%, 81% and 67% greater, respectively, versus TMC125 100 mg twice daily alone."( Pharmacokinetics of darunavir/ritonavir and TMC125 alone and coadministered in HIV-negative volunteers.
De Smedt, G; Hoetelmans, RM; Kakuda, TN; Lefebvre, E; Peeters, M; Schöller-Gyüre, M; Sekar, V; Woodfall, B, 2007
)
0.63
"HIV-infected subjects on abacavir (600 mg once daily) plus two nucleoside reverse transcriptase inhibitors (NRTIs) (excluding tenofovir) underwent a 24 h pharmacokinetic assessment for plasma abacavir concentrations."( Abacavir plasma pharmacokinetics in the absence and presence of atazanavir/ritonavir or lopinavir/ritonavir and vice versa in HIV-infected patients.
Back, D; Boffito, M; Bonora, S; D'Avolio, A; Else, L; Gazzard, B; Mandalia, S; Moyle, G; Nelson, M; Pozniak, A; Waters, LJ, 2007
)
0.57
" The clinical consequences of these substantial pharmacokinetic changes should be investigated."( Lopinavir-ritonavir dramatically affects the pharmacokinetics of irinotecan in HIV patients with Kaposi's sarcoma.
Corona, G; Innocenti, F; Sandron, S; Sartor, I; Tirelli, U; Toffoli, G; Vaccher, E, 2008
)
0.75
" Absorption is followed by a fast distribution/elimination phase and a subsequent slower elimination phase with a terminal elimination half-life of 15 hours in the presence of low-dose ritonavir."( Clinical pharmacokinetics of darunavir.
Arastéh, K; Rittweger, M, 2007
)
0.53
" Data originated from pre-defined pharmacokinetic substudies within two randomized 48-week trials evaluating the safety and efficacy of three protease inhibitor regimens."( Pharmacokinetics of two randomized trials evaluating the safety and efficacy of indinavir, saquinavir and lopinavir in combination with low-dose ritonavir: the MaxCmin1 and 2 trials.
Cahn, P; Castagna, A; Clumeck, N; Dragsted, UB; Fox, Z; Gerstoft, J; Justesen, US; Losso, M; Lundgren, JD; Obel, N; Pedersen, C; Peters, B, 2007
)
0.54
"Fourteen patients participated in the study and seven participated in the intensified pharmacokinetic protocol."( Effect of highly active antiretroviral therapy on tacrolimus pharmacokinetics in hepatitis C virus and HIV co-infected liver transplant recipients in the ANRS HC-08 study.
Abbara, C; Barrail, A; Boissonnas, A; Bonhomme-Faivre, L; Duclos-Vallée, JC; Samuel, D; Taburet, AM; Teicher, E; Vincent, I; Vittecoq, D, 2007
)
0.34
" Coadministration of these drugs might result in complex pharmacokinetic drug-drug interactions."( Amprenavir and lopinavir pharmacokinetics following coadministration of amprenavir or fosamprenavir with lopinavir/ritonavir, with or without efavirenz.
Barditch-Crovo, P; Carson, KA; Flexner, C; Hendrix, CW; Khan, W; Pakes, GE; Parish, M; Parsons, T; Pham, PA; Qaqish, R; Radebaugh, C, 2007
)
0.55
"37) or Cmax (10."( Amprenavir and lopinavir pharmacokinetics following coadministration of amprenavir or fosamprenavir with lopinavir/ritonavir, with or without efavirenz.
Barditch-Crovo, P; Carson, KA; Flexner, C; Hendrix, CW; Khan, W; Pakes, GE; Parish, M; Parsons, T; Pham, PA; Qaqish, R; Radebaugh, C, 2007
)
0.55
"EFV did not appear to significantly alter APV and LPV pharmacokinetic parameters in HIV-infected patients taking APV 750 mg twice daily + LPV 533/133 mg twice daily."( Amprenavir and lopinavir pharmacokinetics following coadministration of amprenavir or fosamprenavir with lopinavir/ritonavir, with or without efavirenz.
Barditch-Crovo, P; Carson, KA; Flexner, C; Hendrix, CW; Khan, W; Pakes, GE; Parish, M; Parsons, T; Pham, PA; Qaqish, R; Radebaugh, C, 2007
)
0.55
" HIV-infected subjects > or =18 to <25 years old receiving (> or =28 days) 300/100 mg atazanavir-ritonavir plus 300 mg tenofovir disoproxil fumarate (TDF) plus one or more other nucleoside analogs underwent intensive 24-h pharmacokinetic studies following a light meal."( Pharmacokinetics of antiretroviral regimens containing tenofovir disoproxil fumarate and atazanavir-ritonavir in adolescents and young adults with human immunodeficiency virus infection.
Cunningham, CK; Fletcher, CV; Flynn, PM; Harris, DR; Havens, PL; Kapogiannis, BG; Kiser, JJ; Liu, NX; Major-Wilson, H; Muenz, LR; Viani, RM; Wilson, CM, 2008
)
0.78
"An intensive steady-state 24-h pharmacokinetic profile of atazanavir was performed in the third trimester of pregnancy and postpartum."( Atazanavir plus low-dose ritonavir in pregnancy: pharmacokinetics and placental transfer.
Airoldi, M; Bertuletti, P; Cattaneo, D; Frigerio, L; Maggiolo, F; Ripamonti, D; Ruggeri, M; Suter, F, 2007
)
0.64
"96, indicating equivalence, whereas Cmax values were slightly although not significantly lower."( Atazanavir plus low-dose ritonavir in pregnancy: pharmacokinetics and placental transfer.
Airoldi, M; Bertuletti, P; Cattaneo, D; Frigerio, L; Maggiolo, F; Ripamonti, D; Ruggeri, M; Suter, F, 2007
)
0.64
"This was an open-label, crossover study to investigate the pharmacokinetic interaction between darunavir (TMC114), coadministered with low-dose ritonavir (darunavir/ritonavir), and the protease inhibitor saquinavir in HIV-negative healthy volunteers."( Pharmacokinetic interaction between darunavir and saquinavir in HIV-negative volunteers.
De Pauw, M; Hoetelmans, RM; Lefebvre, E; Mariën, K; Sekar, VJ; Vangeneugden, T, 2007
)
0.54
" A randomized, open-label, two-period, two-sequence, balanced, crossover drug interaction study was conducted with 22 healthy adult subjects to compare steady-state plasma RFB pharmacokinetic parameters during concomitant administration of FPV-RTV (700/100 mg twice a day [BID]) with a 75%-reduced RFB dose (150 mg every other day [QOD]) to the standard RFB regimen (300 mg once per day [QD]) by geometric least-squares mean ratios."( Pharmacokinetic interaction between fosamprenavir-ritonavir and rifabutin in healthy subjects.
Borland, J; Chen, YC; Ford, SL; Lou, Y; Min, SS; Shelton, MJ; Yuen, GJ, 2008
)
0.6
"This study investigated the steady-state pharmacokinetic interaction between the HIV protease inhibitor, darunavir (TMC114), administered with low-dose ritonavir (darunavir/ritonavir), and clarithromycin in HIV-negative healthy volunteers."( Darunavir/ritonavir pharmacokinetics following coadministration with clarithromycin in healthy volunteers.
De Paepe, E; De Pauw, M; Hoetelmans, RM; Lefebvre, E; Sekar, VJ; Spinosa-Guzman, S; Vangeneugden, T, 2008
)
0.95
" Intensive pharmacokinetic sampling was performed at 2 weeks and predose concentrations were collected every 8 weeks; safety and plasma HIV-1 RNA were monitored every 4-12 weeks for 24 weeks."( Pharmacokinetics, safety and efficacy of lopinavir/ritonavir in infants less than 6 months of age: 24 week results.
Capparelli, EV; Chadwick, EG; Chen, J; Hughes, M; Palumbo, P; Pinto, JA; Robbins, B; Rodman, JH; Serchuck, L; Smith, E; Yogev, R, 2008
)
0.6
"To evaluate the pharmacokinetic compatibility of a ritonavir-boosted indinavir-fosamprenavir combination among patients with human immunodeficiency virus (HIV)."( Pharmacokinetics of an indinavir-ritonavir-fosamprenavir regimen in patients with human immunodeficiency virus.
Acosta, EP; Easley, KA; Lennox, JL; Ofotokun, I; Pan, Y, 2008
)
0.88
"Single-center, nonrandomized, prospective, multiple-dose, two-phase pharmacokinetic study."( Pharmacokinetics of an indinavir-ritonavir-fosamprenavir regimen in patients with human immunodeficiency virus.
Acosta, EP; Easley, KA; Lennox, JL; Ofotokun, I; Pan, Y, 2008
)
0.63
" After 12-hour pharmacokinetic sampling was performed on all patients (period A), fosamprenavir (a prodrug of amprenavir) 700 mg twice/day was coadministered for 5 days, with a repeat 12-hour pharmacokinetic sampling performed on the fifth day (period B)."( Pharmacokinetics of an indinavir-ritonavir-fosamprenavir regimen in patients with human immunodeficiency virus.
Acosta, EP; Easley, KA; Lennox, JL; Ofotokun, I; Pan, Y, 2008
)
0.63
" With the exception of indinavir C(max), the changes in indinavir and ritonavir pharmacokinetic parameters observed after fosamprenavir coadministration were not statistically significant."( Pharmacokinetics of an indinavir-ritonavir-fosamprenavir regimen in patients with human immunodeficiency virus.
Acosta, EP; Easley, KA; Lennox, JL; Ofotokun, I; Pan, Y, 2008
)
0.86
"51 examined the pharmacokinetic profile, safety, and efficacy of RTV-boosted tipranavir (TPV/r), alone and in combination with comparator PIs (CPIs) in 315 triple-class-experienced, HIV-infected patients."( Pharmacokinetics, safety, and efficacy of tipranavir boosted with ritonavir alone or in combination with other boosted protease inhibitors as part of optimized combination antiretroviral therapy in highly treatment-experienced patients (BI Study 1182.51).
Arestéh, K; Blick, G; Johnson, M; Katlama, C; Lazzarin, A; Leith, JG; MacGregor, TR; Meier, U; Pierone, G; Walmsley, SL, 2008
)
0.58
" Lopinavir pharmacokinetic measures (median [interquartile range]) for children with and without rifampicin, respectively, were maximum concentration (Cmax) of 10."( Effect of rifampicin on lopinavir pharmacokinetics in HIV-infected children with tuberculosis.
Egbers, C; Eley, BS; Maartens, G; McIlleron, HM; Meyers, TM; Nuttall, JJ; Ren, Y; Smith, PJ, 2008
)
0.35
" Although the median Cmax and AUC0-12 were lowered by 26% and 31%."( Effect of rifampicin on lopinavir pharmacokinetics in HIV-infected children with tuberculosis.
Egbers, C; Eley, BS; Maartens, G; McIlleron, HM; Meyers, TM; Nuttall, JJ; Ren, Y; Smith, PJ, 2008
)
0.35
"The influence of nevirapine or efavirenz co-administration on ritonavir-boosted amprenavir pharmacokinetics was investigated in HIV-infected patients with a population pharmacokinetic approach."( Impact of nevirapine or efavirenz co-administration on ritonavir-boosted amprenavir pharmacokinetics in HIV-infected patients.
Allavena, C; Biron, C; Dailly, E; Jolliet, P; Raffi, F, 2008
)
0.83
" AMD070 alone had the following pharmacokinetic features, given as medians (ranges): 3 h (0."( Effect of low-dose ritonavir on the pharmacokinetics of the CXCR4 antagonist AMD070 in healthy volunteers.
Becker, S; Cao, YJ; Conley, J; Dunaway, S; Flexner, CW; Hendrix, CW; Kashuba, AD; Klingman, K; MacFarland, R; Park, JG; Radebaugh, C; Wiggins, I, 2008
)
0.67
"Studies on the pharmacokinetic interaction between atazanavir and lopinavir with ritonavir (lopinavir/ritonavir) report contradictory results."( Atazanavir and lopinavir with ritonavir alone or in combination: analysis of pharmacokinetic interaction and predictors of drug exposure.
Bacarelli, A; Cauda, R; De Luca, A; Di Giambenedetto, S; Navarra, P; Ragazzoni, E; Regazzi, M; Villani, P, 2008
)
0.86
"Seventy-seven pharmacokinetic profiles (saquinavir/ritonavir 1000/100 mg twice daily, n = 34; 1600/100 mg once daily, n = 26; 2000/100 mg once daily, n = 17) from five studies were combined, presented as twice- and once-daily percentiles (P10-P90) and compared."( Pharmacokinetic analysis to assess forgiveness of boosted saquinavir regimens for missed or late dosing.
Aarons, LJ; Back, DJ; Boffito, M; Dickinson, L; Khoo, SH; Pozniak, AL; Schutz, M, 2008
)
0.6
" However, pharmacokinetic data can only ever be a guide to the impact on long-term efficacy."( Pharmacokinetic analysis to assess forgiveness of boosted saquinavir regimens for missed or late dosing.
Aarons, LJ; Back, DJ; Boffito, M; Dickinson, L; Khoo, SH; Pozniak, AL; Schutz, M, 2008
)
0.35
" Cmax and AUC increases with gastric acid neutralization were greatest in those with the lowest absorption of DG17 alone."( Phase 1 single dose studies to optimize the pharmacokinetics of DG17, a novel HIV-protease inhibitor pro-drug, using sodium bicarbonate and ritonavir.
Cherry, CL; Hoy, JF; Krum, H; Lewin, SR; Mills, J; Rowe, JS, 2008
)
0.55
" Rats received RTV-boosted ATV or ATV-SLS SD+G for 14 d in the pharmacokinetic study."( Long-term pharmacokinetic efficacy and safety of low-dose ritonavir as a booster and atazanavir pharmaceutical formulation based on solid dispersion system in rats.
Fukushima, K; Haraya, K; Ito, Y; Sugioka, N; Takada, K; Terasaka, S, 2008
)
0.59
" Total (n = 56) and unbound (n = 36) LPV pharmacokinetic parameters were determined at steady-state using validated high-performance liquid chromatography with ultraviolet detection and high-performance liquid chromatography-tandem mass spectrometry methods, respectively."( Lopinavir/ritonavir pharmacokinetics in HIV/HCV-coinfected patients with or without cirrhosis.
Back, DJ; Cargnel, A; Cordier, L; Cusato, M; Dickinson, L; Duca, P; Khoo, SH; Meraviglia, P; Micheli, V; Orlando, G; Regazzi, M; Rizzardini, G; Viganò, P; Villani, P, 2008
)
0.75
" The objective is to study the pharmacokinetic interaction of RSV with atazanavir/ritonavir (ATV/RTV) or fosamprenavir/ritonavir (FPV/RTV)."( Effects of atazanavir/ritonavir or fosamprenavir/ritonavir on the pharmacokinetics of rosuvastatin.
Bain, AM; Bedimo, RG; Busti, AJ; Hall, RG; Leff, RD; Meek, C; Mehvar, R, 2008
)
0.89
"In a prospective pharmacokinetic drug interaction study, six HIV-seronegative, healthy adult volunteers received single 10-mg doses of RSV at baseline and after 6 days of ATV/RTV and FPV/RTV, with 6-day washout periods."( Effects of atazanavir/ritonavir or fosamprenavir/ritonavir on the pharmacokinetics of rosuvastatin.
Bain, AM; Bedimo, RG; Busti, AJ; Hall, RG; Leff, RD; Meek, C; Mehvar, R, 2008
)
0.66
"Compared to baseline, the area under the plasma concentration-time curve (AUC 0-24h) and maximum plasma concentration (Cmax) of RSV increased by 213% and 600%, respectively, and the time to reach Cmax was shorter (1."( Effects of atazanavir/ritonavir or fosamprenavir/ritonavir on the pharmacokinetics of rosuvastatin.
Bain, AM; Bedimo, RG; Busti, AJ; Hall, RG; Leff, RD; Meek, C; Mehvar, R, 2008
)
0.66
" The potential for a pharmacokinetic drug interaction exists when sildenafil and DRV/r are co-administered, as these drugs are primarily metabolized by cytochrome P450 (CYP) 3A, and darunavir and ritonavir are CYP3A inhibitors."( Effect of repeated doses of darunavir plus low-dose ritonavir on the pharmacokinetics of sildenafil in healthy male subjects: phase I randomized, open-label, two-way crossover study.
De Marez, T; De Paepe, E; De Pauw, M; Hoetelmans, RM; Lefebvre, E; Sekar, V; Vangeneugden, T, 2008
)
0.79
" Full pharmacokinetic profiles of sildenafil, N-desmethyl sildenafil, darunavir and ritonavir were determined."( Effect of repeated doses of darunavir plus low-dose ritonavir on the pharmacokinetics of sildenafil in healthy male subjects: phase I randomized, open-label, two-way crossover study.
De Marez, T; De Paepe, E; De Pauw, M; Hoetelmans, RM; Lefebvre, E; Sekar, V; Vangeneugden, T, 2008
)
0.82
" When sildenafil 25 mg was co-administered with DRV/r, the sildenafil maximum plasma concentration (Cmax) was 38% lower compared with Cmax after administration of sildenafil alone at a dose of 100 mg."( Effect of repeated doses of darunavir plus low-dose ritonavir on the pharmacokinetics of sildenafil in healthy male subjects: phase I randomized, open-label, two-way crossover study.
De Marez, T; De Paepe, E; De Pauw, M; Hoetelmans, RM; Lefebvre, E; Sekar, V; Vangeneugden, T, 2008
)
0.6
" Pharmacokinetic assessments were performed on day 14 for each session."( Pharmacokinetic interaction between ethinyl estradiol, norethindrone and darunavir with low-dose ritonavir in healthy women.
De Pauw, M; Felicione, E; Guzman, SS; Hoetelmans, RM; Lefebvre, E; Sekar, VJ; Vangeneugden, T, 2008
)
0.56
"The pharmacokinetic interaction observed here is considered to be clinically relevant as EE concentrations are considerably reduced when DRV/r is co-administered with EE and NE."( Pharmacokinetic interaction between ethinyl estradiol, norethindrone and darunavir with low-dose ritonavir in healthy women.
De Pauw, M; Felicione, E; Guzman, SS; Hoetelmans, RM; Lefebvre, E; Sekar, VJ; Vangeneugden, T, 2008
)
0.56
" Two studies evaluated pharmacokinetic (PK) interactions among EVG and RTV-boosted tipranavir (TPV/r) or darunavir (DRV/r)."( Effect of ritonavir-boosted tipranavir or darunavir on the steady-state pharmacokinetics of elvitegravir.
Enejosa, J; Hinkle, J; Kearney, BP; Mack, R; Mathias, AA; Piliero, PJ; Sekar, V; Shen, G; Tomaka, F, 2008
)
0.75
" Studies were powered to conclude lack of an interaction if the 90% confidence interval for the geometric mean ratios of the AUCtau and Cmax for EVG, TPV, and DRV were within predefined no-effect boundaries."( Effect of ritonavir-boosted tipranavir or darunavir on the steady-state pharmacokinetics of elvitegravir.
Enejosa, J; Hinkle, J; Kearney, BP; Mack, R; Mathias, AA; Piliero, PJ; Sekar, V; Shen, G; Tomaka, F, 2008
)
0.75
" On coadministration, AUCtau and Cmax of EVG and TPV and EVG and DRV were within prespecified no-effect boundaries versus treatment alone; trough concentrations were also not substantially altered."( Effect of ritonavir-boosted tipranavir or darunavir on the steady-state pharmacokinetics of elvitegravir.
Enejosa, J; Hinkle, J; Kearney, BP; Mack, R; Mathias, AA; Piliero, PJ; Sekar, V; Shen, G; Tomaka, F, 2008
)
0.75
" It is coformulated with low doses of ritonavir in order to enhance its pharmacokinetic profile."( Simultaneous population pharmacokinetic model for lopinavir and ritonavir in HIV-infected adults.
Barbanoj, MJ; Blanco, A; Clotet, B; Costa, J; Domingo, P; Miranda, C; Moltó, J; Negredo, E; Santos, JR; Valle, M, 2008
)
0.86
"To develop and validate a population pharmacokinetic model for lopinavir and ritonavir administered simultaneously in a population of HIV-infected adults."( Simultaneous population pharmacokinetic model for lopinavir and ritonavir in HIV-infected adults.
Barbanoj, MJ; Blanco, A; Clotet, B; Costa, J; Domingo, P; Miranda, C; Moltó, J; Negredo, E; Santos, JR; Valle, M, 2008
)
0.81
" First, a population pharmacokinetic model was developed for lopinavir and for ritonavir separately."( Simultaneous population pharmacokinetic model for lopinavir and ritonavir in HIV-infected adults.
Barbanoj, MJ; Blanco, A; Clotet, B; Costa, J; Domingo, P; Miranda, C; Moltó, J; Negredo, E; Santos, JR; Valle, M, 2008
)
0.81
" LPV Cmin was lower than expected in the hemodialysis group."( The pharmacokinetics and pharmacogenomics of efavirenz and lopinavir/ritonavir in HIV-infected persons requiring hemodialysis.
Amorosa, V; Cramer, YS; Gupta, SK; Hall, SD; Koletar, SL; Rosenkranz, SL; Szczech, LA, 2008
)
0.58
"The aim of this study was to develop and validate a population pharmacokinetic model in order to describe ritonavir-boosted saquinavir concentrations dosed twice and once daily in human immunodeficiency virus (HIV)-infected patients from the UK, Uganda and Thailand and to identify factors that may influence saquinavir pharmacokinetics."( Population pharmacokinetics of ritonavir-boosted saquinavir regimens in HIV-infected individuals.
Aarons, LJ; Autar, RS; Back, DJ; Boffito, M; Burger, DM; Dickinson, L; Khoo, SH; Merry, C; Mugyenyi, P; Pozniak, AL, 2008
)
0.85
" Non-linear mixed effects modelling (NONMEM version V) was applied to determine the saquinavir pharmacokinetic parameters, interindividual/interoccasion variability (IIV/IOV) and residual error."( Population pharmacokinetics of ritonavir-boosted saquinavir regimens in HIV-infected individuals.
Aarons, LJ; Autar, RS; Back, DJ; Boffito, M; Burger, DM; Dickinson, L; Khoo, SH; Merry, C; Mugyenyi, P; Pozniak, AL, 2008
)
0.63
" These cases provide a context in which to review pharmacokinetic data pertaining to the coadministration of DRV/r and an antiretroviral agent from the NNRTI class."( Pharmacokinetic interactions between ritonavir-boosted darunavir and NNRTIs: a report of 3 cases.
Rawizza, HE; Sax, PE, 2008
)
0.62
" The pharmacokinetic behavior of elvucitabine was best described by a two-compartment linear model using two absorption rates and a first-order elimination rate."( Effect of a single dose of ritonavir on the pharmacokinetic behavior of elvucitabine, a nucleoside reverse transcriptase inhibitor, administered in healthy volunteers.
Colucci, P; Ducharme, MP; Pottage, JC; Robison, H; Turgeon, J, 2009
)
0.65
"This was an open-label, three-session, pharmacokinetic trial."( Pharmacokinetics of atazanavir/ritonavir once daily and lopinavir/ritonavir twice and once daily over 72 h following drug cessation.
Back, D; Boffito, M; Else, L; Gazzard, B; Khoo, S; Moyle, G; Pozniak, A; Sousa, M; Taylor, J, 2008
)
0.63
"This study investigated the pharmacokinetic forgiveness of two boosted protease inhibitors."( Pharmacokinetics of atazanavir/ritonavir once daily and lopinavir/ritonavir twice and once daily over 72 h following drug cessation.
Back, D; Boffito, M; Else, L; Gazzard, B; Khoo, S; Moyle, G; Pozniak, A; Sousa, M; Taylor, J, 2008
)
0.63
" Pharmacokinetic analysis showed higher volume of distribution median values related to the C/C genotype in comparison with C/T or T/T genotypes for exon 26 (4."( Pharmacokinetic study of the variability of indinavir drug levels when boosted with ritonavir in HIV-infected children.
Bellusci, C; Bologna, R; Bramuglia, GF; Cáceres Guido, P; Curras, V; Hocht, C; Mangano, A; Mariño Hernández, E; Mecikovsky, D; Niselman, V; Rubio, MC; Sen, L, 2009
)
0.58
"Several dose-finding studies of boosted protease inhibitors have demonstrated that doses lower than those recommended in Caucasian populations exhibit in the Thai population similar pharmacokinetic (PK) properties with sustained virological suppression but reduced toxicity."( A low dose of ritonavir-boosted atazanavir provides adequate pharmacokinetic parameters in HIV-1-infected Thai adults.
Avihingsanon, A; Burger, DM; Chanmano, S; Cooper, DA; Gorowara, M; Kerr, SJ; Lange, J; Ohata, P; Phanuphak, P; Ruxrungtham, K; van der Lugt, J, 2009
)
0.71
" Lack of pharmacokinetic alteration bounds for 90% confidence intervals (CI) about the geometric mean ratio (GMR; coadministration versus alone) were 70-143% for elvitegravir and ritonavir pharmacokinetics (maximum concentration [C(max)], concentration at the end of the dosing interval [C(tau)] and area under the plasma concentration-time curve [AUC(tau); 0-24 h] and 80-125% for etravirine pharmacokinetics (AUC(tau) 0-12 h)."( Pharmacokinetics of elvitegravir and etravirine following coadministration of ritonavir-boosted elvitegravir and etravirine.
Kakuda, TN; Kearney, BP; Mack, R; Ramanathan, S; West, S, 2008
)
0.77
" Elvitegravir pharmacokinetic GMR was 6-7% higher following elvitegravir/r plus etravirine dosing versus elvitegravir/r."( Pharmacokinetics of elvitegravir and etravirine following coadministration of ritonavir-boosted elvitegravir and etravirine.
Kakuda, TN; Kearney, BP; Mack, R; Ramanathan, S; West, S, 2008
)
0.57
"Darunavir is a new protease inhibitor (PI) with in vitro activity against wild-type and PI-resistant HIV; it is used with the pharmacokinetic booster ritonavir."( Pharmacokinetics, efficacy, and safety of darunavir/ritonavir 800/100 mg once-daily in treatment-naïve and -experienced patients.
Boffito, M; Hill, A; Miralles, D,
)
0.58
" Statistical analyses were performed on pharmacokinetic parameters: the area under the concentration-time curve from 0 to 4 h (AUC(0-4)), the maximum concentration of the drug (C(max)), and the residual concentration of the drug at the end of the dosing interval (C(trough)) for plasma and the AUC(0-4) and C(trough) for intracellular data."( Pilot pharmacokinetic study of human immunodeficiency virus-infected patients receiving tenofovir disoproxil fumarate (TDF): investigation of systemic and intracellular interactions between TDF and abacavir, lamivudine, or lopinavir-ritonavir.
Ayen, R; Clotet, B; Grassi, J; Levi, M; Negredo, E; Pruvost, A; Puig, J; Théodoro, F, 2009
)
0.54
"The aim of this study was to develop and validate a population pharmacokinetic model to: (i) describe ritonavir-boosted atazanavir concentrations (300/100 mg once daily) and identify important covariates; and (ii) evaluate the predictive performance of the model for lower, unlicensed atazanavir doses (150 and 200 mg once daily) boosted with ritonavir (100 mg once daily)."( Population pharmacokinetics of ritonavir-boosted atazanavir in HIV-infected patients and healthy volunteers.
Aarons, L; Back, D; Boffito, M; Davies, G; Dickinson, L; Else, L; Khoo, S; Pozniak, A; Waters, L, 2009
)
0.85
"Non-linear mixed effects modelling was applied to determine atazanavir pharmacokinetic parameters, inter-individual variability (IIV) and residual error."( Population pharmacokinetics of ritonavir-boosted atazanavir in HIV-infected patients and healthy volunteers.
Aarons, L; Back, D; Boffito, M; Davies, G; Dickinson, L; Else, L; Khoo, S; Pozniak, A; Waters, L, 2009
)
0.64
"A population pharmacokinetic model for ritonavir-boosted atazanavir has been developed and validated."( Population pharmacokinetics of ritonavir-boosted atazanavir in HIV-infected patients and healthy volunteers.
Aarons, L; Back, D; Boffito, M; Davies, G; Dickinson, L; Else, L; Khoo, S; Pozniak, A; Waters, L, 2009
)
0.91
" Pharmacokinetic studies of higher dosages of rifampin are urgently needed in children to assist in placing the dosage of rifampin used in childhood on a more scientific foundation."( Rifampin pharmacokinetics in children, with and without human immunodeficiency virus infection, hospitalized for the management of severe forms of tuberculosis.
Cilliers, K; Donald, PR; Hussey, GD; Labadarios, D; Maritz, JS; McIlleron, H; Schaaf, HS; Smith, P; Willemse, M, 2009
)
0.35
" Pharmacokinetic curves were recorded for 12 h in the second trimester (week 20 +/-2), the third trimester (week 33 +/-2) and post-partum (weeks 4-6)."( The pharmacokinetics, safety and efficacy of boosted saquinavir tablets in HIV type-1-infected pregnant women.
Burger, D; Colbers, A; Hawkins, D; Koopmans, P; Molto, J; Richter, C; Ruxrungtham, K; Schutz, M; van der Ende, M; van der Lugt, J; Vogel, M; Wyen, C, 2009
)
0.35
" For artemether, trends toward Cmax and AUC decreases (Cmax 14."( Lopinavir/ritonavir affects pharmacokinetic exposure of artemether/lumefantrine in HIV-uninfected healthy volunteers.
Aweeka, FT; Charlebois, E; Dorsey, G; German, P; Hanpithakpong, W; Havlir, D; Lawrence, J; Lindegardh, N; Parikh, S; Rosenthal, PJ, 2009
)
0.76
" We performed a population pharmacokinetic analysis with NONMEM and determined that the steady-state pharmacokinetics of saquinavir in 136 HIV type 1-infected adults was modulated by a decrease in saquinavir CL following coadministration of the cytochrome P450 3A inhibitors ritonavir and atazanavir."( Cytochrome P450 3A inhibition by atazanavir and ritonavir, but not demography or drug formulation, influences saquinavir population pharmacokinetics in human immunodeficiency virus type 1-infected adults.
Lötsch, J; von Hentig, N, 2009
)
0.79
" Low bioavailability and extensive first-pass metabolism make benzimidazoles prone to pharmacokinetic drug interactions."( Effect of ritonavir on the pharmacokinetics of the benzimidazoles albendazole and mebendazole: an interaction study in healthy volunteers.
Corti, N; Heck, A; Jetter, A; Pauli-Magnus, C; Rentsch, K; Stieger, B; Zingg, W, 2009
)
0.76
"The AUC(0-24) of benzimidazoles decreased after long-term use of ritonavir, while no changes in pharmacokinetic profiles were observed under short-term administration."( Effect of ritonavir on the pharmacokinetics of the benzimidazoles albendazole and mebendazole: an interaction study in healthy volunteers.
Corti, N; Heck, A; Jetter, A; Pauli-Magnus, C; Rentsch, K; Stieger, B; Zingg, W, 2009
)
0.99
" Full pharmacokinetic profiles were analysed for lopinavir and ritonavir with a validated HPLC tandem mass spectrometry assay."( Pharmacokinetics and tolerability of once- versus twice-daily lopinavir/ritonavir treatment in HIV-1-infected children.
la Porte, C; Mitchell, CD; Parker, J; Rongkavilit, C; van Heeswijk, R; Zhang, G, 2009
)
0.82
"A phase 1, open-label, randomized, crossover, drug interaction study was conducted to assess the pharmacokinetic effects of coadministration of posaconazole (400 mg twice daily), with atazanavir (ATV) (300 mg/d alone) and with ritonavir (100 mg/d) or with efavirenz (400 mg/d) in healthy volunteers."( Effects of oral posaconazole on the pharmacokinetics of atazanavir alone and with ritonavir or with efavirenz in healthy adult volunteers.
Krishna, G; Ma, L; Martinho, M; McLeod, J; Moton, A; Seiberling, M, 2009
)
0.76
"Prospective, open-label, pharmacokinetic study in 12 HIV-infected patients stabilized on FPV/RTV 1400 mg/200 mg + tenofovir disoproxil fumarate (TDF)/emtricitabine (FTC) 300 mg/200 mg QD (TELEX II)."( Steady-state amprenavir, tenofovir, and emtricitabine pharmacokinetics before and after reducing ritonavir boosting of a fosamprenavir/tenofovir/emtricitabine regimen from 200 mg to 100 mg once daily (TELEX II).
Acosta, EP; Jennings, HC; Pakes, GE; Parks, DA; Taylor, C,
)
0.35
" Four weeks after reducing RTV, changes in Cmin and AUC24h were: APV: +26%, +0."( Steady-state amprenavir, tenofovir, and emtricitabine pharmacokinetics before and after reducing ritonavir boosting of a fosamprenavir/tenofovir/emtricitabine regimen from 200 mg to 100 mg once daily (TELEX II).
Acosta, EP; Jennings, HC; Pakes, GE; Parks, DA; Taylor, C,
)
0.35
"Subjects underwent 24-hour pharmacokinetic sampling at baseline and on day 14 of each treatment period."( Pharmacokinetics of concurrent administration of fosamprenavir and atazanavir without ritonavir in human immunodeficiency virus-negative subjects.
Anderson, PL; Clay, PG; Glaros, AG; McRae, M, 2009
)
0.58
" Ritonavir coadministration improves the pharmacokinetic (PK) profiles of concomitant PIs, and represents a cornerstone of PI-containing regimens."( Pharmacokinetic enhancers for HIV drugs.
Desai, MC; Xu, L, 2009
)
1.26
"To investigate the pharmacokinetic interaction between darunavir/ritonavir (DRV/r) and nevirapine (NVP) in 19 HIV-infected patients."( Pharmacokinetic interaction between nevirapine and darunavir with low-dose ritonavir in HIV-1-infected patients.
De Pauw, M; Hoetelmans, RM; Lefebvre, E; Mariën, K; Pozniak, A; Sekar, V; Vangeneugden, T, 2009
)
0.82
"To identify pharmacokinetic (PK) drug-drug interactions between tipranavir-ritonavir (TPV/r) and rosuvastatin and atorvastatin, we conducted two prospective, open-label, single-arm, two-period studies."( Differential effects of tipranavir plus ritonavir on atorvastatin or rosuvastatin pharmacokinetics in healthy volunteers.
Barditch-Crovo, P; Cameron, DW; Elgadi, MM; Flexner, C; Fuchs, E; la Porte, CJ; Lee, LS; Pham, PA; Piliero, PJ; Sabo, JP; van Heeswijk, R, 2009
)
0.85
" Participants received atazanavir 400 mg daily for 7 days followed by atazanavir/ritonavir 300 mg/100 mg daily for 7 days with pharmacokinetic studies on days 7 and 14."( Atazanavir pharmacokinetics in genetically determined CYP3A5 expressors versus non-expressors.
Anderson, PL; Aquilante, CL; Bushman, LR; Gardner, EM; MaWhinney, S; McDaneld, P; Predhomme, J; Ray, M; Zheng, JH, 2009
)
0.58
" Intensive pharmacokinetic (PK) sampling was done on days 10 and 24."( Lack of effect of efavirenz on the pharmacokinetics of tipranavir-ritonavir in healthy volunteers.
Béïque, L; Cameron, DW; la Porte, CJ; Sabo, JP, 2009
)
0.59
" (> or =40 kg); these gave an AUC24h, C0h and Cmax of 102, 114 and 112%, respectively, versus the corresponding mean adult pharmacokinetic parameter."( Pharmacokinetics, safety and efficacy of darunavir/ritonavir in treatment-experienced children and adolescents.
Blanche, S; Bologna, R; Cahn, P; Dierynck, I; Flynn, P; Fortuny, C; Rugina, S; Sekar, V; Spinosa-Guzman, S; van Baelen, B; Vis, P, 2009
)
0.6
" Pharmacokinetic interactions between HIV therapy and opioid dependence treatment medications can occur."( Pharmacokinetic interactions between buprenorphine/naloxone and tipranavir/ritonavir in HIV-negative subjects chronically receiving buprenorphine/naloxone.
Altice, FL; Andrews, L; Bruce, RD; Conner, C; Fang, WB; Friedland, GH; Lin, SN; Moody, DE; Piliero, PJ; Sabo, JP; Wruck, JM, 2009
)
0.58
" A 12 h pharmacokinetic study was done at 4-6 weeks after starting treatment."( Pharmacokinetics and 48 week efficacy of low-dose lopinavir/ritonavir in HIV-infected children.
Ananworanich, J; Boonrak, P; Bunupuradah, T; Burger, D; Gorowara, M; Jupimai, T; Pancharoen, C; Phasomsap, C; Puthanakit, T; Ruxrungtham, K; van der Lugt, J, 2009
)
0.6
"Low-dose lopinavir displayed adequate pharmacokinetic parameters and good efficacy as compared with standard-dose lopinavir in Thai children."( Pharmacokinetics and 48 week efficacy of low-dose lopinavir/ritonavir in HIV-infected children.
Ananworanich, J; Boonrak, P; Bunupuradah, T; Burger, D; Gorowara, M; Jupimai, T; Pancharoen, C; Phasomsap, C; Puthanakit, T; Ruxrungtham, K; van der Lugt, J, 2009
)
0.6
" Noncompartmental and population pharmacokinetic analyses (2-compartment open model) were performed."( Pharmacokinetic evaluation of rifabutin in combination with lopinavir-ritonavir in patients with HIV infection and active tuberculosis.
Ashkin, D; Boulanger, C; Espinoza, LA; Farrell, K; Graham, JJ; Hollender, E; Maasen, D; Peloquin, CA; Rivero, RO; Stambaugh, JJ; Symes, S, 2009
)
0.59
" For the five pharmacokinetic trials of lopinavir/ritonavir, a meta-analysis was used to estimate the effects of lopinavir dose versus ritonavir dose on lopinavir pharmacokinetics."( How much ritonavir is needed to boost protease inhibitors? Systematic review of 17 dose-ranging pharmacokinetic trials.
Boffito, M; Hill, A; Sawyer, W; van der Lugt, J, 2009
)
1.02
"The pharmacokinetic (PK) interaction between ritonavir-boosted elvitegravir (elvitegravir/r) and maraviroc was evaluated."( Pharmacokinetic interaction of ritonavir-boosted elvitegravir and maraviroc.
Abel, S; Hui, J; Kearney, BP; Ramanathan, S; Tweedy, S; West, S, 2010
)
0.91
" Lack of PK alteration was defined as 90% confidence intervals for ratio of geometric least squares means ratio (coadministration:alone) between 70% and 143% for elvitegravir and ritonavir Cmax (maximum concentration), Ctau (trough), and AUCtau (area under plasma concentration-time curve; 0-24 hours); for maraviroc, given a 100% increase in Cmax and AUCtau (0-12 hours); the predicted 90% confidence intervals were 162% to 247% and 136% to 295%, respectively."( Pharmacokinetic interaction of ritonavir-boosted elvitegravir and maraviroc.
Abel, S; Hui, J; Kearney, BP; Ramanathan, S; Tweedy, S; West, S, 2010
)
0.84
"Objective An open-label, three-period pharmacokinetic study was conducted to investigate the drug interaction potential between fosamprenavir (FPV) and tenofovir disoproxil fumarate (TDF)."( Steady-state amprenavir and tenofovir pharmacokinetics after coadministration of unboosted or ritonavir-boosted fosamprenavir with tenofovir disoproxil fumarate in healthy volunteers.
Andrews, M; Condoluci, DV; Luber, AD; Olson, K; Pakes, GE; Pappa, KA; Peloquin, CA; Slowinski, PD, 2010
)
0.58
"* Short-term administration of low-dose ritonavir increases area under the plasma concentration curve following oral midazolam by a factor of 28."( Inhibition of oral midazolam clearance by boosting doses of ritonavir, and by 4,4-dimethyl-benziso-(2H)-selenazine (ALT-2074), an experimental catalytic mimic of glutathione oxidase.
Berkowitz, N; Court, MH; Greenblatt, DJ; Harmatz, JS; MacNab, MW; Oleson, LE; Peters, DE; Zinny, MA, 2009
)
0.86
"We evaluated whether 'boosting' doses of ritonavir can serve as a positive control inhibitor for pharmacokinetic drug-drug interaction studies involving cytochrome P450 3A (CYP3A)."( Inhibition of oral midazolam clearance by boosting doses of ritonavir, and by 4,4-dimethyl-benziso-(2H)-selenazine (ALT-2074), an experimental catalytic mimic of glutathione oxidase.
Berkowitz, N; Court, MH; Greenblatt, DJ; Harmatz, JS; MacNab, MW; Oleson, LE; Peters, DE; Zinny, MA, 2009
)
0.86
"To determine the pharmacokinetic profiles of lopinavir and ritonavir in two newly developed generic co-formulations for HIV-infected children (Lopimune paediatric tablets and granules, 100/25 mg of lopinavir/ritonavir, Cipla Pharmaceuticals), and to compare these with the branded product (Kaletra)."( Pharmacokinetics of two generic co-formulations of lopinavir/ritonavir for HIV-infected children: a pilot study of paediatric Lopimune versus the branded product in healthy adult volunteers.
Burger, DM; Colbers, EP; de Kanter, CT; Fillekes, Q; Hoitsma, A, 2010
)
0.85
" A 32 h pharmacokinetic curve was recorded."( Pharmacokinetics of two generic co-formulations of lopinavir/ritonavir for HIV-infected children: a pilot study of paediatric Lopimune versus the branded product in healthy adult volunteers.
Burger, DM; Colbers, EP; de Kanter, CT; Fillekes, Q; Hoitsma, A, 2010
)
0.6
"Large differences in pharmacokinetic parameters can be excluded for Lopimune paediatric tablets when compared with the branded product and taken on an empty stomach, and also for Lopimune granules when these are taken with food."( Pharmacokinetics of two generic co-formulations of lopinavir/ritonavir for HIV-infected children: a pilot study of paediatric Lopimune versus the branded product in healthy adult volunteers.
Burger, DM; Colbers, EP; de Kanter, CT; Fillekes, Q; Hoitsma, A, 2010
)
0.6
"To investigate the potential for a pharmacokinetic interaction between darunavir (DRV, TMC114, Prezista), indinavir (IDV, Crixivan) and low-dose ritonavir (RTV, Norvir)."( Pharmacokinetic interaction between indinavir and darunavir with low-dose ritonavir in healthy volunteers.
De Marez, T; De Paepe, E; De Pauw, M; Hoetelmans, RM; Lefebvre, E; Sekar, V; Vangeneugden, T, 2010
)
0.79
" On day 7, full pharmacokinetic profiles of DRV, IDV and RTV were determined."( Pharmacokinetic interaction between indinavir and darunavir with low-dose ritonavir in healthy volunteers.
De Marez, T; De Paepe, E; De Pauw, M; Hoetelmans, RM; Lefebvre, E; Sekar, V; Vangeneugden, T, 2010
)
0.59
"To evaluate the pharmacokinetic interactions between ritonavir and quinine in healthy volunteers."( Pharmacokinetic interactions between ritonavir and quinine in healthy volunteers following concurrent administration.
Cook, JM; Omoruyi, SI; Onyeji, CO; Owolabi, AR; Sarma, PV; Soyinka, JO, 2010
)
0.88
"Ten healthy volunteers were each given 600-mg single oral doses of quinine alone, ritonavir alone (200 mg every 12 h for 9 days), and quinine in combination with ritonavir, in a three-period pharmacokinetic nonrandomized sequential design study."( Pharmacokinetic interactions between ritonavir and quinine in healthy volunteers following concurrent administration.
Cook, JM; Omoruyi, SI; Onyeji, CO; Owolabi, AR; Sarma, PV; Soyinka, JO, 2010
)
0.86
"01) in the elimination half-life (11."( Pharmacokinetic interactions between ritonavir and quinine in healthy volunteers following concurrent administration.
Cook, JM; Omoruyi, SI; Onyeji, CO; Owolabi, AR; Sarma, PV; Soyinka, JO, 2010
)
0.63
"We aimed to investigate the extent of pharmacokinetic drug interactions between bosentan and a fixed combination of lopinavir/ritonavir."( Mutual pharmacokinetic interactions between bosentan and lopinavir/ritonavir in healthy participants.
Dingemanse, J; Nilsson, PN; Patat, A; van Giersbergen, PL, 2010
)
0.8
"Nucleoside-sparing combination antiretroviral therapy (cART) regimens might be an attractive therapeutic option for HIV type-1 (HIV-1)-infected patients; however, the pharmacokinetic profiles of such regimens are frequently unknown."( The effects of a nucleoside-sparing antiretroviral regimen on the pharmacokinetics of ritonavir-boosted darunavir in HIV type-1-infected patients.
Back, D; Dickinson, L; Erlwein, OW; Garvey, L; Latch, N; Mackie, NE; McClure, MO; Scullard, G; Walsh, J; Winston, A, 2010
)
0.58
"Fourteen HIV-1-infected patients (age 21-55 years, 64% male) on stable cART with plasma HIV RNA <50 copies/ml entered this Phase I pharmacokinetic study."( The effects of a nucleoside-sparing antiretroviral regimen on the pharmacokinetics of ritonavir-boosted darunavir in HIV type-1-infected patients.
Back, D; Dickinson, L; Erlwein, OW; Garvey, L; Latch, N; Mackie, NE; McClure, MO; Scullard, G; Walsh, J; Winston, A, 2010
)
0.58
"This study characterized the pharmacokinetic effects, safety, and antiretroviral activity of three different doses of the nonpeptidic protease inhibitor tipranavir, in combination with ritonavir administered twice daily for 28 days, on a number of triple-combination regimens containing a nonnucleoside reverse transcriptase inhibitor (efavirenz or nevirapine) plus two nucleoside reverse transcriptase inhibitors (abacavir, didanosine, lamivudine, stavudine, and zidovudine) or a three nucleoside reverse transcriptase inhibitor combination (zidovudine, lamivudine, and abacavir)."( Pharmacokinetic characterization of three doses of tipranavir boosted with ritonavir on highly active antiretroviral therapy in treatment-experienced HIV-1 patients.
Castles, M; Goebel, FD; Johnson, PA; Legg, D; MacGregor, TR; McCallister, S; Sabo, JP,
)
0.55
" The effects of twice-daily tipranavir and ritonavir combinations on the steady-state pharmacokinetics of the antiretrovirals were assessed by comparing pharmacokinetic parameters at baseline and after 3 weeks of coadministration."( Pharmacokinetic characterization of three doses of tipranavir boosted with ritonavir on highly active antiretroviral therapy in treatment-experienced HIV-1 patients.
Castles, M; Goebel, FD; Johnson, PA; Legg, D; MacGregor, TR; McCallister, S; Sabo, JP,
)
0.62
" Six-point pharmacokinetic sampling (0, 2, 4, 6, 8, and 12 h) was undertaken after observed intake with a standardized breakfast."( Pharmacokinetics of lopinavir-ritonavir with and without nonnucleoside reverse transcriptase inhibitors in Ugandan HIV-infected adults.
Dickinson, L; Gibb, DM; Gilks, CF; Kayiwa, J; Khoo, S; Kityo, C; Lutwama, F; Munderi, P; Nalumenya, R; Reid, A; Ssali, F; Tumukunde, D; Walker, AS, 2010
)
0.65
" The aim of this study was to develop a population pharmacokinetic (PK) model and to evaluate and quantify the influence of ritonavir on the PK of docetaxel."( Population pharmacokinetics of intravenously and orally administered docetaxel with or without co-administration of ritonavir in patients with advanced cancer.
Beijnen, JH; Huitema, AD; Koolen, SL; Oostendorp, RL; Schellens, JH, 2010
)
0.78
" Drug concentrations were measured using a validated LC-MS/MS assay and pharmacokinetic analysis was performed using non-linear mixed effect modelling."( Intracellular and plasma steady-state pharmacokinetics of raltegravir, darunavir, etravirine and ritonavir in heavily pre-treated HIV-infected patients.
Beijnen, JH; Huitema, AD; Mulder, JW; Ter Heine, R; van Gorp, EC; Wagenaar, JF, 2010
)
0.58
" LPV/r pharmacokinetic parameters were calculated using noncompartmental analysis."( Pharmacokinetics and virologic response of zidovudine/lopinavir/ritonavir initiated during the third trimester of pregnancy.
Chirayus, S; Cressey, TR; Jourdain, G; Kongpanichkul, R; Lallemant, M; Ngo-Giang-Huong, N; Pattarakulwanich, S; Rawangban, B; Sabsanong, P; Varadisai, S; Voramongkol, N; Yuthavisuthi, P, 2010
)
0.6
" Geometric mean (90% confidence interval) LPV AUC, Cmax and Cmin were 64."( Pharmacokinetics and virologic response of zidovudine/lopinavir/ritonavir initiated during the third trimester of pregnancy.
Chirayus, S; Cressey, TR; Jourdain, G; Kongpanichkul, R; Lallemant, M; Ngo-Giang-Huong, N; Pattarakulwanich, S; Rawangban, B; Sabsanong, P; Varadisai, S; Voramongkol, N; Yuthavisuthi, P, 2010
)
0.6
" Full pharmacokinetic profiles were determined for DRV, ritonavir, and RFB and its active metabolite, 25-O-desacetylrifabutin (desRFB), on day 13."( Pharmacokinetics of darunavir/ritonavir and rifabutin coadministered in HIV-negative healthy volunteers.
Berckmans, C; De Pauw, M; Hoetelmans, R; Lavreys, L; Sekar, V; Spinosa-Guzman, S; Van de Casteele, T; Vangeneugden, T, 2010
)
0.9
"This study was conducted to examine the pharmacokinetic interactions between buprenorphine/naloxone (BUP/NLX) and lopinavir/ritonavir (LPV/r) in HIV-seronegative subjects chronically maintained on BUP/NLX."( Pharmacokinetic interactions between buprenorphine/naloxone and once-daily lopinavir/ritonavir.
Altice, FL; Andrews, L; Bruce, RD; Fang, WB; Friedland, GH; Lin, SN; Ma, Q; Moody, DE; Morse, GD, 2010
)
0.79
"This study was an open labeled pharmacokinetic study in twelve HIV-seronegative subjects stabilized on at least 3 weeks of BUP/NLX therapy."( Pharmacokinetic interactions between buprenorphine/naloxone and once-daily lopinavir/ritonavir.
Altice, FL; Andrews, L; Bruce, RD; Fang, WB; Friedland, GH; Lin, SN; Ma, Q; Moody, DE; Morse, GD, 2010
)
0.58
"2 ng*hr/mL) and Cmax (6."( Pharmacokinetic interactions between buprenorphine/naloxone and once-daily lopinavir/ritonavir.
Altice, FL; Andrews, L; Bruce, RD; Fang, WB; Friedland, GH; Lin, SN; Ma, Q; Moody, DE; Morse, GD, 2010
)
0.58
" Pharmacodynamic responses indicate that the altered norbuprenorphine clearance did not lead to opioid withdrawal."( Pharmacokinetic interactions between buprenorphine/naloxone and once-daily lopinavir/ritonavir.
Altice, FL; Andrews, L; Bruce, RD; Fang, WB; Friedland, GH; Lin, SN; Ma, Q; Moody, DE; Morse, GD, 2010
)
0.58
"A pharmacokinetic trial was conducted to evaluate the potential for once-daily etravirine in antiretroviral regimens without and with darunavir/ritonavir."( Pharmacokinetics of once-daily etravirine without and with once-daily darunavir/ritonavir in antiretroviral-naive HIV type-1-infected adults.
DeJesus, E; Kakuda, TN; Lalezari, JP; Osiyemi, OO; Ruane, PJ; Ryan, R; Witek, J, 2010
)
0.79
"Of 23 enrolled patients (male 87%, Caucasian 39%), pharmacokinetic profiles for etravirine were available for 21 and 20 patients on day 14 and 28, respectively."( Pharmacokinetics of once-daily etravirine without and with once-daily darunavir/ritonavir in antiretroviral-naive HIV type-1-infected adults.
DeJesus, E; Kakuda, TN; Lalezari, JP; Osiyemi, OO; Ruane, PJ; Ryan, R; Witek, J, 2010
)
0.59
" In a subgroup of patients, a complete 24 h pharmacokinetic study was performed by HPLC."( Efficacy, safety and pharmacokinetics of 900/100 mg of darunavir/ritonavir once daily in treatment-experienced patients.
Crespo, M; Curran, A; Deig, E; Domingo, P; Gutirerrez, M; Imaz, A; Lopez, RM; Mateo, G; Ocaña, I; Ribera, E, 2010
)
0.6
" Twenty-five patients were included in the pharmacokinetic study."( Efficacy, safety and pharmacokinetics of 900/100 mg of darunavir/ritonavir once daily in treatment-experienced patients.
Crespo, M; Curran, A; Deig, E; Domingo, P; Gutirerrez, M; Imaz, A; Lopez, RM; Mateo, G; Ocaña, I; Ribera, E, 2010
)
0.6
" Pharmacokinetic data were available in 577 patients randomized to receive etravirine."( Pharmacokinetics and pharmacodynamics of the non-nucleoside reverse-transcriptase inhibitor etravirine in treatment-experienced HIV-1-infected patients.
Corbett, C; De Smedt, G; Hoetelmans, RM; Kakuda, TN; Leopold, L; Nijs, S; Peeters, MP; Schöller-Gyüre, M; Snoeck, E; Vingerhoets, J; Vis, P; Wade, JR; Woodfall, BJ, 2010
)
0.36
" On day 10, a full pharmacokinetic profile was obtained for each participant."( Plasma and intracellular (peripheral blood mononuclear cells) pharmacokinetics of once-daily raltegravir (800 milligrams) in HIV-infected patients.
Back, D; Barbanoj, MJ; Cedeño, S; Clotet, B; Egan, D; Liptrott, N; Miranda, C; Moltó, J; Valle, M; Watson, V, 2011
)
0.37
" All pharmacokinetic parameters appeared to be highly variable regardless to the addition of etravirine."( Pharmacokinetics of etravirine, raltegravir and darunavir/ritonavir in treatment experienced patients.
Barrail-Tran, A; Bollens, D; Chêne, G; Colin, C; Descamps, D; Fagard, C; Goldwirt, L; Katlama, C; Molina, JM; Piketty, C; Taburet, AM; Yazdanpanah, Y, 2010
)
0.6
"The study was an open label, one-sequence cross-over pharmacokinetic study in HIV-negative adults."( Lack of a pharmacokinetic interaction between steady-state tipranavir/ritonavir and single-dose valacyclovir in healthy volunteers.
Castles, MA; Cong, XJ; Kraft, MF; MacGregor, TR; Mauss, S; Sabo, JP; Wallace, L, 2011
)
0.6
" A semiphysiological pharmacokinetic model incorporating a population pharmacokinetic analysis [nonlinear mixed-effects model (NONMEM)] was developed to analyze plasma concentration-time profiles after administration via each of the three above-mentioned routes."( A pharmacokinetic model for evaluating the impact of hepatic and intestinal first-pass loss of saquinavir in the rat.
Casabó, VG; Lledó-García, R; Merino-Sanjuán, M; Nácher, A, 2011
)
0.37
"The aim of this review is to discuss the effect of pharmacokinetic drug-drug interactions (DDIs) in the antiretroviral treatment of HIV infection."( Drug-drug interactions in the treatment of HIV infection: focus on pharmacokinetic enhancement through CYP3A inhibition.
Josephson, F, 2010
)
0.36
" It has shown promising in vitro anti-HIV-1 activities and favourable human pharmacokinetic properties after co-administration with ritonavir (RTV)."( Evaluation of steady-state pharmacokinetic interactions between ritonavir-boosted BILR 355, a non-nucleoside reverse transcriptase inhibitor, and lamivudine/zidovudine in healthy subjects.
Allen, L; Castles, MA; Huang, DB; Huang, F; MacGregor, TR; Moy, F; Nguyen, T; Robinson, P; Rowland, L; Vinisko, R, 2012
)
0.82
" Intensive blood samples were taken on days 7 and 14 for pharmacokinetic assessments."( Evaluation of steady-state pharmacokinetic interactions between ritonavir-boosted BILR 355, a non-nucleoside reverse transcriptase inhibitor, and lamivudine/zidovudine in healthy subjects.
Allen, L; Castles, MA; Huang, DB; Huang, F; MacGregor, TR; Moy, F; Nguyen, T; Robinson, P; Rowland, L; Vinisko, R, 2012
)
0.62
"After co-administration with BILR 355/r, the AUC(12,ss) and C(max,ss) of 3TC increased by 45% and 24%, respectively; the elimination half-life (t(1/2) ,ss) of 3TC was significantly increased."( Evaluation of steady-state pharmacokinetic interactions between ritonavir-boosted BILR 355, a non-nucleoside reverse transcriptase inhibitor, and lamivudine/zidovudine in healthy subjects.
Allen, L; Castles, MA; Huang, DB; Huang, F; MacGregor, TR; Moy, F; Nguyen, T; Robinson, P; Rowland, L; Vinisko, R, 2012
)
0.62
"The goal of this article is to review the ritonavir 100 mg heat-stable tablet formulation for the treatment of HIV, focusing on recent pharmacokinetic studies, safety and tolerability data, administration, and storage."( Heat-stable ritonavir tablets: a new formulation of a pharmacokinetic enhancer for HIV.
Sherman, EM; Steinberg, JG, 2011
)
1.01
" Pharmacokinetic parameters were compared using the 400/100 mg twice daily dose as reference, by determining geometric mean ratios (GMRs) and 90% confidence intervals."( Pharmacokinetics of plasma lopinavir/ritonavir following the administration of 400/100 mg, 200/150 mg and 200/50 mg twice daily in HIV-negative volunteers.
Amin, J; Back, D; Boffito, M; Else, L; Emery, S; Gazzard, B; Hill, A; Jackson, A; Khoo, S; Lin, E; Morley, R; Puls, R, 2011
)
0.64
"These pharmacokinetic data show that therapeutic plasma concentrations of lopinavir can be achieved with 200/150 mg of lopinavir/ritonavir twice daily (one Meltrex tablet and one 100 mg ritonavir capsule twice daily)."( Pharmacokinetics of plasma lopinavir/ritonavir following the administration of 400/100 mg, 200/150 mg and 200/50 mg twice daily in HIV-negative volunteers.
Amin, J; Back, D; Boffito, M; Else, L; Emery, S; Gazzard, B; Hill, A; Jackson, A; Khoo, S; Lin, E; Morley, R; Puls, R, 2011
)
0.85
" Pharmacokinetic parameters were determined in a noncompartmental analysis."( Effect of tipranavir/ritonavir combination on the pharmacokinetics of tadalafil in healthy volunteers.
Dellamonica, P; Durant, J; Ferrando, S; Garraffo, R; Lavrut, T; MacGregor, TR; Rouyrre, N; Sabo, JP, 2011
)
0.69
" This 2-step, multicenter, pharmacokinetic study enrolled human immunodeficiency virus (HIV)-infected adults on lopinavir/ritonavir (LPV/r) capsules (400/100 mg bid) plus 1 or more nucleoside reverse transcriptase inhibitors."( Sex differences in lopinavir and ritonavir pharmacokinetics among HIV-infected women and men.
Cramer, Y; Currier, JS; Fletcher, CV; Hermes, AE; Park, JG; Umeh, OC, 2011
)
0.86
"Intensive steady-state 24-hour pharmacokinetic profiles were performed during the third trimester and at 6-12 weeks postpartum."( Atazanavir pharmacokinetics with and without tenofovir during pregnancy.
Basar, M; Best, BM; Burchett, SK; Capparelli, EV; Hawkins, E; Hu, C; Mirochnick, M; Read, JS; Rossi, SS; Smith, E; Stek, AM, 2011
)
0.37
"S/GSK1349572 is an unboosted, once daily, next generation integrase inhibitor with potent activity, low pharmacokinetic (PK) variability and a novel resistance profile."( Effect of atazanavir and atazanavir/ritonavir on the pharmacokinetics of the next-generation HIV integrase inhibitor, S/GSK1349572.
Borland, J; Chen, S; Lou, Y; Min, S; Peppercorn, A; Piscitelli, SC; Song, I; Wajima, T, 2011
)
0.64
"The log-transformed data for vicriviroc primary pharmacokinetic parameters on appropriate days were statistically analysed using a one-way analysis of variance (ANOVA) model extracting the effects due to treatment."( Pharmacokinetic interaction of vicriviroc with other antiretroviral agents: results from a series of fixed-sequence and parallel-group clinical trials.
Kasserra, C; O'Mara, E, 2011
)
0.37
" Data were modeled first separately and then together by using individually predicted ritonavir pharmacokinetic parameters in the final lopinavir model."( Sequential population pharmacokinetic modeling of lopinavir and ritonavir in healthy volunteers and assessment of different dosing strategies.
Aarons, L; Back, D; Boffito, M; Davies, G; Dickinson, L; Else, L; Khoo, S; Moyle, G; Pozniak, A; von Hentig, N, 2011
)
0.83
" There were no significant differences in the LPV area under the plasma concentration-time curve from 0 to 12 h (AUC(0-12)), C(0), C(12), maximum concentration of drug in serum (C(max)), or half-life (t(1/2)) between the baseline and double-dose LPV/r time points."( Pharmacokinetics of lopinavir in HIV-infected adults receiving rifampin with adjusted doses of lopinavir-ritonavir tablets.
Decloedt, EH; Maartens, G; McIlleron, H; Merry, C; Orrell, C; Smith, P, 2011
)
0.58
" Intensive pharmacokinetic sampling occurred 7 days after starting ATV."( Atazanavir and atazanavir/ritonavir pharmacokinetics in HIV-infected infants, children, and adolescents.
Aldrovandi, G; Brundage, RC; Fenton, T; Fletcher, CV; Graham, B; Kiser, JJ; Mofenson, LM; Rutstein, RM; Samson, P; Schnittman, S; Smith, E, 2011
)
0.67
"Cohorts satisfied protocol-defined pharmacokinetic criteria if the median ATV AUC0-24hr was 60 μg × h/ml or less, and AUC0-24hr and ATV concentrations 24-h postdose (C24) were more than 30 μg × h/ml and at least 60 ng/ml, respectively, in at least 80% of the children, with no individual AUC0-24hr less than 15 μg × h/ml."( Atazanavir and atazanavir/ritonavir pharmacokinetics in HIV-infected infants, children, and adolescents.
Aldrovandi, G; Brundage, RC; Fenton, T; Fletcher, CV; Graham, B; Kiser, JJ; Mofenson, LM; Rutstein, RM; Samson, P; Schnittman, S; Smith, E, 2011
)
0.67
"Unboosted ATV capsules satisfied pharmacokinetic criteria at a dose of 520 mg/m for those aged more than 2 to 13 years or less and 620 mg/m for those aged more than 13 to 21 years or less."( Atazanavir and atazanavir/ritonavir pharmacokinetics in HIV-infected infants, children, and adolescents.
Aldrovandi, G; Brundage, RC; Fenton, T; Fletcher, CV; Graham, B; Kiser, JJ; Mofenson, LM; Rutstein, RM; Samson, P; Schnittman, S; Smith, E, 2011
)
0.67
" A total of 151 ATV plasma concentrations were obtained, and a population pharmacokinetic model was developed with NONMEM."( Population pharmacokinetics of atazanavir/ritonavir in HIV-1-infected children and adolescents.
Blanche, S; Dollfus, C; Firtion, G; Foissac, F; Hirt, D; Laurent, C; Treluyer, JM; Urien, S, 2011
)
0.63
" We report the pharmacokinetic parameters of tenofovir in combination with efavirenz, darunavir-ritonavir, or atazanavir-ritonavir in HIV-infected children."( Steady-state pharmacokinetics of tenofovir-based regimens in HIV-infected pediatric patients.
Acosta, EP; Britto, P; Carey, V; Graham, B; Hazra, R; Jean-Philippe, P; King, JR; Wiznia, A; Yogev, R, 2011
)
0.59
" Full pharmacokinetic profiles were assessed for each phase for the 72 h following day 10."( Pharmacokinetics of once-daily darunavir-ritonavir and atazanavir-ritonavir over 72 hours following drug cessation.
Amara, A; Back, D; Boffito, M; Gazzard, B; Higgs, C; Jackson, A; Khoo, S; Moyle, G; Seymour, N, 2011
)
0.64
" The lopinavir/ritonavir dose of 500/125 mg bid administered with efavirenz most closely approximates the pharmacokinetic exposure of lopinavir/ritonavir 400/100 mg bid administered alone."( Pharmacokinetics and safety of the lopinavir/ritonavir tablet 500/125 mg twice daily coadministered with efavirenz in healthy adult participants.
Awni, W; Bernstein, B; Causemaker, SJ; Chiu, YL; Klein, CE; Ng, J, 2012
)
0.99
"Significant pharmacokinetic interactions can result between acid-suppressing agents and some protease inhibitors (PIs) in the management of HIV infection."( Effects of the H2-receptor antagonist famotidine on the pharmacokinetics of atazanavir-ritonavir with or without tenofovir in HIV-infected patients.
Bertz, R; Boffito, M; Child, M; Chung, E; Kashuba, A; Mahnke, L; Patterson, K; Tebas, P; Wang, X; Wu, Y; Zhang, J; Zhu, L, 2011
)
0.59
" Each subject had 2 separate (within 30 days) steady-state 12-hour pharmacokinetic (PK) studies with crushed and whole 200/50 mg lopinavir/ritonavir tablets."( Pharmacokinetics of lopinavir/ritonavir crushed versus whole tablets in children.
Best, BM; Capparelli, EV; Diep, H; Farrell, MJ; Lee, G; Rakhmanina, N; Rossi, SS; van den Anker, JN; Williams, E, 2011
)
0.86
" Pharmacokinetic parameters, including AUC(0-24), C(max), and C(min), were calculated using noncompartmental methods, and drug interactions were evaluated using an ANOVA model by treatment group."( Effect of vicriviroc with or without ritonavir on oral contraceptive pharmacokinetics: a randomized, open-label, parallel-group, fixed-sequence crossover trial in healthy women.
Kasserra, C; Li, J; March, B; O'Mara, E, 2011
)
0.64
" Twenty-one subjects completed the study and were included in the pharmacokinetic analysis; 4 discontinued for reasons unrelated to study drug and 2 discontinued because of adverse events."( Effect of vicriviroc with or without ritonavir on oral contraceptive pharmacokinetics: a randomized, open-label, parallel-group, fixed-sequence crossover trial in healthy women.
Kasserra, C; Li, J; March, B; O'Mara, E, 2011
)
0.64
" At week 24, the geometric mean values for SQV area under the plasma concentration-time curve from 0-12 hours (AUC₀₋₁₂), the maximum observed plasma concentration (C(max)), trough plasma concentration (C(min)), and the elimination half-life (t(1/2)) were 24."( Pharmacokinetic study of saquinavir 500 mg plus ritonavir (1000/100 mg twice a day) in HIV-positive pregnant women.
Blanco, JL; Brunet, M; Calvo, M; Coll, O; Gatell, JM; Hernandez, S; Laguno, M; Larrousse, M; Lonca, M; Mallolas, J; Martin, R; Martínez, E; Martinez-Rebollar, M; Milinkovic, A; Perez, I; Soy, D, 2011
)
0.62
"HIV-infected subjects on abacavir (600 mg once daily) underwent steady-state pharmacokinetic assessments without and with darunavir/ritonavir or raltegravir."( Pharmacokinetics of abacavir and its anabolite carbovir triphosphate without and with darunavir/ritonavir or raltegravir in HIV-infected subjects.
Abongomera, G; Back, D; Boffito, M; Dickinson, L; Gazzard, B; Gedela, K; Jackson, A; Khoo, S; Moyle, G; Taylor, J, 2012
)
0.8
" This study determined the pharmacokinetic (PK) parameters of a low-dose LPV/r tablet (70% of standard dose) in HIV-infected Thai adolescents."( Low dose lopinavir/ritonavir tablet achieves adequate pharmacokinetic parameters in HIV-infected Thai adolescents.
Ananworanich, J; Burger, D; Gorowara, M; Kerr, S; Klinklom, A; Pancharoen, C; Phasomsap, C; Puthanakit, T; Ruxrungtham, K; Sriheara, C, 2012
)
0.71
" A mechanistic evaluation was undertaken using various in vitro and ex vivo studies to support the in vivo pharmacokinetic data."( Effect of grapefruit juice and ritonavir on pharmacokinetics of lopinavir in Wistar rats.
Aditya, N; Ravi, PR; Srivani, S; Thakur, R; Vats, R, 2012
)
0.66
" The pharmacokinetic analysis was performed with NONMEM."( Population pharmacokinetic analysis and pharmacogenetics of raltegravir in HIV-positive and healthy individuals.
Aouri, M; Arab-Alameddine, M; Boffito, M; Buclin, T; Cavassini, M; Csajka, C; Decosterd, LA; di Iulio, J; Fayet-Mello, A; Günthard, HF; Lubomirov, R; Neely, M; Owen, A; Rentsch, K; Rotger, M; Telenti, A, 2012
)
0.38
" Individual darunavir pharmacokinetic parameters were calculated by noncompartmental analysis and compared between days 0 and 14 by means of the geometric mean ratio (GMR) and its 90% confidence interval (CI)."( Effect of milk thistle on the pharmacokinetics of darunavir-ritonavir in HIV-infected patients.
Cedeño, S; Clotet, B; Miranda, C; Moltó, J; Negredo, E; Valle, M, 2012
)
0.62
" Pharmacokinetic (PK) and pharmacokinetic/pharmacodynamic (PK/PD) analyses were conducted using a 2-step approach: individual non-model-based PK parameters from observed sparse concentration data were determined, followed by statistical analysis of potential relationships between PK and efficacy response parameters after 48 weeks of treatment."( Pharmacokinetics and pharmacodynamics of once-daily versus twice-daily raltegravir in treatment-naive HIV-infected patients.
Campbell, H; Eron, JJ; Hang, Y; Luo, WL; Nguyen, BY; Rizk, ML; Sklar, P; Su, J; Wenning, L; Zhao, J, 2012
)
0.38
" A phase I pharmacokinetic study was conducted to assess the impact of long-term use of ritonavir-boosted lopinavir (LPV/r) on quinine pharmacokinetics in healthy volunteers."( Effects of ritonavir-boosted lopinavir on the pharmacokinetics of quinine.
El-Gasim, M; Hendrix, CW; Lu, Y; Nyunt, MM; Parsons, TL; Petty, BG, 2012
)
0.99
" Intensive pharmacokinetic sampling was performed 7 days apart after LPV/r dosing under moderate fat, high fat, and fasted meal conditions."( Steady-state pharmacokinetics of lopinavir plus ritonavir when administered under different meal conditions in HIV-infected Ugandan adults.
Back, D; Boffito, M; Byakika-Kibwika, P; Khoo, S; Lamorde, M; Mayito, J; Merry, C; Nabukeera, L; Ogwal-Okeng, J; Ryan, M; Tjia, J, 2012
)
0.63
"A pharmacokinetic (PK) study was performed in 12 HIV-negative men receiving 600 mg of darunavir, 100 mg of ritonavir, and 200 mg of etravirine orally, twice daily for 8 days."( Single- and multiple-dose pharmacokinetics of darunavir plus ritonavir and etravirine in semen and rectal tissue of HIV-negative men.
Asher Prince, HM; Brown, KC; Cohen, MS; Jennings, SH; Kashuba, AD; Malone, SA; Patterson, KB; Shaheen, NJ; Spacek, M, 2012
)
0.83
" Ritonavir, an HIV-1 protease inhibitor (PI) and potent CYP3A inhibitor, is used as a pharmacokinetic enhancer at subtherapeutic doses in combination with other HIV PIs."( Impact of low-dose ritonavir on danoprevir pharmacokinetics: results of computer-based simulations and a clinical drug-drug interaction study.
Blotner, S; Chen, Y; Fretland, J; Haznedar, JO; Reddy, MB; Smith, P; Tran, JQ, 2012
)
1.62
" The clinical results demonstrate that low-dose ritonavir enhances the pharmacokinetic profile of low-dose danoprevir such that overall danoprevir exposures can be reduced while sustaining danoprevir trough concentrations."( Impact of low-dose ritonavir on danoprevir pharmacokinetics: results of computer-based simulations and a clinical drug-drug interaction study.
Blotner, S; Chen, Y; Fretland, J; Haznedar, JO; Reddy, MB; Smith, P; Tran, JQ, 2012
)
0.96
" The primary objective was to investigate pharmacokinetic (PK) effects of lopinavir/ritonavir 400 mg/100 mg twice daily on pitavastatin 4 mg when coadministered."( Effects of steady-state lopinavir/ritonavir on the pharmacokinetics of pitavastatin in healthy adult volunteers.
Campbell, SE; Medlock, MM; Morgan, RE; Sponseller, CA; Suehira, K; Yu, CY, 2012
)
0.88
"To evaluate the pharmacokinetic profile of ritonavir-boosted lopinavir in HIV-infected patients during rifabutin-based anti-mycobacterial therapy."( Lopinavir pharmacokinetic profiles in HIV-infected patients during rifabutin-based anti-mycobacterial therapy.
Apostoli, A; Bigoni, S; Calabresi, A; Carvalho, AC; Cusato, M; Dal Zoppo, S; El-Hamad, I; Marcantoni, C; Matteelli, A; Regazzi, M; Villani, P, 2012
)
0.64
"A longitudinal, cross-over pharmacokinetic evaluation of lopinavir with and without rifabutin in HIV-infected subjects with mycobacterial disease was done."( Lopinavir pharmacokinetic profiles in HIV-infected patients during rifabutin-based anti-mycobacterial therapy.
Apostoli, A; Bigoni, S; Calabresi, A; Carvalho, AC; Cusato, M; Dal Zoppo, S; El-Hamad, I; Marcantoni, C; Matteelli, A; Regazzi, M; Villani, P, 2012
)
0.38
"In 10 patients with complete lopinavir curves at T1, T2 and T3 pharmacokinetic values were, respectively: AUC(0-12), 187."( Lopinavir pharmacokinetic profiles in HIV-infected patients during rifabutin-based anti-mycobacterial therapy.
Apostoli, A; Bigoni, S; Calabresi, A; Carvalho, AC; Cusato, M; Dal Zoppo, S; El-Hamad, I; Marcantoni, C; Matteelli, A; Regazzi, M; Villani, P, 2012
)
0.38
" These findings warrant further evaluation of their properties in an effort to identify valuable alternatives to Ritonavir as pharmacokinetic enhancers."( Benzoxazole and benzothiazole amides as novel pharmacokinetic enhancers of HIV protease inhibitors.
Baumeister, J; Haché, G; Hallenberger, S; Jonckers, TH; Rouan, MC; Sasaki, JC; Schepens, W, 2012
)
0.59
"Once-daily LPV/r monotherapy in HIV-HCV coinfected individuals offers a safe and effective approach to the management of the HIV infection, with a predictable pharmacokinetic profile."( A pilot, prospective, open-label simplification study to evaluate the safety, efficacy, and pharmacokinetics of once-daily lopinavir-ritonavir monotherapy in HIV-HCV coinfected patients: the MONOCO study.
Ackad, N; Conway, B; Cooper, C; la Porte, C; Sampalis, J; Tossonian, H,
)
0.34
"The Asian population, in general, has higher antiretroviral concentrations than those who are not Asian, but there are limited pharmacokinetic data for darunavir/ritonavir in Asian children."( Pharmacokinetics of darunavir/ritonavir in Asian HIV-1-infected children aged ≥7 years.
Ananworanich, J; Burger, D; Chokephaibulkit, K; Gorowara, M; Kerr, SJ; Pasomsap, C; Phongsamart, W; Prasitsuebsai, W; Puthanakit, T; Sekar, V; Sophonphan, J; Suwanlerk, T; Vanprapar, N; Wittawatmongkol, O, 2012
)
0.86
" The darunavir pharmacokinetic parameters were similar to those in non-Asian individuals from the DELPHI study, in which 13 of 20 with BW<40 kg used 50 or 60 mg ritonavir boosting."( Pharmacokinetics of darunavir/ritonavir in Asian HIV-1-infected children aged ≥7 years.
Ananworanich, J; Burger, D; Chokephaibulkit, K; Gorowara, M; Kerr, SJ; Pasomsap, C; Phongsamart, W; Prasitsuebsai, W; Puthanakit, T; Sekar, V; Sophonphan, J; Suwanlerk, T; Vanprapar, N; Wittawatmongkol, O, 2012
)
0.86
"The objective of this study was to develop a simultaneous population pharmacokinetic (PK) model to describe atazanavir/ritonavir (ATV/RTV) PK (300/100 mg) and to assess the effect of RTV dose reduction on ATV PK."( Simultaneous population pharmacokinetic modelling of atazanavir and ritonavir in HIV-infected adults and assessment of different dose reduction strategies.
Austin, R; Back, D; Boffito, M; Davies, G; Dickinson, L; Khoo, S; Owen, A; Schipani, A, 2013
)
0.83
"Boceprevir represents a new treatment option for hepatitis C (HCV)-infected patients, including those with HCV/human immunodeficiency virus coinfection; however, little is known about pharmacokinetic interactions between boceprevir and antiretroviral drugs."( Pharmacokinetic interactions between the hepatitis C virus protease inhibitor boceprevir and ritonavir-boosted HIV-1 protease inhibitors atazanavir, darunavir, and lopinavir.
Butterton, JR; Feng, HP; Hughes, EA; Hulskotte, EG; O'Mara, E; Treitel, MA; van Zutven, MG; Wagner, JA; Xuan, F; Youngberg, SP, 2013
)
0.61
"A randomized, open-label study to assess the pharmacokinetic interactions between boceprevir and ritonavir-boosted protease inhibitors (PI/r) was conducted in 39 healthy adults."( Pharmacokinetic interactions between the hepatitis C virus protease inhibitor boceprevir and ritonavir-boosted HIV-1 protease inhibitors atazanavir, darunavir, and lopinavir.
Butterton, JR; Feng, HP; Hughes, EA; Hulskotte, EG; O'Mara, E; Treitel, MA; van Zutven, MG; Wagner, JA; Xuan, F; Youngberg, SP, 2013
)
0.83
" Therefore, to prevent overexposure directly posttransplantation in HIV-infected patients on ritonavir-containing cART, the predictive value of a pretransplantation pharmacokinetic curve of tacrolimus was explored."( Pretransplantation pharmacokinetic curves of tacrolimus in HIV-infected patients on ritonavir-containing cART: a pilot study.
Crommelin, HA; Mudrikova, T; van den Broek, MP; van Maarseveen, EM; van Zuilen, AD, 2013
)
0.83
"A pretransplantation pharmacokinetic model of tacrolimus in these patients was developed, and a posttransplantation dosing advice was established for each individual patient."( Pretransplantation pharmacokinetic curves of tacrolimus in HIV-infected patients on ritonavir-containing cART: a pilot study.
Crommelin, HA; Mudrikova, T; van den Broek, MP; van Maarseveen, EM; van Zuilen, AD, 2013
)
0.61
" As the simulated pharmacokinetic curves lacked an absorption peak every 12 h, the mean 12 h-AUC was approximately 40 % lower compared with AUC's reported in HIV-negative recipients, when similar trough levels were targeted."( Pretransplantation pharmacokinetic curves of tacrolimus in HIV-infected patients on ritonavir-containing cART: a pilot study.
Crommelin, HA; Mudrikova, T; van den Broek, MP; van Maarseveen, EM; van Zuilen, AD, 2013
)
0.61
" However, the pharmacokinetic profiles of such regimens are often not established."( Pharmacokinetic profile and safety of 150 mg of maraviroc dosed with 800/100 mg of darunavir/ritonavir all once daily, with and without nucleoside analogues, in HIV-infected subjects.
Back, D; Croucher, A; Else, LJ; Khoo, S; Mora-Peris, B; Scullard, G; Vera, JH; Winston, A, 2013
)
0.61
" At steady-state (days 10 and 20), intensive pharmacokinetic sampling was undertaken."( Pharmacokinetic profile and safety of 150 mg of maraviroc dosed with 800/100 mg of darunavir/ritonavir all once daily, with and without nucleoside analogues, in HIV-infected subjects.
Back, D; Croucher, A; Else, LJ; Khoo, S; Mora-Peris, B; Scullard, G; Vera, JH; Winston, A, 2013
)
0.61
" The objective of this study was to develop a population pharmacokinetic model describing the pharmacokinetic differences of lopinavir and ritonavir, with and without rifampicin, between children and adults."( Model-based evaluation of the pharmacokinetic differences between adults and children for lopinavir and ritonavir in combination with rifampicin.
Decloedt, EH; Denti, P; Karlsson, MO; McIlleron, H; Ren, Y; Zhang, C, 2013
)
0.81
"An integrated population pharmacokinetic model developed in nonmem 7 was used to describe the pharmacokinetics of lopinavir and ritonavir in 21 HIV infected adults, 39 HIV infected children and 35 HIV infected children with tuberculosis, who were established on lopinavir/ritonavir-based antiretroviral therapy with and without rifampicin-containing antituberculosis therapy."( Model-based evaluation of the pharmacokinetic differences between adults and children for lopinavir and ritonavir in combination with rifampicin.
Decloedt, EH; Denti, P; Karlsson, MO; McIlleron, H; Ren, Y; Zhang, C, 2013
)
0.81
" In children, the absorption half-life of lopinavir and the mean transit time of ritonavir were lengthened, compared with those in adults."( Model-based evaluation of the pharmacokinetic differences between adults and children for lopinavir and ritonavir in combination with rifampicin.
Decloedt, EH; Denti, P; Karlsson, MO; McIlleron, H; Ren, Y; Zhang, C, 2013
)
0.83
" Noncompartmental pharmacokinetic analysis was used to estimate PK parameters [area under the concentration-time curve over 24 h (AUC0-24h ) and maximal concentration (Cmax )]."( Pharmacokinetics of two common antiretroviral regimens in older HIV-infected patients: a pilot study.
Adams, JL; Corbett, AH; Dumond, JB; Forrest, A; Jennings, SH; Kashuba, AD; Kendrick, RL; Malone, S; Patterson, KB; Prince, HM; Sykes, C; Wang, R; White, N, 2013
)
0.39
" Compared with the general population, these elderly subjects had 8-13% decreased TFV AUC0-24h and Cmax , and 19-78% increased FTC and RTV AUC0-24h and Cmax ."( Pharmacokinetics of two common antiretroviral regimens in older HIV-infected patients: a pilot study.
Adams, JL; Corbett, AH; Dumond, JB; Forrest, A; Jennings, SH; Kashuba, AD; Kendrick, RL; Malone, S; Patterson, KB; Prince, HM; Sykes, C; Wang, R; White, N, 2013
)
0.39
" The current study evaluated the pharmacokinetic interaction between etravirine and the lopinavir/ritonavir melt extrusion formulation."( Steady-state pharmacokinetics of etravirine and lopinavir/ritonavir melt extrusion formulation, alone and in combination, in healthy HIV-negative volunteers.
Akuma, SH; De Smedt, G; Hoetelmans, RM; Kakuda, TN; Schöller-Gyüre, M; Spittaels, K; Vyncke, V; Witek, J, 2013
)
0.85
" Steady-state pharmacokinetics were assessed for all antiretrovirals alone and coadministered; pharmacokinetic parameters were obtained by noncompartmental analysis."( Steady-state pharmacokinetics of etravirine and lopinavir/ritonavir melt extrusion formulation, alone and in combination, in healthy HIV-negative volunteers.
Akuma, SH; De Smedt, G; Hoetelmans, RM; Kakuda, TN; Schöller-Gyüre, M; Spittaels, K; Vyncke, V; Witek, J, 2013
)
0.63
" The objective was to investigate pharmacokinetic interactions between darunavir/ritonavir or etravirine and arthemether/lumefrantrine."( Pharmacokinetic interaction between etravirine or darunavir/ritonavir and artemether/lumefantrine in healthy volunteers: a two-panel, two-way, two-period, randomized trial.
DeMasi, R; Kakuda, TN; Mohammed, P; van Delft, Y, 2013
)
0.86
"To investigate the pharmacokinetic and pharmacodynamic relationships of the human immunodeficiency virus (HIV)-protease inhibitor atazanavir (ATV) in the presence and absence of the pharmacokinetic booster ritonavir, utilizing ATV plasma trough concentrations (Ctrough ) and clinical biomarkers of antiviral efficacy and safety over 48 weeks."( Pharmacokinetics and pharmacodynamics of atazanavir-containing antiretroviral regimens, with or without ritonavir, in patients who are HIV-positive and treatment-naïve.
Bertz, RJ; Chung, E; Grasela, D; McGrath, D; Persson, A; Zhang, J; Zhu, L, 2013
)
0.79
" Pharmacokinetics and drug-drug interaction were simulated using the full physiologically based pharmacokinetic model implemented in the Simcyp™ ADME simulator."( Prediction of drug-drug interactions between various antidepressants and efavirenz or boosted protease inhibitors using a physiologically based pharmacokinetic modelling approach.
Almond, L; Back, D; Khoo, S; Kirov, A; Marzolini, C; Owen, A; Seden, K; Siccardi, M, 2013
)
0.39
" Although the simulated drug-drug interactions with antidepressants were overall weak to moderate according to the classification of the US FDA, fluoxetine and venlafaxine represent better candidates from a pharmacokinetic standpoint for patients on efavirenz and venlafaxine or citalopram for patients on boosted protease inhibitors."( Prediction of drug-drug interactions between various antidepressants and efavirenz or boosted protease inhibitors using a physiologically based pharmacokinetic modelling approach.
Almond, L; Back, D; Khoo, S; Kirov, A; Marzolini, C; Owen, A; Seden, K; Siccardi, M, 2013
)
0.39
" To enhance its pharmacokinetic profile, darunavir must be co-administered with ritonavir."( Simultaneous pharmacogenetics-based population pharmacokinetic analysis of darunavir and ritonavir in HIV-infected patients.
Cedeño, S; Clotet, B; Miranda, C; Moltó, J; Owen, A; Pushpakom, S; Valle, M; Xinarianos, G, 2013
)
0.84
"The aim of this study was to develop a semi-mechanistic population pharmacokinetic model for darunavir and ritonavir administered in HIV-infected adults."( Simultaneous pharmacogenetics-based population pharmacokinetic analysis of darunavir and ritonavir in HIV-infected patients.
Cedeño, S; Clotet, B; Miranda, C; Moltó, J; Owen, A; Pushpakom, S; Valle, M; Xinarianos, G, 2013
)
0.82
"A population pharmacokinetic analysis was performed with 705 plasma samples from 75 Caucasian individuals receiving darunavir/ritonavir (600/100 mg twice daily) for at least 4 weeks."( Simultaneous pharmacogenetics-based population pharmacokinetic analysis of darunavir and ritonavir in HIV-infected patients.
Cedeño, S; Clotet, B; Miranda, C; Moltó, J; Owen, A; Pushpakom, S; Valle, M; Xinarianos, G, 2013
)
0.82
"A population pharmacokinetic model to simultaneously describe the pharmacokinetics of darunavir and ritonavir was developed in HIV-infected patients."( Simultaneous pharmacogenetics-based population pharmacokinetic analysis of darunavir and ritonavir in HIV-infected patients.
Cedeño, S; Clotet, B; Miranda, C; Moltó, J; Owen, A; Pushpakom, S; Valle, M; Xinarianos, G, 2013
)
0.83
"Open-label, prospective, randomized pharmacokinetic and pharmacodynamic study in healthy volunteers."( Influence of low-dose ritonavir with and without darunavir on the pharmacokinetics and pharmacodynamics of inhaled beclomethasone.
Alfaro, RM; Boyd, SD; Calderon, MM; Chairez, C; Hadigan, C; Kovacs, JA; McManus, M; Nieman, LK; Pau, AK; Penzak, SR, 2013
)
0.7
" Pharmacokinetic sampling for 17-BMP was performed on days 14 and 28, and pharmacokinetic parameter values were compared within patients and between groups."( Influence of low-dose ritonavir with and without darunavir on the pharmacokinetics and pharmacodynamics of inhaled beclomethasone.
Alfaro, RM; Boyd, SD; Calderon, MM; Chairez, C; Hadigan, C; Kovacs, JA; McManus, M; Nieman, LK; Pau, AK; Penzak, SR, 2013
)
0.7
"The aim of this study was to investigate the pharmacokinetic interaction between ritonavir (RTV) and an anti-HIV agent 3-cyanomethyl-4-methyl-DCK (CMDCK)."( Investigation of the pharmacokinetic interaction between ritonavir and CMDCK, a new non-nucleoside reverse transcriptase inhibitor.
Li, H; Shen, GL; Yuan, M; Zhuang, XM, 2013
)
0.86
" In this paper, we review and unite pharmacokinetic data, case reports and current research regarding this drug-drug interaction in order to suggest options for the clinical management of HIV-positive patients requiring treatment with protease inhibitors and inhaled or intranasal corticosteroids."( Inhaled corticosteroid use in HIV-positive individuals taking protease inhibitors: a review of pharmacokinetics, case reports and clinical management.
Nguyen, DP; Phengrasamy, T; Saberi, P, 2013
)
0.39
" Although there were no significant changes in conjugated bilirubin, unconjugated bilirubin Cmax and AUC(0-24) of were lower (17 and 19%, phase 2; 20 and 23% during phase 3)."( Coadministration of atazanavir-ritonavir and zinc sulfate: impact on hyperbilirubinemia and pharmacokinetics.
Back, D; Boffito, M; Else, L; Gazzard, B; Jackson, A; Karolia, Z; Moyle, G; Ringner-Nackter, L; Seymour, N; Yapa, MH, 2013
)
0.68
" Blood samples were collected serially with each dose for pharmacokinetic assessment."( Pharmacokinetics of a three-way drug interaction between danoprevir, ritonavir and the organic anion transporting polypeptide (OATP) inhibitor ciclosporin.
Asthappan, J; Brennan, BJ; Goelzer, P; Morcos, PN; Moreira, SA; Navarro, MT; Smith, PF; Weigl, P, 2013
)
0.62
" A pharmacokinetic interaction between tenofovir and ritonavir may have resulted in the toxicity."( Tenofovir induced Fanconi syndrome: a possible pharmacokinetic interaction.
Desai, C; Desai, M; Dikshit, RK; Kapadia, J; Patel, S; Shah, AN; Shah, S,
)
0.38
" Pharmacokinetic parameters were derived using noncompartmental analysis."( Total and unbound darunavir pharmacokinetics in pregnant women infected with HIV-1: results of a study of darunavir/ritonavir 600/100 mg administered twice daily.
Baugh, B; Brown, K; Coate, B; Falcon, R; Kakuda, TN; Mrus, J; Osiyemi, OO; Verboven, P; Wright, R; Yasin, S; Zorrilla, CD, 2014
)
0.61
"To address the need for nucleos(t)ide reverse transcriptase inhibitor (NRTI)-sparing regimens, we explored the virologic and pharmacokinetic characteristics of maraviroc plus ritonavir-boosted darunavir in a single-arm, open-label, 96-week study."( Virologic response, early HIV-1 decay, and maraviroc pharmacokinetics with the nucleos(t)ide-free regimen of maraviroc plus darunavir/ritonavir in a pilot study.
Acosta, EP; Adeyemi, O; Berzins, B; Castro, J; Eron, JJ; Kuritzkes, DR; Lalezari, J; Lu, D; Ryscavage, P; Swindells, S; Taiwo, B; Tsibris, A, 2013
)
0.79
"Results from intensive pharmacokinetic sampling of subjects enrolled in single dose visits was used to determine individualized dosing for the first 6-10 subjects in each of 2 cohorts (4 weeks to <6 months, 6 months to <2 years); steady state pharmacokinetic data were then used to select the dosage regimen for the remaining subjects recruited to the cohort."( Pharmacokinetics, safety and antiviral activity of fosamprenavir/ritonavir-containing regimens in HIV-infected children aged 4 weeks to 2 years-48-week study data.
Cassim, H; Cheng, K; Cotton, M; Ford, SL; Garges, HP; Givens, N; Lou, Y; Pavía-Ruz, N; Perger, T; Ross, LL; Sievers, J; Wire, MB, 2014
)
0.64
" FPV 45 mg/kg boosted with RTV 7 to 10 mg/kg BID achieved average plasma amprenavir area under curve(0-τ) values 26% to 28% lower and Cmax similar to historical adult data for FPV/RTV 700/100 mg BID; amprenavir Cτ values were lower in the subjects <6 months of age."( Pharmacokinetics, safety and antiviral activity of fosamprenavir/ritonavir-containing regimens in HIV-infected children aged 4 weeks to 2 years-48-week study data.
Cassim, H; Cheng, K; Cotton, M; Ford, SL; Garges, HP; Givens, N; Lou, Y; Pavía-Ruz, N; Perger, T; Ross, LL; Sievers, J; Wire, MB, 2014
)
0.64
"Serial pharmacokinetic samples were collected at week 2 and predose samples every 4-12 weeks."( Pharmacokinetics and 48-week safety and antiviral activity of fosamprenavir-containing regimens in HIV-infected 2- to 18-year-old children.
Cheng, K; Cotton, M; Duiculescu, D; Ford, SL; Fortuny, C; Garges, HP; Givens, N; Lou, Y; Perger, T; Ross, LL; Sievers, J; Tamarirt, DP; Wire, MB, 2014
)
0.4
" However, data on the pharmacokinetic (PK) profiles of these regimens are limited."( Pharmacokinetic study of dual therapy with raltegravir 400 mg twice daily and Darunavir/Ritonavir 800/100 mg once daily in HIV-1-infected patients.
Blanco, JL; Brunet, M; Calvo, M; Gatell, JM; González, A; Hidalgo, S; Laguno, M; Loncà, M; Mallolas, J; Martínez, E; Martínez-Rebollar, M; Muñoz, A; Pérez, I, 2013
)
0.61
" The geometric mean values for DRV were AUC0-24 68,730 ng·h·mL [95% confidence interval (CI): 58,970-86,480], Ctrough 1330 ng/mL (95% CI: 1110-1760; IC-50 for wild-type and resistant HIV-1 strains was 55 and 550 ng/mL, respectively), Cmax 7630 ng/mL (95% CI: 6740-9000), and t1/2 10."( Pharmacokinetic study of dual therapy with raltegravir 400 mg twice daily and Darunavir/Ritonavir 800/100 mg once daily in HIV-1-infected patients.
Blanco, JL; Brunet, M; Calvo, M; Gatell, JM; González, A; Hidalgo, S; Laguno, M; Loncà, M; Mallolas, J; Martínez, E; Martínez-Rebollar, M; Muñoz, A; Pérez, I, 2013
)
0.61
" The aim of this study was to investigate the potential pharmacokinetic interactions between Rh2 and the HIV protease inhibitor ritonavir."( Pharmacokinetic interactions between 20(S)-ginsenoside Rh2 and the HIV protease inhibitor ritonavir in vitro and in vivo.
Aa, JY; Cao, B; Ge, C; Gu, RR; Li, MJ; Liu, CX; Liu, LS; Ma, T; Mao, Y; Shi, J; Sun, RB; Wang, GJ; Wang, XW; Wu, XL; Xia, WJ; Xiao, WJ; Yu, XY; Zha, WB; Zheng, T; Zhou, J, 2013
)
0.82
"The mean Cmax and AUC0-24 of rifabutin in patients on rifabutin 150 mg every other day were 36% and 26% lower than on 300 mg/day rifabutin, while the mean Cmax and AUC0-24 of 25-O-desacetyl rifabutin were 186% and 152% higher, respectively."( Pharmacokinetics of rifabutin in Japanese HIV-infected patients with or without antiretroviral therapy.
Aoki, T; Gatanaga, H; Honda, H; Kikuchi, Y; Oka, S; Sano, K; Tanuma, J; Teruya, K; Tsukada, K; Watanabe, K; Yazaki, H, 2013
)
0.39
"Open-label, two-period, single-sequence pharmacokinetic study."( Effect of ritonavir-boosted danoprevir, a potent hepatitis C virus protease inhibitor, on the pharmacokinetics of methadone in healthy subjects undergoing methadone maintenance therapy.
Bech, N; Brennan, BJ; Morcos, PN; Moreira, SA; Navarro, MT; Smith, PF, 2014
)
0.8
" Pharmacokinetic parameters for the total concentrations of (R)- and (S)-methadone on Days -1 and 10 were determined using noncompartmental methods."( Effect of ritonavir-boosted danoprevir, a potent hepatitis C virus protease inhibitor, on the pharmacokinetics of methadone in healthy subjects undergoing methadone maintenance therapy.
Bech, N; Brennan, BJ; Morcos, PN; Moreira, SA; Navarro, MT; Smith, PF, 2014
)
0.8
"Coadministration of DNVr with MMT resulted in no significant pharmacokinetic interactions or signs of methadone withdrawal."( Effect of ritonavir-boosted danoprevir, a potent hepatitis C virus protease inhibitor, on the pharmacokinetics of methadone in healthy subjects undergoing methadone maintenance therapy.
Bech, N; Brennan, BJ; Morcos, PN; Moreira, SA; Navarro, MT; Smith, PF, 2014
)
0.8
"The pharmacokinetic data support coadministration of daclatasvir with atazanavir/ritonavir, efavirenz and/or tenofovir."( Assessment of pharmacokinetic interactions of the HCV NS5A replication complex inhibitor daclatasvir with antiretroviral agents: ritonavir-boosted atazanavir, efavirenz and tenofovir.
Bertz, R; Bifano, M; Grasela, D; Hartstra, J; Hwang, C; Kandoussi, H; Oosterhuis, B; Sevinsky, H; Tiessen, R; Velinova-Donga, M, 2013
)
0.82
"This open-label, fixed-sequence, phase 1 study evaluated the pharmacokinetic interaction between maraviroc (MVC) and ritonavir-boosted fosamprenavir (FPV/r) in healthy subjects."( Pharmacokinetic interaction between maraviroc and fosamprenavir-ritonavir: an open-label, fixed-sequence study in healthy subjects.
Fang, A; Heera, J; Mendes da Costa, L; Plotka, A; Vourvahis, M, 2013
)
0.84
" DNV AUC0-∞, Cmax and C12h GMR% (90% CI) with ranitidine: 81."( Effect of meal and antisecretory agents on the pharmacokinetics of danoprevir/ritonavir in healthy volunteers.
Bech, N; Brennan, BJ; Morcos, PN; Moreira, SA; Navarro, MT; Quatkemeyer, A; Smith, PF, 2014
)
0.63
" The mean Cmax and area under plasma concentration-time curve values (single-dose: 1200 mg TMC310911) were higher under fasted conditions than in fed condition."( Safety and pharmacokinetics of the HIV-1 protease inhibitor TMC310911 coadministered with ritonavir in healthy participants: results from 2 phase 1 studies.
Dierynck, I; Hoetelmans, RM; Jacquemyn, B; Marien, K; Meyvisch, P; Simmen, K; Smyej, I; Verloes, R, 2014
)
0.62
"TMC310911 exhibited a linear pharmacokinetic profile after the single- (up to 2000 mg) and multiple-dose (up to 900 mg) administrations; ritonavir improved the pharmacokinetic profile of TMC310911."( Safety and pharmacokinetics of the HIV-1 protease inhibitor TMC310911 coadministered with ritonavir in healthy participants: results from 2 phase 1 studies.
Dierynck, I; Hoetelmans, RM; Jacquemyn, B; Marien, K; Meyvisch, P; Simmen, K; Smyej, I; Verloes, R, 2014
)
0.83
"Intensive pharmacokinetic studies of tenofovir in a large, diverse cohort of HIV-infected women over 24 h at steady state were performed and factors that influenced exposure [assessed by areas under the concentration-time curves (AUCs)] identified."( Common clinical conditions - age, low BMI, ritonavir use, mild renal impairment - affect tenofovir pharmacokinetics in a large cohort of HIV-infected women.
Anastos, K; Bacchetti, P; Baxi, SM; Cohen, M; Gandhi, M; Gange, SJ; Greenblatt, RM; Huang, Y; Minkoff, H; Scherzer, R; Shlipak, MG; Young, M, 2014
)
0.67
"HIV-infected women (n = 101) on tenofovir-based therapy underwent intensive 24-h pharmacokinetic sampling."( Common clinical conditions - age, low BMI, ritonavir use, mild renal impairment - affect tenofovir pharmacokinetics in a large cohort of HIV-infected women.
Anastos, K; Bacchetti, P; Baxi, SM; Cohen, M; Gandhi, M; Gange, SJ; Greenblatt, RM; Huang, Y; Minkoff, H; Scherzer, R; Shlipak, MG; Young, M, 2014
)
0.67
" The primary objective was to develop an integrated LPV population pharmacokinetic model to investigate the influence of α-1-acid glycoprotein and link total and free LPV exposure to pharmacodynamic changes in HIV-1 RNA and assess viral dynamic and drug efficacy parameters."( Integrated population pharmacokinetic/viral dynamic modelling of lopinavir/ritonavir in HIV-1 treatment-naïve patients.
Acosta, EP; D'Argenio, DZ; Delille, C; Kerstner-Wood, C; Lennox, JL; Ofotokun, I; Sheth, AN; Wang, K, 2014
)
0.63
" We used a pharmacokinetic model developed from published data to simulate the effect of sample patient-controlled epidural labor analgesic regimens on plasma fentanyl concentrations in the absence and presence of ritonavir-induced cytochrome P450 3A4 inhibition."( Effect of ritonavir-induced cytochrome P450 3A4 inhibition on plasma fentanyl concentrations during patient-controlled epidural labor analgesia: a pharmacokinetic simulation.
Avram, MJ; Cambic, CR; Gupta, DK; Wong, CA, 2014
)
0.99
" Systemic fentanyl disposition was described using a three-compartment pharmacokinetic model."( Effect of ritonavir-induced cytochrome P450 3A4 inhibition on plasma fentanyl concentrations during patient-controlled epidural labor analgesia: a pharmacokinetic simulation.
Avram, MJ; Cambic, CR; Gupta, DK; Wong, CA, 2014
)
0.8
"For the total group, LPV geometric mean AUC0-12, Cmax and C12 were 106."( Pharmacokinetics of pediatric lopinavir/ritonavir tablets in children when administered twice daily according to FDA weight bands.
Bastiaans, DE; Burger, DM; Chalermpantmetagul, S; Colbers, AP; Compagnucci, A; Cressey, TR; Forcat, S; Giaquinto, C; Hansudewechakul, R; Harper, LM; Inshaw, JR; Kanjanavanit, S; Königs, C; Lyall, H; Noguera-Julian, A; Saïdi, Y, 2014
)
0.67
"Sixteen and seventeen patients were respectively randomized to the two arms, and pharmacokinetic analysis carried out in 12 and 13 respectively."( Randomised pharmacokinetic trial of rifabutin with lopinavir/ritonavir-antiretroviral therapy in patients with HIV-associated tuberculosis in Vietnam.
Barrail-Tran, A; Borand, L; Connolly, C; Duc, NH; Dung, NH; Harries, AD; Lagarde, D; Lan, NN; Lan, NT; Laureillard, D; Lien, TT; Lienhardt, C; Pym, A; Quillet, C; Taburet, AM; Thu, NT, 2014
)
0.64
" Use of ritonavir as a pharmacokinetic enhancer may have abrogated genetic associations with atazanavir pharmacokinetics."( Genomewide association study of atazanavir pharmacokinetics and hyperbilirubinemia in AIDS Clinical Trials Group protocol A5202.
Daar, ES; Haas, DW; Johnson, DH; McLaren, PJ; Morse, GD; Ritchie, MD; Venuto, C, 2014
)
0.84
"Steady-state 12-hour pharmacokinetic profiles of lopinavir/ritonavir were assessed in patients with (group 1, n = 20) or without (group 2, n = 36) hepatitis coinfection, taking lopinavir/ritonavir 400/100 mg twice a day plus nucleoside reverse transcriptase inhibitors, measured by means of high-performance liquid chromatography-tandem mass spectrometry."( Lopinavir/ritonavir pharmacokinetics, efficacy, and safety in HIV and hepatitis B or C coinfected adults without symptoms of hepatic impairment.
Bickel, M; Brodt, R; Haberl, A; Khaykin, P; Kotzerke, P; Kurowski, M; Nisius, G; Stephan, C; Stürmer, M; von Hentig, N, 2014
)
1.05
" Darunavir AUC24h following administration of the FDCs (G003: 74,780 ng ∙ h/mL and G004: 76,490 ng ∙ h/mL) was comparable to that following darunavir/ritonavir (78,410 ng ∙ h/mL), as was Cmax (6,666 and 6,917 ng/mL versus 6,973 ng/mL, respectively)."( Pharmacokinetics of darunavir in fixed-dose combination with cobicistat compared with coadministration of darunavir and ritonavir as single agents in healthy volunteers.
Hillewaert, V; Hoetelmans, RM; Iterbeke, K; Kakuda, TN; Opsomer, M; Petrovic, R; Timmers, M; Van De Casteele, T, 2014
)
0.81
" Comparative oral pharmacokinetic studies and tissue distribution studies of optimized SLNs and LPV/RTV coformulation were done in Wistar rats."( A hybrid design to optimize preparation of lopinavir loaded solid lipid nanoparticles and comparative pharmacokinetic evaluation with marketed lopinavir/ritonavir coformulation.
Dalal, V; Murthy, AN; Ravi, PR; Vats, R, 2014
)
0.6
" We evaluated the effect of 2 highly active antiretroviral therapy (HAART) regimens (1 including efavirenz and the other ritonavir-boosted lopinavir) on the pharmacokinetic (PK) parameters of an ENG-releasing implant in HIV-positive women."( Effect of antiretroviral therapy including lopinavir/ritonavir or efavirenz on etonogestrel-releasing implant pharmacokinetics in HIV-positive women.
Amaral, E; Bahamondes, L; Bahamondes, MV; Brito, MB; de Souza, RM; Duarte, G; Ferriani, RA; Quintana, SM; Rocha Prandini, TR; Scaranari, C; Vieira, CS, 2014
)
0.86
" A population pharmacokinetic approach was used to characterize the pharmacokinetics of both drugs and their interaction in a cohort of unselected patients and to compare darunavir exposure expected under alternative dosage regimens."( Population pharmacokinetic modelling and evaluation of different dosage regimens for darunavir and ritonavir in HIV-infected individuals.
Aouri, M; Arab-Alameddine, M; Buclin, T; Cavassini, M; Csajka, C; Décosterd, LA; Fayet-Mello, A; Gatri, M; Guidi, M; Ledergerber, B; Lubomirov, R; Panchaud, A; Rentsch, K; Rotger, M; Telenti, A; Widmer, N, 2014
)
0.62
" Firstly, a population pharmacokinetic analysis for darunavir and ritonavir was conducted, with inclusion of patients' demographic, clinical and genetic characteristics as potential covariates (NONMEM(®))."( Population pharmacokinetic modelling and evaluation of different dosage regimens for darunavir and ritonavir in HIV-infected individuals.
Aouri, M; Arab-Alameddine, M; Buclin, T; Cavassini, M; Csajka, C; Décosterd, LA; Fayet-Mello, A; Gatri, M; Guidi, M; Ledergerber, B; Lubomirov, R; Panchaud, A; Rentsch, K; Rotger, M; Telenti, A; Widmer, N, 2014
)
0.86
" The simulations predicted darunavir Cmin much higher than the IC50 thresholds for wild-type and protease inhibitor-resistant HIV-1 strains (55 and 550 ng/mL, respectively) under standard dosing in >98% of experienced and naive patients."( Population pharmacokinetic modelling and evaluation of different dosage regimens for darunavir and ritonavir in HIV-infected individuals.
Aouri, M; Arab-Alameddine, M; Buclin, T; Cavassini, M; Csajka, C; Décosterd, LA; Fayet-Mello, A; Gatri, M; Guidi, M; Ledergerber, B; Lubomirov, R; Panchaud, A; Rentsch, K; Rotger, M; Telenti, A; Widmer, N, 2014
)
0.62
" Pharmacokinetic blood sampling was performed on days 5, 11 and 16."( Steady-state pharmacokinetics of darunavir/ritonavir and pitavastatin when co-administered to healthy adult volunteers.
Campbell, SE; Medlock, MM; Morgan, RE; Small, DS; Sponseller, CA; Yu, CY, 2014
)
0.67
"5 years) were enrolled, and pharmacokinetic data were available for 27 subjects."( Steady-state pharmacokinetics of darunavir/ritonavir and pitavastatin when co-administered to healthy adult volunteers.
Campbell, SE; Medlock, MM; Morgan, RE; Small, DS; Sponseller, CA; Yu, CY, 2014
)
0.67
" Twelve-hour intensive pharmacokinetic sampling after observed LPV/r intake (plus 2 nucleoside reverse transcriptase inhibitors) following World Health Organization 2010 dosing with food was performed 4 weeks after enrollment."( The pharmacokinetics and acceptability of lopinavir/ritonavir minitab sprinkles, tablets, and syrups in african HIV-infected children.
Burger, D; Fillekes, Q; Gibb, DM; Keishanyu, R; Kekitiinwa, A; Kendall, L; Lallemant, M; Musiime, V; Namuddu, R; Opilo, W; Walker, AS; Young, N, 2014
)
0.65
" Among 132 evaluable pharmacokinetic profiles, there were 13/21/25 within-child comparisons in cohort A/B/C."( The pharmacokinetics and acceptability of lopinavir/ritonavir minitab sprinkles, tablets, and syrups in african HIV-infected children.
Burger, D; Fillekes, Q; Gibb, DM; Keishanyu, R; Kekitiinwa, A; Kendall, L; Lallemant, M; Musiime, V; Namuddu, R; Opilo, W; Walker, AS; Young, N, 2014
)
0.65
" Pharmacokinetic sampling occurred at the end of each dosing period."( Phase I safety, pharmacokinetics, and pharmacogenetics study of the antituberculosis drug PA-824 with concomitant lopinavir-ritonavir, efavirenz, or rifampin.
Allen, R; Aweeka, F; Bao, J; Cramer, Y; Dooley, KE; Haas, DW; Koletar, SL; Luetkemeyer, AF; Marzan, F; Murray, S; Park, JG; Savic, R; Sutherland, D, 2014
)
0.61
" The authors studied ATV pharmacokinetic (PK) parameters among children who received atazanavir/ritonavir co-administered with TDF."( Pharmacokinetics of atazanavir/ritonavir among HIV-infected Thai children concomitantly taking tenofovir disoproxil fumarate.
Ananworanich, J; Bunupuradah, T; Keadpudsa, S; Prasitsuebsai, W; Puthanakit, T; Sahakijpicharn, T; Srimuan, A; Techasaensiri, C; Thammajaruk, N, 2014
)
0.91
"Single-sequence, open-label, single-center pharmacokinetic investigation."( Influence of Panax ginseng on the steady state pharmacokinetic profile of lopinavir-ritonavir in healthy volunteers.
Alfaro, RM; Calderón, MM; Chairez, CL; Gordon, LA; Kovacs, JA; Penzak, SR, 2014
)
0.63
" Lopinavir and ritonavir pharmacokinetic parameter values were determined using noncompartmental methods, and preadministration and postadministration ginseng values were compared using a Student t test, where p<0."( Influence of Panax ginseng on the steady state pharmacokinetic profile of lopinavir-ritonavir in healthy volunteers.
Alfaro, RM; Calderón, MM; Chairez, CL; Gordon, LA; Kovacs, JA; Penzak, SR, 2014
)
0.98
" Co-administration with EFV resulted in decreases of 57, 39 and 75% in DTG AUC(0-τ), Cmax and Cτ, respectively."( Effects of enzyme inducers efavirenz and tipranavir/ritonavir on the pharmacokinetics of the HIV integrase inhibitor dolutegravir.
Borland, J; Castellino, S; Chen, S; Guta, P; Hosking, L; Lou, Y; Mosteller, M; Peppercorn, A; Piscitelli, SC; Rubio, JP; Savina, P; Song, I; Wagner, D; Wajima, T; Wilfret, D, 2014
)
0.65
" Serial pharmacokinetic samples for DTG and amprenavir and safety assessments were obtained throughout the study."( Effect of fosamprenavir-ritonavir on the pharmacokinetics of dolutegravir in healthy subjects.
Borland, J; Chen, S; Peppercorn, A; Piscitelli, SC; Song, I; Wajima, T, 2014
)
0.71
" This is the basis of pharmacokinetic enhancement."( Pharmacokinetic enhancers in HIV therapeutics.
Acosta, EP; Delille, C; Larson, KB; Otofokun, I; Wang, K, 2014
)
0.4
"The drug-drug interactions between pitavastatin and darunavir/ritonavir (DRV/r) as well as pitavastatin and efavirenz (EFV) were examined in an open-label, parallel-arm, pharmacokinetic (PK) study in HIV-uninfected healthy volunteers."( Lack of pharmacokinetic interactions between pitavastatin and efavirenz or darunavir/ritonavir.
Aberg, JA; Ma, Q; Malvestutto, CD; Morse, GD; Underberg, JA, 2014
)
0.87
"In the EFV arm, the geometric mean area under the concentration time curve (AUC0-τ) and Cmax of pitavastatin were 85."( Lack of pharmacokinetic interactions between pitavastatin and efavirenz or darunavir/ritonavir.
Aberg, JA; Ma, Q; Malvestutto, CD; Morse, GD; Underberg, JA, 2014
)
0.63
" A pediatric population pharmacokinetic model for APV was developed and simulation was used to identify dosing regimens for pediatric patients receiving FPV in combination with ritonavir (RTV) which resulted in concentrations similar to those in adults receiving FPV/RTV 700/100 mg BID."( Population pharmacokinetic modeling and simulation of amprenavir following fosamprenavir/ritonavir administration for dose optimization in HIV infected pediatric patients.
Barbour, AM; Gibiansky, L; Wire, MB, 2014
)
0.82
" Intensive steady-state pharmacokinetic sampling was conducted after six doses."( Pharmacokinetics and safety of rifabutin in young HIV-infected children receiving rifabutin and lopinavir/ritonavir.
Gous, H; Kellermann, T; Kindra, G; McIlleron, H; Moultrie, H; Sawry, S; Van Rie, A; Wiesner, L, 2015
)
0.63
"Rifabutin dosed at 5 mg/kg three times per week resulted in lower AUC0-48, AUC0-24 and Cmax values for rifabutin and 25-O-desacetyl rifabutin compared with adults receiving 150 mg of rifabutin daily, the current recommended dose."( Pharmacokinetics and safety of rifabutin in young HIV-infected children receiving rifabutin and lopinavir/ritonavir.
Gous, H; Kellermann, T; Kindra, G; McIlleron, H; Moultrie, H; Sawry, S; Van Rie, A; Wiesner, L, 2015
)
0.63
" Such formulations would serve to extend the drug half-life while improving the pharmacokinetic profile and biodistribution to reservoirs of human immunodeficiency virus (HIV) infection."( Pharmacokinetics, biodistribution, and toxicity of folic acid-coated antiretroviral nanoformulations.
Alnouti, Y; Balkundi, S; Fox, HS; Gautam, N; Gendelman, HE; Liu, XM; McMillan, J; Puligujja, P; Thakare, R, 2014
)
0.4
"5 h for 20 mg group; the mean Cmax of last dose was 498 ± 54 ng/mL for 10 mg group and 897 ± 136 ng/mL for 20 mg group."( Pharmacokinetics of sifuvirtide in treatment-naive and treatment-experienced HIV-infected patients.
Che, J; Chen, X; Cheng, Y; Dong, T; Meng, Q; Qian, X; Tong, B, 2014
)
0.4
"Long-acting nanoformulated antiretroviral therapy (nanoART) that targets monocyte-macrophages could improve the drug's half-life and protein-binding capacities while facilitating cell and tissue depots."( Pharmacodynamics of long-acting folic acid-receptor targeted ritonavir-boosted atazanavir nanoformulations.
Bade, AN; Baldridge, HM; Balkundi, SS; Dash, PK; Dimitrov, DS; Feng, Y; Gendelman, HE; Gorantla, S; Hilaire, JR; Kendrick, LM; Liu, XM; McMillan, JM; Poluektova, LY; Puligujja, P; Wang, Y; Ying, T; Zhang, G, 2015
)
0.66
" A compartmental pharmacokinetic (PK) model of LPV/r was developed, including a mechanistic description of competitive inhibition."( A pharmacokinetic model of lopinavir in combination with ritonavir in human.
Duangchaemkarn, K; Lohitnavy, M; Reisfeld, B, 2014
)
0.65
"Inhibition of the cytochrome p450 3A4 enzyme system leads to increases in plasma concentrations of coadministered antiretroviral agents - a concept known as pharmacokinetic boosting."( Pharmacokinetic enhancement in HIV antiretroviral therapy: a comparison of ritonavir and cobicistat.
Bow, D; Ng, J; Norton, M; Renjifo, B; Salem, AH; van Wyk, J,
)
0.36
" From the interaction between atazanavir and DTG via CYP3A4 and UGT1A1 and placental efflux transporter inhibition and considering the infant's probable enzymatic immaturity, the DTG elimination half-life was estimated to be 4-fold longer in neonates than in adults."( Pharmacokinetics of dolutegravir in a premature neonate after HIV treatment intensification during pregnancy.
Amiel, C; Caseris, M; Charpentier, C; Descamps, D; Desnoyer, A; Farnoux, C; Lassel, L; Lê, MP; Pain, JB; Peytavin, G; Pialoux, G, 2015
)
0.42
" Similarly, coadministration of BMS-663068 with RTV increased the BMS-626529 Cmax and AUCtau by 53% and 45%, respectively."( Pharmacokinetic interactions between BMS-626529, the active moiety of the HIV-1 attachment inhibitor prodrug BMS-663068, and ritonavir or ritonavir-boosted atazanavir in healthy subjects.
Bertz, R; Furlong, M; Hanna, GJ; Hruska, M; Hwang, C; Landry, IS; Shah, V; Zhu, L, 2015
)
0.62
"Intensive steady-state 12- or 24-hour pharmacokinetic profiles were performed during the second trimester, third trimester, and postpartum."( Pharmacokinetics of Once Versus Twice Daily Darunavir in Pregnant HIV-Infected Women.
Best, BM; Burchett, SK; Capparelli, EV; Cressey, TR; Kreitchmann, R; Mirochnick, M; Mofenson, LM; Rungruengthanakit, K; Shapiro, D; Smith, E; Stek, A; Wang, J, 2015
)
0.42
" Finally the method was successfully applied for human pharmacokinetic study in eight healthy male volunteers after the oral administration of 600 mg darunavir along with 100 mg ritonavir and 100 mg tenofovir as boosters."( Development and validation of a rapid ultra high performance liquid chromatography with tandem mass spectrometry method for the simultaneous determination of darunavir, ritonavir, and tenofovir in human plasma: Application to human pharmacokinetics.
Aris, AB; Jaafar, J; Madhavi, G; Majid, ZA; Reddy, AV; Talib, J; Umar, K, 2015
)
0.8
" Plasma samples were collected through 144 hours after administration for pharmacokinetic assessments."( Pharmacokinetics and safety of co-administered paritaprevir plus ritonavir, ombitasvir, and dasabuvir in hepatic impairment.
Awni, WM; Bernstein, BM; Ding, B; Dutta, S; Khatri, A; Lawitz, EJ; Marbury, TC; Menon, RM; Mullally, VM; Podsadecki, TJ, 2015
)
0.65
"To evaluate the pharmacokinetic profiles of lopinavir (LPV) in Chinese HIV-infected patients."( [Pharmacokinetic profiles of lopinavir (LPV) in Chinese HIV-infected patients].
Du, X; Fu, Q; Li, T; Liu, Z; Zhang, X, 2015
)
0.42
"A total of 16 patients were enrolled in the LPV pharmacokinetic study."( [Pharmacokinetic profiles of lopinavir (LPV) in Chinese HIV-infected patients].
Du, X; Fu, Q; Li, T; Liu, Z; Zhang, X, 2015
)
0.42
"The non-compartment model pharmacokinetic (PK) parameters were as follows: the peak time of LPV (T(max)) (3."( [Pharmacokinetic profiles of lopinavir (LPV) in Chinese HIV-infected patients].
Du, X; Fu, Q; Li, T; Liu, Z; Zhang, X, 2015
)
0.42
"The pharmacokinetic profiles of LPV in Chinese HIV-1 infected patients demonstrate lower C(min) than those of reported studies, while other parameters are similar."( [Pharmacokinetic profiles of lopinavir (LPV) in Chinese HIV-infected patients].
Du, X; Fu, Q; Li, T; Liu, Z; Zhang, X, 2015
)
0.42
"Tenofovir is an efficacious drug with a long half-life and high activity against both HIV and HBV."( Population pharmacokinetics of tenofovir in HIV/HBV co-infected patients.
Avihingsanon, A; Burger, D; Lewin, SR; Matthews, G; Punyawudho, B; Ruxrungtham, K; Thammajaruk, N; Thongpeang, P, 2015
)
0.42
" The population pharmacokinetic model of tenofovir was developed by a nonlinear mixed-effects modeling approach (NONMEM®)."( Population pharmacokinetics of tenofovir in HIV/HBV co-infected patients.
Avihingsanon, A; Burger, D; Lewin, SR; Matthews, G; Punyawudho, B; Ruxrungtham, K; Thammajaruk, N; Thongpeang, P, 2015
)
0.42
" Physiologically based pharmacokinetic (PBPK) modelling may provide a method to predict pharmacokinetics in pregnant women, without the need to perform extensive in vivo clinical trials."( Physiologically Based Modelling of Darunavir/Ritonavir Pharmacokinetics During Pregnancy.
Burger, D; Colbers, A; Greupink, R; Litjens, C; Russel, FG, 2016
)
0.69
"This study aimed to investigate the pharmacokinetic interactions between quinine and lopinavir boosted with ritonavir (LPV/r) in healthy Thai adults (8 males and 12 females)."( Pharmacokinetic Interactions Between Quinine and Lopinavir/Ritonavir in Healthy Thai Adults.
Cressey, TR; Kongjam, P; Na-Bangchang, K; Rattanapunya, S; Rueangweerayut, R; Tawon, Y, 2015
)
0.87
" The aim of the study was to investigate the pharmacokinetic interaction between the anti-malarial drugs, artesunate-mefloquine and the antiretroviral drug, lopinavir boosted with ritonavir (LPV/r)."( Pharmacokinetic interactions between artesunate-mefloquine and ritonavir-boosted lopinavir in healthy Thai adults.
Cressey, TR; Kongjam, P; Na-Bangchang, K; Rattanapunya, S; Rueangweerayut, R; Tawon, Y, 2015
)
0.85
"The study was an open-label, three-way, sequential, cross-over, pharmacokinetic study in healthy Thai adults."( Pharmacokinetic interactions between artesunate-mefloquine and ritonavir-boosted lopinavir in healthy Thai adults.
Cressey, TR; Kongjam, P; Na-Bangchang, K; Rattanapunya, S; Rueangweerayut, R; Tawon, Y, 2015
)
0.66
" We studied the effect of INH on LPV concentrations by administering INH for 7 days and performing intensive pharmacokinetic sampling in 16 human immunodeficiency virus infected patients established on LPV/r-based ART."( The pharmacokinetics of lopinavir/ritonavir when given with isoniazid in South African HIV-infected individuals.
Decloedt, EH; Maartens, G; McIlleron, H; van der Walt, JS; Wiesner, L, 2015
)
0.7
" We have compared non-compartmental analysis (NCA) and model-based predictions of DDIs for long half-life drugs by conducting simulation studies and reviewing published trials, using antituberculosis drug bedaquiline (BDQ) as a model compound."( Pharmacokinetic Interactions for Drugs with a Long Half-Life—Evidence for the Need of Model-Based Analysis.
Acharya, C; Clauson, B; Dooley, KE; Karlsson, MO; Svensson, EM, 2016
)
0.43
" We conducted a population pharmacokinetic analysis of lopinavir and ritonavir concentrations collected from 84 pregnant and 595 nonpregnant treatment-naive and -experienced HIV-1-infected subjects enrolled in six clinical studies."( No Need for Lopinavir Dose Adjustment during Pregnancy: a Population Pharmacokinetic and Exposure-Response Analysis in Pregnant and Nonpregnant HIV-Infected Subjects.
Jones, AK; Klein, CE; Nilius, AM; Patterson, KB; Salem, AH; Santini-Oliveira, M; Taylor, GP, 2016
)
0.67
" Co-administration with ritonavir increases paritaprevir exposure and half-life without adversely influencing tolerability."( Pharmacokinetics and tolerability of paritaprevir, a direct acting antiviral agent for hepatitis C virus treatment, with and without ritonavir in healthy volunteers.
Awni, WM; Chiu, YL; Dutta, S; Klein, CE; Menon, RM; Podsadecki, TJ, 2016
)
0.95
" Population pharmacokinetic modelling was applied to investigate the interaction and generate alternative doses to inform clinical trial design."( Simulation of the impact of rifampicin on once-daily darunavir/ritonavir pharmacokinetics and dose adjustment strategies: a population pharmacokinetic approach.
Back, D; Boffito, M; Dickinson, L; Khoo, S; Siccardi, M; Winston, A, 2016
)
0.67
"Open-label, single-sequence pharmacokinetic study."( Lack of an Effect of Ritonavir Alone and Lopinavir-Ritonavir on the Pharmacokinetics of Fenofibric Acid in Healthy Volunteers.
Alfaro, RM; Calderón, MM; Gordon, LA; Hadigan, C; Kovacs, JA; Malati, CY; McLaughlin, M; Penzak, SR, 2016
)
0.75
" Fenofibric acid pharmacokinetic parameter values were compared before and after concomitant ritonavir or lopinavir-ritonavir administration."( Lack of an Effect of Ritonavir Alone and Lopinavir-Ritonavir on the Pharmacokinetics of Fenofibric Acid in Healthy Volunteers.
Alfaro, RM; Calderón, MM; Gordon, LA; Hadigan, C; Kovacs, JA; Malati, CY; McLaughlin, M; Penzak, SR, 2016
)
0.97
"Linear regression equations describing the relationships of lopinavir peak and trough concentrations to lopinavir AUC values were established using pharmacokinetic data from published studies of patients or healthy subjects receiving lopinavir and ritonavir at standard dosages."( Prediction of area under the concentration-time curve for lopinavir from peak or trough lopinavir concentrations in patients receiving lopinavir-ritonavir therapy.
Srinivas, NR, 2016
)
0.82
" A population pharmacokinetic model for paritaprevir was developed using data from formulation, bioavailability, and drug-drug interaction studies that evaluated the pharmacokinetics of paritaprevir (coadministered with ritonavir to enhance exposure) with or without ombitasvir and/or dasabuvir at different paritaprevir dose levels."( Dose- and Formulation-Dependent Non-Linear Pharmacokinetic Model of Paritaprevir, a Protease Inhibitor for the Treatment of Hepatitis C Virus Infection: Combined Analysis from 12 Phase I Studies.
Awni, WM; Beck, D; Dutta, S; Khatri, A; Liu, W; Menon, RM; Mensing, S; Polepally, AR, 2016
)
0.62
" Coadministration of dasabuvir increased paritaprevir bioavailability by 59 %; however, ombitasvir coadministration did not affect the pharmacokinetic profile of paritaprevir."( Dose- and Formulation-Dependent Non-Linear Pharmacokinetic Model of Paritaprevir, a Protease Inhibitor for the Treatment of Hepatitis C Virus Infection: Combined Analysis from 12 Phase I Studies.
Awni, WM; Beck, D; Dutta, S; Khatri, A; Liu, W; Menon, RM; Mensing, S; Polepally, AR, 2016
)
0.43
" Here we present a population pharmacokinetic model to describe the longitudinal change of unbound lopinavir/ritonavir (LPV/RTV) PK parameters with gestational age, and to predict unbound LPV concentrations under different dosing regimens."( Model-Based Analysis of Unbound Lopinavir Pharmacokinetics in HIV-Infected Pregnant Women Supports Standard Dosing in the Third Trimester.
Chen, J; Dumond, JB; Malone, S; Patterson, KB; Prince, HM, 2016
)
0.65
" In part 1, 90% confidence intervals for darunavir Cmax and AUC were all within 80-125% for suspension (fed or fasted) versus tablets (fed)."( Pharmacokinetics of darunavir after administration of an oral suspension with low-dose ritonavir and with or without food.
De Paepe, E; Hoetelmans, RM; Kakuda, TN; Lavreys, L; Sekar, V; Stevens, T; Vangeneugden, T; Vanstockem, M, 2014
)
0.63
"Total exposure, measured by area under the plasma concentration-time curve (AUC), was generated for the DAAs, ritonavir, and ribavirin using population pharmacokinetic modeling of data (N = 2093 patients) from 6 Phase 3 studies and 1 Phase 2 study."( Effects of Mild and Moderate Renal Impairment on Ombitasvir, Paritaprevir, Ritonavir, Dasabuvir, and Ribavirin Pharmacokinetics in Patients with Chronic HCV Infection.
Badri, PS; Eckert, D; Menon, RM; Mensing, S; Polepally, AR, 2017
)
0.9
"The pharmacokinetic parameters like peak plasma concentration (Cmax), area under the plasma concentration time profile (AUC) and elimination half life of repaglinide were significantly (p<0."( Altered pharmacokinetics and pharmacodynamics of repaglinide by ritonavir in rats with healthy, diabetic and impaired hepatic function.
Goud, T; Maddi, S; Nayakanti, D; Thatipamula, RP, 2016
)
0.67
" Pharmacokinetic parameters were derived using noncompartmental analysis."( Pharmacokinetics of once-daily darunavir/ritonavir in HIV-1-infected pregnant women.
Baugh, B; Brown, K; Crauwels, HM; Hillewaert, V; Kakuda, TN; Osiyemi, OO; Ryan, B; Verboven, P; Yasin, S; Zorrilla, C, 2016
)
0.7
" The success of these dosing recommendations was evaluated by analyzing pharmacokinetic data from liver transplant recipients in the CORAL-I study."( Pharmacokinetics of Tacrolimus and Cyclosporine in Liver Transplant Recipients Receiving 3 Direct-Acting Antivirals as Treatment for Hepatitis C Infection.
Awni, WM; Badri, PS; Coakley, EP; Ding, B; Dutta, S; Menon, RM; Parikh, A, 2016
)
0.43
"A population pharmacokinetic model was developed using tacrolimus dosing and Ctrough data before and during 3D treatment (n = 29)."( Pharmacokinetics of Tacrolimus and Cyclosporine in Liver Transplant Recipients Receiving 3 Direct-Acting Antivirals as Treatment for Hepatitis C Infection.
Awni, WM; Badri, PS; Coakley, EP; Ding, B; Dutta, S; Menon, RM; Parikh, A, 2016
)
0.43
"A one-compartment model with first-order absorption adequately described tacrolimus pharmacokinetic profiles during the first 4 weeks of 3D treatment."( Pharmacokinetics of Tacrolimus and Cyclosporine in Liver Transplant Recipients Receiving 3 Direct-Acting Antivirals as Treatment for Hepatitis C Infection.
Awni, WM; Badri, PS; Coakley, EP; Ding, B; Dutta, S; Menon, RM; Parikh, A, 2016
)
0.43
"Using data from a phase III clinical trial (GIFT-I) that enrolled Japanese patients with HCV genotype 1b infection, population pharmacokinetic models were developed for the drugs that comprise the 2D regimen: paritaprevir, ombitasvir, and ritonavir."( Population Pharmacokinetics of Paritaprevir, Ombitasvir, and Ritonavir in Japanese Patients with Hepatitis C Virus Genotype 1b Infection.
Gopalakrishnan, SM; Khatri, A; Menon, RM; Mensing, S; Polepally, AR, 2017
)
0.88
"Population pharmacokinetic modeling did not reveal any patient-related or clinical parameters that would require dose adjustment of the 2D regimen when used for the treatment of HCV genotype 1b infection in Japanese patients."( Population Pharmacokinetics of Paritaprevir, Ombitasvir, and Ritonavir in Japanese Patients with Hepatitis C Virus Genotype 1b Infection.
Gopalakrishnan, SM; Khatri, A; Menon, RM; Mensing, S; Polepally, AR, 2017
)
0.7
"The population pharmacokinetic models for each component of the 3D ± ribavirin regimen (DAAs and ritonavir, n = 2348) and ribavirin (n = 1841) adequately described their respective plasma concentration-time data."( Population pharmacokinetics of paritaprevir, ombitasvir, dasabuvir, ritonavir and ribavirin in hepatitis C virus genotype 1 infection: analysis of six phase III trials.
Awni, WM; Dutta, S; Eckert, D; Khatri, A; Menon, RM; Mensing, S; Podsadecki, TJ; Polepally, AR; Sharma, S, 2017
)
0.91
" Here we aimed to quantify nevirapine and LPV/r drug-drug interaction effects on bedaquiline and M2 in patients co-infected with HIV and multidrug-resistant tuberculosis (MDR-TB) using population pharmacokinetic (PK) analysis and compare these with model-based predictions from single-dose studies in subjects without TB."( Confirming model-predicted pharmacokinetic interactions between bedaquiline and lopinavir/ritonavir or nevirapine in patients with HIV and drug-resistant tuberculosis.
Brill, MJ; Karlsson, MO; Maartens, G; Pandie, M; Svensson, EM, 2017
)
0.68
" Results from the five available pharmacokinetic studies show reductions in total darunavir plasma concentrations of between 20-50% during the third trimester of pregnancy."( Pharmacokinetics and Safety of Darunavir/Ritonavir in HIV-Infected Pregnant Women.
Brown, K; Burger, D; Hadacek, MB; Hill, A; Khoo, S; La Porte, C; Moecklinghoff, C; Peytavin, G,
)
0.4
"To our knowledge, there is no published data on the pharmacokinetic (PK) profile of antiretroviral (ART) drugs on patients undergoing extracorporeal membrane oxygenation (ECMO) therapy."( The effect of veno-venous ECMO on the pharmacokinetics of Ritonavir, Darunavir, Tenofovir and Lamivudine.
Ashworth, A; Barker, J; Davies, A; Feddy, L; Fedor, I; Ghazi Suliman, MA; Hayes, T; Kosmidis, C; Malagon, I; Ogungbenro, K; Stirling, S; Szabo-Barnes, A, 2017
)
0.7
"The aim of this study was to determine the pharmacokinetic interaction between ivacaftor and ritonavir."( The pharmacokinetic interaction between ivacaftor and ritonavir in healthy volunteers.
Barry, MG; D'Arcy, DM; Liddy, AM; McLaughlin, G; Schmitz, S, 2017
)
0.92
" Three pharmacokinetic profiles were assessed for each volunteer: ivacaftor 150 mg alone (study A), ivacaftor 150 mg plus ritonavir 50 mg daily (study B), and ivacaftor 150 mg plus ritonavir 50 mg daily after two weeks of ritonavir 50 mg daily (study C)."( The pharmacokinetic interaction between ivacaftor and ritonavir in healthy volunteers.
Barry, MG; D'Arcy, DM; Liddy, AM; McLaughlin, G; Schmitz, S, 2017
)
0.91
" The overall objective of this project was to develop separate physiologically based pharmacokinetic (PBPK) substrate models for the protease inhibitors darunavir and lopinavir."( Physiologically Based Pharmacokinetic Modeling for Predicting the Effect of Intrinsic and Extrinsic Factors on Darunavir or Lopinavir Exposure Coadministered With Ritonavir.
Arya, V; Au, S; Mullick, C; Struble, K; Wagner, C; Zhao, P, 2017
)
0.65
"Ethanol (20 mM) significantly decreased apparent half-life and increased degradation rate constant of RTV-boosted DRV but not for DRV alone."( Influence of Ethanol on Darunavir Hepatic Clearance and Intracellular PK/PD in HIV-Infected Monocytes, and CYP3A4-Darunavir Interactions Using Inhibition and in Silico Binding Studies.
Cory, TJ; Gong, Y; Kumar, S; Li, J; Li, W; Meibohm, B; Midde, NM, 2017
)
0.46
"A literature search of MEDLINE was performed (1985 to April 2017) using the following search terms: cobicistat, ritonavir, pharmacokinetic, drug interactions, booster, pharmacokinetic enhancer, HIV, antiretrovirals."( Cobicistat Versus Ritonavir: Similar Pharmacokinetic Enhancers But Some Important Differences.
Hughes, CA; Phillips, EJ; Seet, J; Tseng, A; Wu, J, 2017
)
1
" Pharmacokinetic properties of the boosted antiretroviral can also affect interaction outcome with comedications."( Cobicistat Versus Ritonavir: Similar Pharmacokinetic Enhancers But Some Important Differences.
Hughes, CA; Phillips, EJ; Seet, J; Tseng, A; Wu, J, 2017
)
0.79
"The aim of this study was to incorporate placental transfer into a pregnancy physiologically based pharmacokinetic model to simulate and evaluate fetal darunavir exposure at term."( Prediction of Fetal Darunavir Exposure by Integrating Human Ex-Vivo Placental Transfer and Physiologically Based Pharmacokinetic Modeling.
Buaben, AO; Burger, DM; Colbers, AP; Freriksen, JJM; Greupink, R; Russel, FGM; Schalkwijk, S, 2018
)
0.48
"An existing and validated pregnancy physiologically based pharmacokinetic model of maternal darunavir/ritonavir exposure was extended with a feto-placental unit."( Prediction of Fetal Darunavir Exposure by Integrating Human Ex-Vivo Placental Transfer and Physiologically Based Pharmacokinetic Modeling.
Buaben, AO; Burger, DM; Colbers, AP; Freriksen, JJM; Greupink, R; Russel, FGM; Schalkwijk, S, 2018
)
0.7
" Scaled placental transfer was integrated into the pregnancy physiologically based pharmacokinetic model."( Prediction of Fetal Darunavir Exposure by Integrating Human Ex-Vivo Placental Transfer and Physiologically Based Pharmacokinetic Modeling.
Buaben, AO; Burger, DM; Colbers, AP; Freriksen, JJM; Greupink, R; Russel, FGM; Schalkwijk, S, 2018
)
0.48
" The antiretroviral pharmacokinetic enhancers, ritonavir and cobicistat, inhibit both these transporters."( Differential Influence of the Antiretroviral Pharmacokinetic Enhancers Ritonavir and Cobicistat on Intestinal P-Glycoprotein Transport and the Pharmacokinetic/Pharmacodynamic Disposition of Dabigatran.
Alfaro, RM; Brooks, KM; George, JM; Gordon, LA; Hadigan, C; Kellogg, A; Kumar, P; Lozier, J; McManus, M; Nghiem, K; Penzak, SR, 2017
)
0.95
"To develop a population pharmacokinetic model and identify sources of variability, genetic and nongenetic factors, of tenofovir."( Pharmacogenetics-based population pharmacokinetic analysis of tenofovir in Thai HIV-infected patients.
Avihingsanon, A; Burger, DM; Mitruk, S; Punyawudho, B; Rungtivasuwan, K; Ruxrungtham, K; Sukasem, C; Thammajaruk, N, 2017
)
0.46
" A nonlinear mixed effects model was used to develop the population pharmacokinetic model and investigate the influence of these polymorphisms and other patient specific covariates on the pharmacokinetics of tenofovir."( Pharmacogenetics-based population pharmacokinetic analysis of tenofovir in Thai HIV-infected patients.
Avihingsanon, A; Burger, DM; Mitruk, S; Punyawudho, B; Rungtivasuwan, K; Ruxrungtham, K; Sukasem, C; Thammajaruk, N, 2017
)
0.46
" A combined population pharmacokinetic model was developed to jointly describe the pharmacokinetics of lopinavir and ritonavir in 32 HIV-infected children (16 with MDR-TB receiving treatment with combinations of high-dose isoniazid, pyrazinamide, ethambutol, ethionamide, terizidone, a fluoroquinolone, and amikacin and 16 without TB) who were established on a lopinavir-ritonavir-containing antiretroviral regimen."( Pharmacokinetics and Drug-Drug Interactions of Lopinavir-Ritonavir Administered with First- and Second-Line Antituberculosis Drugs in HIV-Infected Children Treated for Multidrug-Resistant Tuberculosis.
de Kock, M; Denti, P; Garcia-Prats, AJ; Hesseling, AC; McIlleron, H; Norman, J; Schaaf, HS; Tikiso, T; van der Laan, LE; Wiesner, L; Winckler, J, 2018
)
0.94
"Intensive 24-hour pharmacokinetic profiles at steady state compared ATV exposures (area under the concentration-time curve in one dosing interval) in 5 ATV + RTV baseline weight-band dosing categories, with historic data in adults receiving ATV + RTV 300/100 mg capsules."( Pharmacokinetics and Pharmacodynamics of Atazanavir in HIV-1-Infected Children Treated With Atazanavir Powder and Ritonavir: Combined Analysis of the PRINCE-1 and -2 Studies.
Correll, TA; Eley, T; Pikora, C; Sevinsky, H; Wang, R; Xu, X; Zaru, L, 2018
)
0.69
" Ritonavir (RTV) is a potent mechanism-based and reversible CYP3A inhibitor and moderate inducer that is used as a pharmacokinetic enhancer in several antiviral treatment regimens."( Guiding dose adjustment of amlodipine after co-administration with ritonavir containing regimens using a physiologically-based pharmacokinetic/pharmacodynamic model.
Menon, RM; Mukherjee, D; Shebley, M; Zha, J, 2018
)
1.63
" To provide insights into the complex mechanisms of absorption and disposition of TLC-ART101, we constructed novel mechanism-based pharmacokinetic (MBPK) models."( Mechanism-based pharmacokinetic (MBPK) models describe the complex plasma kinetics of three antiretrovirals delivered by a long-acting anti-HIV drug combination nanoparticle formulation.
Collier, AC; Collins, C; Ho, RJY; Kinman, L; Koehn, J; Kraft, JC; McConnachie, LA; Shen, DD; Sun, J, 2018
)
0.48
" The present study is to develop a physiologically based pharmacokinetic (PBPK) model to predict several scenarios in clinical practice."( Application of physiologically based pharmacokinetic modeling to the prediction of drug-drug and drug-disease interactions for rivaroxaban.
Ge, W; Jiang, Q; Xu, R, 2018
)
0.48
"In this multicenter pharmacokinetic study in HIV-infected children (6-12 years of age), we validated the approved once-daily darunavir/ritonavir dosing recommendations."( Pharmacokinetics, Short-term Safety and Efficacy of the Approved Once-daily Darunavir/Ritonavir Dosing Regimen in HIV-infected Children.
Bastiaans, DET; Burger, DM; Colbers, APH; Geelen, SPM; Roukens, M; van der Flier, M; van Rossum, AMC; Vermont, CL; Visser, EG, 2018
)
0.91
" A 24 h intensive pharmacokinetic blood sampling and a trough seminal sampling were performed before (week 0) and after (week 12) dose reduction."( Pharmacokinetic modelling of darunavir/ritonavir dose reduction (800/100 to 400/100 mg once daily) in a darunavir/ritonavir-containing regimen in virologically suppressed HIV-infected patients: ANRS 165 DARULIGHT sub-study.
Bertrand, J; Chaix, ML; Chevret, S; Delobel, P; El Abbassi, EMB; Gallien, S; Katlama, C; Lê, MP; Molina, JM; Peytavin, G; Raffi, F; Saillard, J; Yazdanpanah, Y, 2018
)
0.75
"Fifteen patients completed the intensive pharmacokinetic analysis."( Pharmacokinetic modelling of darunavir/ritonavir dose reduction (800/100 to 400/100 mg once daily) in a darunavir/ritonavir-containing regimen in virologically suppressed HIV-infected patients: ANRS 165 DARULIGHT sub-study.
Bertrand, J; Chaix, ML; Chevret, S; Delobel, P; El Abbassi, EMB; Gallien, S; Katlama, C; Lê, MP; Molina, JM; Peytavin, G; Raffi, F; Saillard, J; Yazdanpanah, Y, 2018
)
0.75
" Intensive or sparse pharmacokinetic sampling was performed for assessments of plasma drug concentrations."( Pharmacokinetics of Ombitasvir, Paritaprevir, Ritonavir, and Dasabuvir in Healthy Chinese Subjects and HCV GT1b-Infected Chinese, South Korean and Taiwanese Patients.
Alves, K; Ding, B; Luo, Y; Menon, RM; Mobashery, N; Wang, H; Yu, C; Zha, J; Zhao, W, 2019
)
0.77
"Collectively, the results of these pharmacokinetic analyses support the use of the same dose of the 3D regimen for Asian and Western patients."( Pharmacokinetics of Ombitasvir, Paritaprevir, Ritonavir, and Dasabuvir in Healthy Chinese Subjects and HCV GT1b-Infected Chinese, South Korean and Taiwanese Patients.
Alves, K; Ding, B; Luo, Y; Menon, RM; Mobashery, N; Wang, H; Yu, C; Zha, J; Zhao, W, 2019
)
0.77
"A population pharmacokinetic analysis was conducted based on data from 85 women."( Evaluating darunavir/ritonavir dosing regimens for HIV-positive pregnant women using semi-mechanistic pharmacokinetic modelling.
Best, BM; Burger, DM; Capparelli, E; Colbers, A; Cressey, TR; Greupink, R; Karlsson, MO; Mirochnick, M; Moltó, J; Russel, FGM; Schalkwijk, S; Ter Heine, R, 2019
)
0.83
"To evaluate the proportion of children with lopinavir Cmin ≥1 mg/L when receiving a novel 8-hourly lopinavir/ritonavir dosing strategy during rifampicin co-treatment."( Pharmacokinetics of adjusted-dose 8-hourly lopinavir/ritonavir in HIV-infected children co-treated with rifampicin.
McIlleron, H; Rabie, H; Rawizza, H; Van Rie, A; Wiesner, L; Winckler, J; Zar, H; Zuidewind, P, 2019
)
0.98
"HIV-infected children on lopinavir/ritonavir and rifampicin were enrolled in a prospective pharmacokinetic study."( Pharmacokinetics of adjusted-dose 8-hourly lopinavir/ritonavir in HIV-infected children co-treated with rifampicin.
McIlleron, H; Rabie, H; Rawizza, H; Van Rie, A; Wiesner, L; Winckler, J; Zar, H; Zuidewind, P, 2019
)
1.04
"A population pharmacokinetic (PopPK) model for pretomanid was developed using data from 14 studies in the pretomanid development program: six phase 1 studies, six phase 2 studies, and two phase 3 studies."( Population Pharmacokinetics of the Antituberculosis Agent Pretomanid.
Everitt, D; Nedelman, JR; Salinger, DH; Subramoney, V, 2019
)
0.51
"We did a parallel, three-group, pharmacokinetic evaluation at HIV clinics in Asia (two sites), South America (five), sub-Saharan Africa (three), and the USA (11) between Dec 30, 2014, and Sept 12, 2016."( Antiretroviral therapy and vaginally administered contraceptive hormones: a three-arm, pharmacokinetic study.
Akelo, V; Aweeka, F; Aziz, M; Berzins, B; Cohn, SE; Coombs, RW; Coughlin, K; Cramer, YS; Friedman, RK; Gingrich, D; Godfrey, C; Moran, LE; Rosenkranz, SL; Scarsi, KK; Swaminathan, S; Zorrilla, CD, 2019
)
0.51
" Further studies designed to examine pharmacodynamic endpoints, such as ovulation, when intravaginal ring hormones are combined with efavirenz are warranted."( Antiretroviral therapy and vaginally administered contraceptive hormones: a three-arm, pharmacokinetic study.
Akelo, V; Aweeka, F; Aziz, M; Berzins, B; Cohn, SE; Coombs, RW; Coughlin, K; Cramer, YS; Friedman, RK; Gingrich, D; Godfrey, C; Moran, LE; Rosenkranz, SL; Scarsi, KK; Swaminathan, S; Zorrilla, CD, 2019
)
0.51
" The aim of the current study was to develop a population pharmacokinetic (PK) model for docetaxel and ritonavir based on the phase 1 studies and to support drug development of this combination treatment."( A Population Pharmacokinetic Model of Oral Docetaxel Coadministered With Ritonavir to Support Early Clinical Development.
Beijnen, JH; de Weger, VA; Huitema, ADR; Janssen, JM; Nuijen, B; Sawicki, E; Schellens, JHM; van Hasselt, JGC; Yu, H, 2020
)
1
" The objective of the study was to apply physiologically-based pharmacokinetic (PBPK) modeling to predict optimal dosage regimens of quinine when coadministered with lopinavir/ritonavir in malaria and HIV coinfected patients with different conditions."( Physiologically-Based Pharmacokinetic Modeling for Optimal Dosage Prediction of Quinine Coadministered With Ritonavir-Boosted Lopinavir.
Karbwang, J; Na-Bangchang, K; Rajoli, RKR; Saeheng, T; Siccardi, M, 2020
)
0.96
"The potential for clinically significant drug interactions (CSDIs) for patients taking ritonavir and cobicistat is high because of their powerful pharmacokinetic effect on the cytochrome P450 (CYP) enzyme system, most notably their inhibitory effect on CYP3A4."( Pharmacokinetic enhancers (cobicistat/ritonavir) and the potential for drug-drug interactions.
de Barra, E; Hollywood, P; Lorigan, D; MacCann, R; McConkey, S, 2020
)
1.05
"Two hundred individuals attending a regional specialist human immunodeficiency virus (HIV) clinic between June and September 2014 who were receiving the pharmacokinetic enhancers ritonavir or cobicistat were interviewed to determine a medication history including medications prescribed by their general practitioner (GP), over-the-counter (OTC) medicines, herbal remedies and recreational drugs."( Pharmacokinetic enhancers (cobicistat/ritonavir) and the potential for drug-drug interactions.
de Barra, E; Hollywood, P; Lorigan, D; MacCann, R; McConkey, S, 2020
)
1.02
"3) were applied to determine pharmacokinetic parameters and assess demographic covariates and relationships with SNPs (SLCO3A1, SLCO1B1, NR1I2, NR1I3, CYP3A5*3, CYP3A4*22, ABCC2, ABCC10, ABCG2 and SCL47A1)."( Population pharmacokinetics and pharmacogenetics of ritonavir-boosted darunavir in the presence of raltegravir or tenofovir disoproxil fumarate/emtricitabine in HIV-infected adults and the relationship with virological response: a sub-study of the NEAT001
Boffito, M; Bonora, S; Cursley, A; D'Avolio, A; Di Perri, G; Dickinson, L; Fäetkenheuer, G; Gurjar, R; Molina, JM; Owen, A; Pozniak, A; Raffi, F; Richert, L; Stöhr, W; Vandekerckhove, L, 2020
)
0.81
"Darunavir (DRV) and ritonavir (RTV; r) intensive pharmacokinetic evaluations were performed at steady state during the second and third trimesters of pregnancy (DRV/r 800/100 mg bid) and 2-3 weeks postpartum (DRV/r 600/100 mg twice daily)."( Darunavir Pharmacokinetics With an Increased Dose During Pregnancy.
Best, BM; Capparelli, EV; Chakhtoura, N; Eke, AC; Kreitchmann, R; Mirochnick, M; Shapiro, DE; Smith, E; Stek, AM; Wang, J, 2020
)
0.88
"To evaluate the safety and pharmacokinetic profile of adjusted doses of darunavir/ritonavir with rifampicin."( Pharmacokinetic profile and safety of adjusted doses of darunavir/ritonavir with rifampicin in people living with HIV.
Ebrahim, I; Maartens, G; McIlleron, H; Orrell, C; Smythe, W; Wiesner, L, 2020
)
1.02
" Therefore, an intravenous study of TLC-ART 101 in nonhuman primates was conducted to elucidate the degree of association of drugs in vivo, estimate subcutaneous bioavailability, and refine a mechanism-based pharmacokinetic (MBPK2) model."( Integration of Computational and Experimental Approaches to Elucidate Mechanisms of First-Pass Lymphatic Drug Sequestration and Long-Acting Pharmacokinetics of the Injectable Triple-HIV Drug Combination TLC-ART 101.
Collier, AC; Ho, RJY; Kinman, L; Koehn, J; Lane, S; Lee, W; McConnachie, LA; Perazzolo, S; Shen, DD; Shireman, LM, 2020
)
0.56
" A population pharmacokinetic meta-analysis was conducted using individual participant data from 10 studies with 6,100 lumefantrine concentrations from 793 nonpregnant adult participants (41% HIV-malaria-coinfected, 36% malaria-infected, 20% HIV-infected, and 3% healthy volunteers)."( An Individual Participant Data Population Pharmacokinetic Meta-analysis of Drug-Drug Interactions between Lumefantrine and Commonly Used Antiretroviral Treatment.
Aweeka, FT; Barnes, KI; Byakika-Kibwika, P; Bygbjerg, IC; Chijioke-Nwauche, I; Denti, P; Francis, J; Hoglund, RM; Khoo, SH; Kredo, T; Lamorde, M; Lemnge, MM; Merry, C; Nyagonde, N; Parikh, S; Scarsi, KK; Sutherland, CJ; Tarning, J; Vestergaard, LS; Walimbwa, SI; Workman, L, 2020
)
0.56
" The aim of current analysis was to develop a pharmacokinetic (PK)-toxicodynamic (TOX) model to quantify the relationship between docetaxel plasma exposure and DLTs."( Quantification of the pharmacokinetic-toxicodynamic relationship of oral docetaxel co-administered with ritonavir.
Beijnen, JH; de Weger, VA; Dorlo, TPC; Huitema, ADR; Janssen, JM; Marchetti, S; Nuijen, B; Schellens, JHM; Stuurman, RE; Yu, H, 2020
)
0.77
" A physiologically-based pharmacokinetic (PBPK) model was developed integrating in vitro, preclinical, and clinical data of HVs and patients with cancer."( Ribociclib Drug-Drug Interactions: Clinical Evaluations and Physiologically-Based Pharmacokinetic Modeling to Guide Drug Labeling.
Chakraborty, A; Dhuria, SV; Elmeliegy, M; He, H; Heimbach, T; Huth, F; Ji, Y; Miller, M; Samant, TS; Schiller, H; Umehara, K, 2020
)
0.56
"This study aimed to assess the pharmacokinetic profile of 150 mg rifabutin (RBT) taken every other day (every 48 h) versus 300 mg RBT taken every other day (E."( Pharmacokinetic study of two different rifabutin doses co-administered with lopinavir/ritonavir in African HIV and tuberculosis co-infected adult patients.
Cisse, K; Compaoré, TR; Diagbouga, S; Kouanda, S; Matteelli, A; Ouedraogo, HG; Regazzi, M; Roggi, A; Sangare, L; Simporé, J; Sulis, G; Tarnagda, G; Villani, P, 2020
)
0.78
" Lopinavir and ritonavir pharmacokinetic parameters were determined after an intensive pharmacokinetic analysis."( Pharmacokinetics of lopinavir/ritonavir oral solution to treat COVID-19 in mechanically ventilated ICU patients.
Bouadma, L; de Montmollin, E; Descamps, D; Jaquet, P; Kauv, J; Laouénan, C; Le Hingrat, Q; Lê, MP; Patrier, J; Peytavin, G; Sonneville, R; Timsit, JF; Veyrier, M; Visseaux, B; Wicky, PH, 2020
)
1.2
" After the dose reduction to 400/100 mg q24h, lopinavir/ritonavir pharmacokinetic parameters were assessed in nine patients."( Pharmacokinetics of lopinavir/ritonavir oral solution to treat COVID-19 in mechanically ventilated ICU patients.
Bouadma, L; de Montmollin, E; Descamps, D; Jaquet, P; Kauv, J; Laouénan, C; Le Hingrat, Q; Lê, MP; Patrier, J; Peytavin, G; Sonneville, R; Timsit, JF; Veyrier, M; Visseaux, B; Wicky, PH, 2020
)
1.09
" However, it remains difficult to safely recommend its dose reduction without compromising the benefit of the antiviral strategy, and careful pharmacokinetic and toxicity monitoring are needed."( Pharmacokinetics of lopinavir/ritonavir oral solution to treat COVID-19 in mechanically ventilated ICU patients.
Bouadma, L; de Montmollin, E; Descamps, D; Jaquet, P; Kauv, J; Laouénan, C; Le Hingrat, Q; Lê, MP; Patrier, J; Peytavin, G; Sonneville, R; Timsit, JF; Veyrier, M; Visseaux, B; Wicky, PH, 2020
)
0.85
" However, darunavir displays a large, poorly characterized, inter-individual pharmacokinetic variability."( Exploration of Reduced Doses and Short-Cycle Therapy for Darunavir/Cobicistat in Patients with HIV Using Population Pharmacokinetic Modeling and Simulations.
Belkhir, L; Elens, L; Haufroid, V; Stillemans, G; Vandercam, B; Vincent, A, 2021
)
0.62
"Sparse pharmacokinetic samples were collected in 127 patients with human immunodeficiency virus type 1 infection, then supplemented with rich sampling data from a subset of 12 individuals."( Exploration of Reduced Doses and Short-Cycle Therapy for Darunavir/Cobicistat in Patients with HIV Using Population Pharmacokinetic Modeling and Simulations.
Belkhir, L; Elens, L; Haufroid, V; Stillemans, G; Vandercam, B; Vincent, A, 2021
)
0.62
" However, the heterogeneity in pharmacokinetic response should be considered when assessing whether individual patients could benefit from a particular regimen, for instance through the use of population pharmacokinetic models."( Exploration of Reduced Doses and Short-Cycle Therapy for Darunavir/Cobicistat in Patients with HIV Using Population Pharmacokinetic Modeling and Simulations.
Belkhir, L; Elens, L; Haufroid, V; Stillemans, G; Vandercam, B; Vincent, A, 2021
)
0.62
" Pharmacokinetic modelling can explore utility of drug in hair."( A population pharmacokinetic model is beneficial in quantifying hair concentrations of ritonavir-boosted atazanavir: a study of HIV-infected Zimbabwean adolescents.
Chawana, TD; Ngara, B; Nhachi, CFB; Rusakaniko, S; Stray-Pedersen, B; Zvada, S, 2020
)
0.78
" The use of boosted regimens in pregnant women living with HIV has been studied for a variety of ARVs; however, a recent recommendation by the US Food and Drug Administration advised against cobicistat-boosted regimens in pregnancy due to substantially lower drug exposures observed in clinical pharmacokinetic studies."( Pharmacokinetic Enhancement of HIV Antiretroviral Therapy During Pregnancy.
Best, BM; Eke, AC; Mirochnick, M; Momper, JD; Salama, E, 2020
)
0.56
" Pharmacokinetic sampling was conducted until 48 h after both study drug administrations."( Effect of Food on the Pharmacokinetics of the Oral Docetaxel Tablet Formulation ModraDoc006 Combined with Ritonavir (ModraDoc006/r) in Patients with Advanced Solid Tumours.
Beijnen, JH; de Weger, VA; Huitema, ADR; Janssen, JM; Keessen, M; Lopez-Yurda, MI; Marchetti, S; Rosing, H; Thijssen, B; Vermunt, MAC, 2021
)
0.83
" We used physiologically-based pharmacokinetic (PBPK) modeling to simulate DDI magnitudes of various scenarios to guide the clinical DDI management of bictegravir."( Physiologically-Based Pharmacokinetic Modeling to Support the Clinical Management of Drug-Drug Interactions With Bictegravir.
Battegay, M; Marzolini, C; Stader, F, 2021
)
0.62
"The anticancer drug docetaxel exhibits large interpatient pharmacokinetic and pharmacodynamic variability."( Investigating the influence of relevant pharmacogenetic variants on the pharmacokinetics and pharmacodynamics of orally administered docetaxel combined with ritonavir.
Beijnen, JH; Dorlo, TPC; Huitema, ADR; Marchetti, S; Pluim, D; van Eijk, M, 2021
)
0.82
" The aim of this study was to establish an insect model for pharmacokinetic and drug-drug interaction studies to develop sustainable endectocides for vector control."( The pharmacokinetics and drug-drug interactions of ivermectin in Aedes aegypti mosquitoes.
Chaccour, C; Duthaler, U; Hammann, F; Hofer, L; Krähenbühl, S; Maia, M; Müller, P; Weber, M, 2021
)
0.62
" This study evaluated S-warfarin limited sampling strategy with a population pharmacokinetic (PK) approach to estimate CYP2C9 activity in healthy adults."( S-warfarin limited sampling strategy with a population pharmacokinetic approach to estimate exposure and cytochrome P450 (CYP) 2C9 activity in healthy adults.
Bertino, JS; Capparelli, EV; Kashuba, ADM; Ma, JD; Nafziger, AN; Nikanjam, M; Tran, L; Turpault, S, 2021
)
0.62
" However, interaction of both drugs, with Cytochrome P 3A4 (CYP 3A4) isoenzyme, may spawn clinically significant pharmacokinetic interactions."( Evaluation of the effects of atazanavir-ritonavir on the pharmacokinetics of lumefantrine in patients living with HIV in Lagos University Teaching Hospital, South-Western Nigeria.
Abideen, G; Adewumi, OO; Agbaje, EO; Akanmu, AS; Akinleye, MO; Akinyede, A; Busari, AA; Hassan, OO; Kadri, MR; Oreagba, IA; Usman, SO, 2021
)
0.89
" The concentration of lumefantrine in each sample was quantified with high-performance liquid chromatography (HPLC) and used to determine its pharmacokinetic parameters which were compared between the test and control groups."( Evaluation of the effects of atazanavir-ritonavir on the pharmacokinetics of lumefantrine in patients living with HIV in Lagos University Teaching Hospital, South-Western Nigeria.
Abideen, G; Adewumi, OO; Agbaje, EO; Akanmu, AS; Akinleye, MO; Akinyede, A; Busari, AA; Hassan, OO; Kadri, MR; Oreagba, IA; Usman, SO, 2021
)
0.89
" Ivacaftor concentrations were measured in all blood samples in order to calculate the pharmacokinetic parameters for ivacaftor."( Pharmacokinetic interactions between ivacaftor and cytochrome P450 3A4 inhibitors in people with cystic fibrosis and healthy controls.
Heijerman, HGM; Sturm, R; van der Meer, R; Wilms, EB, 2021
)
0.62
"3) ADVAN 13, based on a previously established pharmacokinetic model."( Pharmacokinetic-pharmacodynamic modelling of atazanavir in hair among adolescents on antiretroviral treatment in Zimbabwe.
Chawana, TD; Ngara, B; Nhachi, CFB; Rusakaniko, S; Zvada, S, 2021
)
0.62
" A pooled population pharmacokinetic analysis with 744 plasma medroxyprogesterone acetate concentrations from 138 women treated with depot medroxyprogesterone and antiretroviral/antituberculosis treatment across three clinical trials was performed."( A Semimechanistic Pharmacokinetic Model for Depot Medroxyprogesterone Acetate and Drug-Drug Interactions With Antiretroviral and Antituberculosis Treatment.
Cohn, SE; Denti, P; Dooley, KE; Firnhaber, C; Francis, J; Godfrey, C; Kendall, MA; McIlleron, H; Mngqibisa, R; Wu, X, 2021
)
0.62
" Pooled LPV AUC12 (157 203 hours × ng/mL) and Ctrough (9876 ng/mL) were similar to historical controls; RBT AUC24 (7374 hours × ng/mL) and Ctrough (208 ng/mL) were higher, although 3 participants in arm C had RBT Cmax <250 ng/mL."( Safety and Pharmacokinetics of Double-Dose Lopinavir/Ritonavir + Rifampin Versus Lopinavir/Ritonavir + Daily Rifabutin for Treatment of Human Immunodeficiency Virus-Tuberculosis Coinfection.
Benson, CA; Cardoso, SW; Fletcher, CV; Ive, P; Kendall, MA; Lalloo, U; Podany, AT; Wu, X, 2021
)
0.87
" Faster RBT clearance and low Cmax in 3 participants on RBT+RAL requires further study."( Safety and Pharmacokinetics of Double-Dose Lopinavir/Ritonavir + Rifampin Versus Lopinavir/Ritonavir + Daily Rifabutin for Treatment of Human Immunodeficiency Virus-Tuberculosis Coinfection.
Benson, CA; Cardoso, SW; Fletcher, CV; Ive, P; Kendall, MA; Lalloo, U; Podany, AT; Wu, X, 2021
)
0.87
" We used physiologically-based pharmacokinetic (PBPK) modeling to simulate the effect of inflammation on the pharmacokinetics of CYP3A metabolized drugs."( Physiologically Based Pharmacokinetic Modelling to Investigate the Impact of the Cytokine Storm on CYP3A Drug Pharmacokinetics in COVID-19 Patients.
Battegay, M; Marzolini, C; Sendi, P; Stader, F, 2022
)
0.72
"This is an open-label, randomized, fixed sequence single intravenous dosing study to assess pharmacokinetic interactions between remdesivir and TDF/3TC (Study A, crossover design) or TDF/3TC plus ATV/r (Study B)."( An open-label, randomized, single intravenous dosing study to investigate the effect of fixed-dose combinations of tenofovir/lamivudine or atazanavir/ritonavir on the pharmacokinetics of remdesivir in Ugandan healthy volunteers (RemTLAR).
Byakika-Kibwika, P; D'Avolio, A; Kaboggoza, JP; Lamorde, M; Waitt, C; Walimbwa, SI, 2021
)
0.82
" Furthermore, this study will deliver pharmacokinetic datasets for remdesivir drug concentrations and demographic characteristics which could support pharmacometric approaches for simulation of remdesivir treatment regimens in patients concurrently using tenofovir/lamivudine and/or atazanavir/ritonavir."( An open-label, randomized, single intravenous dosing study to investigate the effect of fixed-dose combinations of tenofovir/lamivudine or atazanavir/ritonavir on the pharmacokinetics of remdesivir in Ugandan healthy volunteers (RemTLAR).
Byakika-Kibwika, P; D'Avolio, A; Kaboggoza, JP; Lamorde, M; Waitt, C; Walimbwa, SI, 2021
)
1
" In stage 1, term neonates exposed to HIV on standard antiretroviral prophylaxis (nevirapine ± zidovudine) received single dose(s) of the 4-in-1 formulation, followed by intensive pharmacokinetic sampling and safety assessments."( Pharmacokinetics and Safety of the Abacavir/Lamivudine/Lopinavir/Ritonavir Fixed-Dose Granule Formulation (4-in-1) in Neonates: PETITE Study.
Andrieux-Meyer, I; Bekker, A; Capparelli, E; Cotton, MF; Cressey, R; Cressey, TR; du Toit, S; Groenewald, M; Kumar, M; Lallemant, M; Nielsen, J; Rabie, H; Salvadori, N; Than-In-At, K, 2022
)
0.96
"Sixteen neonates, with a median (range) birth weight of 3130 g (2790-3590 g), completed 24 pharmacokinetic visits."( Pharmacokinetics and Safety of the Abacavir/Lamivudine/Lopinavir/Ritonavir Fixed-Dose Granule Formulation (4-in-1) in Neonates: PETITE Study.
Andrieux-Meyer, I; Bekker, A; Capparelli, E; Cotton, MF; Cressey, R; Cressey, TR; du Toit, S; Groenewald, M; Kumar, M; Lallemant, M; Nielsen, J; Rabie, H; Salvadori, N; Than-In-At, K, 2022
)
0.96
" CYP3A-mediated DDI of elexacaftor-tezacaftor-ivacaftor was evaluated using a physiologically-based pharmacokinetic modeling approach."( Physiologically-Based Pharmacokinetic-Led Guidance for Patients With Cystic Fibrosis Taking Elexacaftor-Tezacaftor-Ivacaftor With Nirmatrelvir-Ritonavir for the Treatment of COVID-19.
Almond, LM; Beringer, PM; Chung, PS; Hong, E; Rao, AP, 2022
)
0.92
" Boosting pharmacokinetic exposure of KIs metabolized by cytochrome P450 (CYP)3A4 with ritonavir might result in lower doses needed and subsequently reduces treatment costs."( Ritonavir-Boosted Exposure of Kinase Inhibitors: an Open Label, Cross-over Pharmacokinetic Proof-of-Concept Trial with Erlotinib.
Baas, P; Beijnen, JH; Boosman, RJ; Burgers, JA; de Gooijer, CJ; Groenland, SL; Huitema, ADR; Steeghs, N; van der Noort, V, 2022
)
2.39
" Complete pharmacokinetic profiles at steady-state were taken up to 24 h after erlotinib intake for both dosing strategies."( Ritonavir-Boosted Exposure of Kinase Inhibitors: an Open Label, Cross-over Pharmacokinetic Proof-of-Concept Trial with Erlotinib.
Baas, P; Beijnen, JH; Boosman, RJ; Burgers, JA; de Gooijer, CJ; Groenland, SL; Huitema, ADR; Steeghs, N; van der Noort, V, 2022
)
2.16
" We aimed to apply physiologically-based pharmacokinetic (PBPK) modeling to simulate the complex drug-drug interactions (DDIs) of ritonavir with two anticoagulants, rivaroxaban and racemic warfarin, to address this important clinical conundrum."( Physiologically-Based Pharmacokinetic Modeling-Guided Dose Management of Oral Anticoagulants when Initiating Nirmatrelvir/Ritonavir (Paxlovid) for COVID-19 Treatment.
Chan, ECY; Wang, Z, 2022
)
1.13
" We aimed to characterize the pharmacokinetics of different paclitaxel formulations, co-administered with ritonavir, and to investigate a pharmacodynamic relationship between low-dose metronomic (LDM) treatment with oral paclitaxel and the anti-angiogenic marker thrombospondin-1 (TSP-1)."( Development of a population pharmacokinetic/pharmacodynamic model for various oral paclitaxel formulations co-administered with ritonavir and thrombospondin-1 based on data from early phase clinical studies.
Beijnen, JH; de Weger, VA; Dorlo, TPC; Huitema, ADR; Nuijen, B; Sawicki, E; van Eijk, M; Yu, H, 2022
)
1.14
"Fifty-eight patients treated with different oral paclitaxel formulations were included for pharmacokinetic analysis."( Development of a population pharmacokinetic/pharmacodynamic model for various oral paclitaxel formulations co-administered with ritonavir and thrombospondin-1 based on data from early phase clinical studies.
Beijnen, JH; de Weger, VA; Dorlo, TPC; Huitema, ADR; Nuijen, B; Sawicki, E; van Eijk, M; Yu, H, 2022
)
0.93
"The developed pharmacokinetic model adequately described the paclitaxel plasma concentrations for the different oral formulations co-administered with ritonavir."( Development of a population pharmacokinetic/pharmacodynamic model for various oral paclitaxel formulations co-administered with ritonavir and thrombospondin-1 based on data from early phase clinical studies.
Beijnen, JH; de Weger, VA; Dorlo, TPC; Huitema, ADR; Nuijen, B; Sawicki, E; van Eijk, M; Yu, H, 2022
)
1.13
"Physiologically based pharmacokinetic (PBPK) models have gained in popularity in the last decade in both drug development and regulatory science."( A Comparative Analysis of Physiologically Based Pharmacokinetic Models for Human Immunodeficiency Virus and Tuberculosis Infections.
Jacobs, BA; Mtshali, S, 2022
)
0.72
" Then, the established method was used for determining the pharmacokinetic profile of Paxlovid in healthy Chinese volunteers."( Simultaneous determination of nirmatrelvir and ritonavir in human plasma using LC-MS/MS and its pharmacokinetic application in healthy Chinese volunteers.
Boucetta, H; Cao, L; Hang, T; Liu, C; Lu, Y; Song, M; Zhu, M, 2022
)
0.98
"Certain drugs inherently have unfavourable pharmacokinetic properties; for example, they are poorly absorbed or broken down too quickly in the liver."( [CYP3A inhibitors as a pharmacokinetic enhancer: pros and cons of drug interactions].
Burger, DM; Ter Heine, R; van Erp, PH, 2022
)
0.72
"Two separate, parallel, three-group, non-randomized, pharmacokinetic studies evaluated either etonogestrel or levonorgestrel in women receiving rilpivirine- or darunavir-based ART compared with women without HIV (control group)."( Pharmacokinetics of levonorgestrel and etonogestrel contraceptive implants over 48 weeks with rilpivirine- or darunavir-based antiretroviral therapy.
Byakika-Kibwika, P; Chappell, CA; Fletcher, CV; Jeppson, J; Kaboggoza, J; Kyohairwe, I; Lamorde, M; Mbabazi, L; Musaazi, J; Nakalema, S; Nakijoba, R; Nassiwa, S; Pham, M; Scarsi, KK; Siccardi, M; Walimbwa, SI; Winchester, L, 2022
)
0.72
"Therapy failure caused by complex population-drug-drug (PDDI) interactions including CYP3A4 can be predicted using mechanistic physiologically-based pharmacokinetic (PBPK) modeling."( The investigation of the complex population-drug-drug interaction between ritonavir-boosted lopinavir and chloroquine or ivermectin using physiologically-based pharmacokinetic modeling.
Alsmadi, MM, 2023
)
1.14
" This study aimed to develop a population pharmacokinetic model of LPV/r and provide dosage optimization in Thai PLWH."( Dose optimization with population pharmacokinetics of ritonavir-boosted lopinavir for Thai people living with HIV with and without active tuberculosis.
Avihingsanon, A; Chaivichacharn, P; Gatechompol, S; Punyawudho, B; Ubolyam, S, 2022
)
0.97
"The population pharmacokinetic model was developed by integrating the interaction between LPV and RTV."( Dose optimization with population pharmacokinetics of ritonavir-boosted lopinavir for Thai people living with HIV with and without active tuberculosis.
Avihingsanon, A; Chaivichacharn, P; Gatechompol, S; Punyawudho, B; Ubolyam, S, 2022
)
0.97
" This study compared the bioequivalence and safety of 2 lopinavir/ritonavir (200/50 mg) formulations under fasted and fed conditions in healthy Chinese volunteers and compared the pharmacokinetic parameters of lopinavir and ritonavir."( Pharmacokinetics, Safety, and Bioequivalence of 2 Lopinavir/Ritonavir (200/50 mg) Tablets in Healthy Chinese Volunteers: Effect of Food on Absorption.
Bao, D; Fu, W; Hu, W; Lin, L; Liu, Y; Zhang, Q; Zhang, W; Zheng, L, 2023
)
1.39
" Within a nested pharmacokinetic (PK) substudy, we performed a population PK analysis to describe total and unbound dolutegravir plasma concentrations in children and adolescents receiving this dual therapy."( Population pharmacokinetics of unbound and total dolutegravir concentrations in children aged 12 years and older: a PK substudy of the SMILE trial.
Abdalla, S; Coelho, A; Compagnucci, A; Cressey, TR; Hirt, D; Ramos, JT; Riault, Y; Saidi, Y; Tréluyer, JM; Zheng, Y, 2023
)
0.91
" This is the first disclosure of Paxlovid physiologically-based pharmacokinetic (PBPK) model."( Physiologically-Based Pharmacokinetic Modeling of PAXLOVID™ with First-Order Absorption Kinetics.
Di, L; Jaini, R; Lin, J; Sagawa, K, 2023
)
0.91
" At steady state, 8-9 blood samples were taken to construct pharmacokinetic curves."( First Pharmacokinetic Data of Tenofovir Alafenamide Fumarate and Tenofovir With Dolutegravir or Boosted Protease Inhibitors in African Children: A Substudy of the CHAPAS-4 Trial.
Bamford, A; Burger, DM; Bwakura-Dangarembizi, M; Chabala, C; Colbers, A; Denti, P; Doerholt, K; Gibb, DM; Griffiths, AL; Makumbi, S; McIlleron, HM; Monkiewicz, LN; Mulenga, V; Mumbiro, V; Musiime, V; Nangiya, J; Szubert, AJ; Waalewijn, H; Wasmann, RE; Wiesner, L, 2023
)
0.91
" For each combination, tenofovir GM (CV%) AUCtau and Cmax remained below reference values in adults taking 25 mg TAF with a boosted protease inhibitors."( First Pharmacokinetic Data of Tenofovir Alafenamide Fumarate and Tenofovir With Dolutegravir or Boosted Protease Inhibitors in African Children: A Substudy of the CHAPAS-4 Trial.
Bamford, A; Burger, DM; Bwakura-Dangarembizi, M; Chabala, C; Colbers, A; Denti, P; Doerholt, K; Gibb, DM; Griffiths, AL; Makumbi, S; McIlleron, HM; Monkiewicz, LN; Mulenga, V; Mumbiro, V; Musiime, V; Nangiya, J; Szubert, AJ; Waalewijn, H; Wasmann, RE; Wiesner, L, 2023
)
0.91
" To evaluate and quantify the DDIs between them and provide rational dose management strategies of BTK inhibitors, we conducted this study using physiologically-based pharmacokinetic (PBPK) models."( Drug-drug interactions and dose management of BTK inhibitors when initiating nirmatrelvir/ritonavir (paxlovid) based on physiologically-based pharmacokinetic models.
Chen, L; Chen, W; Li, C; Li, L, 2023
)
1.13
"Physicochemical properties and pharmacokinetic parameters were acquired from the published literature and databases."( Drug-drug interactions and dose management of BTK inhibitors when initiating nirmatrelvir/ritonavir (paxlovid) based on physiologically-based pharmacokinetic models.
Chen, L; Chen, W; Li, C; Li, L, 2023
)
1.13

Compound-Compound Interactions

Nirmatrelvir/ritonavir has a high potential to cause harm from drug-drug interactions (DDIs) with other drugs metabolized through this pathway. Danoprevir is a potent macrocyclic inhibitor of the hepatitis C virus NS3/4A protease, which is currently in development in combination with low-dose ritonavir.

ExcerptReferenceRelevance
" In this study we investigated the kinetics of SQV when administered alone and in combination with RIT in HIV-infected patients."( Saquinavir pharmacokinetics alone and in combination with ritonavir in HIV-infected patients.
Back, DJ; Barry, MG; Breckenridge, AM; Gibbons, SE; Heavey, J; Merry, C; Mulcahy, F; Ryan, M; Tjia, JF, 1997
)
0.54
" Steady-state SQV profiles were obtained on two occasions following treatment with SQV 600 mg three times daily alone and when administered with RIT 300 mg twice daily."( Saquinavir pharmacokinetics alone and in combination with ritonavir in HIV-infected patients.
Back, DJ; Barry, MG; Breckenridge, AM; Gibbons, SE; Heavey, J; Merry, C; Mulcahy, F; Ryan, M; Tjia, JF, 1997
)
0.54
" Many of the drugs commonly taken by patients with HIV have a strong potential to interact with the protease inhibitors."( Drug interactions of HIV protease inhibitors.
Kuper, JJ; Malaty, LI, 1999
)
0.3
"4 ng/ml, when combined with RTV and proved to be significantly higher (p <0."( Single daily doses of saquinavir achieve HIV-inhibitory concentrations when combined with baby-dose ritonavir.
Donath, F; Kurowski, M; Möcklinghoff, C; Mrozikiewicz, M; Müller, M, 1999
)
0.52
"Due to changes in hepatic metabolism, protease inhibitors may interact with concurrent treatment."( [HIV protease inhibitors: drug interactions].
Dubreuil, L; Gérard, Y; Maulin, L; Mouton, Y, 1999
)
0.3
"0 microM) were marginal and are not likely to produce clinically significant drug-drug interactions."( Potent inhibition of the cytochrome P-450 3A-mediated human liver microsomal metabolism of a novel HIV protease inhibitor by ritonavir: A positive drug-drug interaction.
Dykstra, J; Granneman, GR; Hickman, D; Jayanti, VK; Kumar, GN; Roberts, EM; Surber, B; Thomas, S; Uchic, J; Yao, Y, 1999
)
0.51
" Our results suggest that the response to SQV combined with RTV therapy is complicated by previous long-term treatment with PIs, probably owing to multiple PI resistance mutations."( Anti-HIV effect of saquinavir combined with ritonavir is limited by previous long-term therapy with protease inhibitors.
Aizawa, S; Gatanaga, H; Genka, I; Kikuchi, Y; Oka, S; Tachikawa, N; Yamamoto, Y; Yasuoka, A; Yoshizawa, S, 1999
)
0.56
" Saquinavir (SQV) is used alone or in combination with ritonavir (RTV) or nelfinavir (NLF), respectively, in the context of the HAART drug regimen."( Therapeutic drug monitoring of saquinavir in patients during protease inhibitor therapy with saquinavir alone or in combination with ritonavir or nelfinavir.
Klinker, H; Langmann, P; Richter, E; Schlör, C; Väth, T; Weissbrich, B; Zilly, M, 2000
)
0.76
"A retrospective study of 76 patients was carried out using ritonavir in an antiretroviral regimen combined with two reverse transcriptase inhibitors to treat outpatients from July, 1996, to April, 1998, with the objective of evaluating clinical efficacy and tolerability."( Clinical evaluation of the efficacy and tolerability of ritonavir in combination with two reverse transcriptase inhibitors.
Ary Duque, I; Gomes de Arruda, EA; Martins Maia, F; Pedrosa De Oliveira Júnior, FR, 2000
)
0.8
" These drug-drug interactions were not expected, the mechanism(s) is (are) not clear, and additional studies are warranted."( Competing drug-drug interactions among multidrug antiretroviral regimens used in the treatment of HIV- infected subjects: ACTG 884.
Acosta, EP; Cheng, H; Fischl, M; Fletcher, CV; Gulick, RM; Haubrich, R; Hu, XJ; Katzenstein, D; Mills, C; Raasch, R; Remmel, RP, 2000
)
0.31
" Therefore a number of drug-drug interactions are likely to occur."( [Drug interactions with antiretroviral agents].
Furlan, V; Taburet, AM,
)
0.13
"St John's Wort (SJW) is widely used in the treatment of depression but concerns have been raised about its potential to interact with other drugs."( St Johns wort increases expression of P-glycoprotein: implications for drug interactions.
Back, D; Barry, M; Feely, J; Hennessy, M; Kavanagh, P; Kelleher, D; Mulcahy, F; Spiers, JP, 2002
)
0.31
"To determine the long-term antiretroviral efficacy and tolerability of dual protease inhibitor (PI) therapy with indinavir (IDV)/ritonavir (RTV) at 400/400 mg twice a day (BID) in combination with two nucleoside reverse trancriptase inhibitors (NRTIs)."( Long-term efficacy and safety of ritonavir/indinavir at 400/400 mg twice a day in combination with two nucleoside reverse transcriptase inhibitors as first line antiretroviral therapy.
Bergmann, F; Carls, H; Fätkenheuer, G; Fenske, S; Knechten, H; Lichterfeld, M; Nadler, M; Nischalke, HD; Oette, M; Rieke, A; Rockstroh, JK; Theisen, A; Wasmuth, JC; Wiesel, W, 2002
)
0.8
"The steady-state pharmacokinetics and pharmacodynamics of two oral doses of lopinavir-ritonavir (lopinavir/r; 400/100 and 533/133 mg) twice daily (BID) when dosed in combination with efavirenz, plus two nucleoside reverse transcriptase inhibitors, were assessed in a phase II, open-label, randomized, parallel arm study in 57 multiple protease inhibitor-experienced but non-nucleoside reverse transcriptase inhibitor-naive human immunodeficiency virus (HIV)-infected subjects."( Pharmacokinetic-pharmacodynamic analysis of lopinavir-ritonavir in combination with efavirenz and two nucleoside reverse transcriptase inhibitors in extensively pretreated human immunodeficiency virus-infected patients.
Bernstein, B; Bertz, R; Brun, S; Foit, C; Granneman, GR; Hsu, A; Isaacson, J; Kempf, DJ; King, M; Lam, W; Richards, B; Rode, R; Rynkiewicz, K; Sun, E, 2003
)
0.79
"ESS40011 was a 24-week, multicenter, open-label, clinical trial in which antiretroviral therapy-naïve and -experienced HIV-1-infected adults were randomized 3:1 to receive either APV600/RTV BID or APV1200 BID, in combination with > or = 2 non-protease inhibitor antiretroviral drugs."( Twice-daily amprenavir 1200 mg versus amprenavir 600 mg/ritonavir 100 mg, in combination with at least 2 other antiretroviral drugs, in HIV-1-infected patients.
Gathe, JC; Griffith, S; Hernandez, JE; Lancaster, CT; Liao, Q; Nadler, JP; Pappa, KA; Pollard, RB; Richmond, GJ, 2003
)
0.57
"To evaluate the long-term efficacy and pharmacokinetics of indinavir (IDV)/ritonavir (RTV) 400/100 mg twice a day in combination with two nucleoside reverse transcriptase inhibitors."( Low-dose indinavir in combination with low-dose ritonavir: steady-state pharmacokinetics and long-term clinical outcome follow-up.
Justesen, US; Levring, AM; Lindberg, JA; Pedersen, C; Tauris, P; Thomsen, A, 2003
)
0.8
" When combined with ritonavir (RTV), plasma concentration of SQV is increased."( [Comparison of pharmacokinetics of saquinavir soft-gel capsule (SQV-SGC) combined with ritonavir (RTV), SQV hard-gel capsule with RTV, and SQV-SGC alone].
Genka, I; Hirabayashi, Y; Imai, K; Kikuchi, Y; Kimura, S; Oka, S; Tachikawa, N; Teruya, K; Tsuchiya, K; Yasuoka, A, 2003
)
0.87
" This study evaluated the steady-state pharmacokinetics of indinavir/ritonavir at 800/100 mg twice daily (bid) in combination with efavirenz at 600 mg once daily (qd) in HIV-infected Thai subjects who used this nucleoside-sparing combination in The HIV Netherlands Australia Thailand Research Collaboration 009 study."( Pharmacokinetics of indinavir/ritonavir (800/100 mg) in combination with efavirenz (600 mg) in HIV-1-infected subjects.
Aarnoutse, RE; Boyd, MA; Burger, DM; Cooper, DA; Lange, JM; Phanuphak, P; Ruxrungtham, K; Stek, M; van Heeswijk, RP, 2003
)
0.84
"Randomized, controlled, open-label trial of 233 protease inhibitor- and non-nucleoside reverse transcriptase inhibitor-naive HIV-infected patients allocated to a regimen of nelfinavir and nevirapine (1250/200 mg twice daily; n = 118) or ritonavir and saquinavir (400/400 mg twice daily; n = 115), both in combination with two nucleoside reverse transcriptase inhibitors."( A randomized trial comparing initial HAART regimens of nelfinavir/nevirapine and ritonavir/saquinavir in combination with two nucleoside reverse transcriptase inhibitors.
Gerstoft, J; Katzenstein, TL; Kirk, O; Lundgren, JD; Mathiesen, LR; Nielsen, H; Obel, N; Pedersen, C, 2003
)
0.73
"A regimen of nelfinavir/nevirapine had a favourable virological effect and tolerability over a 48-week period compared with ritonavir/saquinavir, when administered in combination with two nucleoside reverse transcriptase inhibitors."( A randomized trial comparing initial HAART regimens of nelfinavir/nevirapine and ritonavir/saquinavir in combination with two nucleoside reverse transcriptase inhibitors.
Gerstoft, J; Katzenstein, TL; Kirk, O; Lundgren, JD; Mathiesen, LR; Nielsen, H; Obel, N; Pedersen, C, 2003
)
0.75
"To evaluate the efficacy and tolerability of indinavir/ritonavir (IDV/RTV) 400/100 mg twice daily in combination with two nucleoside reverse transcriptase inhibitors in antiretroviral-naive patients."( Efficacy and safety of ritonavir/indinavir 100/400 mg twice daily in combination with two nucleoside analogues in antiretroviral treatment-naive HIV-infected individuals.
Agher, R; Ait-Mohand, H; Bricaire, F; Calvez, V; Costagliola, D; Duvivier, C; Ghosn, J; Katlama, C; Marcelin, AG; Myrto, A; Peytavin, G; Schneider, L, 2003
)
0.88
"We describe a drug-drug interaction between coformulated lopinavir/ritonavir and itraconazole in a patient infected with human immunodeficiency virus type 1 who had disseminated histoplasmosis."( Drug-drug interaction between itraconazole and the antiretroviral drug lopinavir/ritonavir in an HIV-1-infected patient with disseminated histoplasmosis.
Beijnen, JH; Crommentuyn, KM; Huitema, AD; Mulder, JW; Schellens, JH; Sparidans, RW, 2004
)
0.79
" In the present study, two adjusted-dose regimens of lopinavir-ritonavir were tested in combination with rifampin."( Pharmacokinetics of adjusted-dose lopinavir-ritonavir combined with rifampin in healthy volunteers.
Bertz, R; Boeree, MJ; Burger, DM; Colbers, EP; Hekster, YA; Koopmans, PP; la Porte, CJ; Voncken, DS; Wikstrom, K, 2004
)
0.82
"So far, no pediatric doses for indinavir combined with ritonavir have been defined."( Pharmacokinetics of indinavir combined with low-dose ritonavir in human immunodeficiency virus type 1-infected children.
Bergshoeff, AS; Burger, DM; de Groot, R; Fraaij, PL; Leavitt, RY; Nguyen, BY; van Rossum, AM; Verweel, G; Winchell, GA; Wynne, LH, 2004
)
0.82
" The tissue distribution of radioactivity was examined in rats dosed with [14C]lopinavir in combination with ritonavir."( Metabolism and disposition of the HIV-1 protease inhibitor lopinavir (ABT-378) given in combination with ritonavir in rats, dogs, and humans.
Denissen, JF; Grabowski, BA; Jayanti, VK; Johnson, MK; Kempf, DJ; Kumar, GN; Lee, RD; Marsh, KC; Roberts, SA; Sham, HL; Sun, E; Thomas, S; Uchic, J, 2004
)
0.75
"The aim of the study was to characterize the population pharmacokinetics of indinavir, define the relationship between the pharmacokinetics of indinavir and ritonavir, and to identify the factors influencing the pharmacokinetics of indinavir alone or when given with ritonavir."( Population pharmacokinetics of indinavir alone and in combination with ritonavir in HIV-1-infected patients.
Beijnen, JH; Huitema, AD; Kappelhoff, BS; Meenhorst, PL; Mulder, JW; Prins, JM; Sankatsing, SU; Van Gorp, EC, 2005
)
0.76
"To develop a population pharmacokinetic model for lopinavir in combination with ritonavir, in which the interaction between both drugs was characterized, and in which relationships between patient characteristics and pharmacokinetics were identified."( Population pharmacokinetics of lopinavir in combination with ritonavir in HIV-1-infected patients.
Beijnen, JH; Crommentuyn, KM; Huitema, AD; Kappelhoff, BS; Mairuhu, AT; Meenhorst, PL; Mulder, JW; van Gorp, EC, 2005
)
0.8
"To compare the effect of ritonavir on plasma amprenavir pharmacokinetics, healthy adults received either fosamprenavir (700 mg twice a day [BID]) or amprenavir (600 mg BID) alone and in combination with ritonavir (100 mg BID)."( Ritonavir increases plasma amprenavir (APV) exposure to a similar extent when coadministered with either fosamprenavir or APV.
Baker, KL; Berrey, MM; Jones, LS; Lou, Y; Shelton, MJ; Thomas, GJ; Wire, MB, 2006
)
2.08
" Fosamprenavir-ritonavir has shown similar efficacy and safety to lopinavir-ritonavir when each is combined with two nucleoside reverse transcriptase inhibitors."( The KLEAN study of fosamprenavir-ritonavir versus lopinavir-ritonavir, each in combination with abacavir-lamivudine, for initial treatment of HIV infection over 48 weeks: a randomised non-inferiority trial.
DeJesus, E; Eron, J; Estrada, V; Gathe, J; Katlama, C; Lackey, P; Patel, L; Shaefer, M; Staszewski, S; Sutherland-Phillips, D; Vavro, C; Wannamaker, P; Yau, L; Yeni, P; Yeo, J; Young, B, 2006
)
0.97
"Fosamprenavir-ritonavir twice daily in treatment-naive patients provides similar antiviral efficacy, safety, tolerability, and emergence of resistance as lopinavir-ritonavir, each in combination with abacavir-lamivudine."( The KLEAN study of fosamprenavir-ritonavir versus lopinavir-ritonavir, each in combination with abacavir-lamivudine, for initial treatment of HIV infection over 48 weeks: a randomised non-inferiority trial.
DeJesus, E; Eron, J; Estrada, V; Gathe, J; Katlama, C; Lackey, P; Patel, L; Shaefer, M; Staszewski, S; Sutherland-Phillips, D; Vavro, C; Wannamaker, P; Yau, L; Yeni, P; Yeo, J; Young, B, 2006
)
0.98
" Together, these in vitro results indicate that combination with other antiretrovirals does not significantly increase the toxic potential of TFV in RPTECs."( In vitro cytotoxicity and mitochondrial toxicity of tenofovir alone and in combination with other antiretrovirals in human renal proximal tubule cells.
Alvarez, ML; Cihlar, T; Cordobilla, B; Domingo, JC; Domingo, P; Giralt, M; Guallar, J; López-Dupla, M; Sánchez de la Rosa, R; Saumoy, M; Torres, F; Vidal, F; Villarroya, F, 2006
)
0.33
" This unexpected drug-drug interaction warrants further investigation."( Unexpected drug-drug interaction between tipranavir/ritonavir and enfuvirtide.
Bargiacchi, O; Bonora, S; Calcagno, A; D'Avolio, A; Di Perri, G; González de Requena, D; Ladetto, L; Sciandra, M; Siccardi, M; Sinicco, A, 2006
)
0.58
" No correlation was observed between the ABCB1 C3435T, G2677T/A, and C1236T polymorphisms, separately and in haplotypes, with saquinavir pharmacokinetics, administered with or without ritonavir and with PBMC P-gp expression and activity."( The effect of ABCB1 polymorphism on the pharmacokinetics of saquinavir alone and in combination with ritonavir.
Béïque, L; Cameron, DW; Chauhan, B; Foster, BC; Garber, GE; la Porte, CJ; Li, Y; van Heeswijk, RP, 2007
)
0.75
"The pharmacokinetic and metabolite profiles of the antiretroviral agent tipranavir (TPV), administered with ritonavir (RTV), in nine healthy male volunteers were characterized."( Steady-state disposition of the nonpeptidic protease inhibitor tipranavir when coadministered with ritonavir.
Brinkman, A; Castles, M; Chen, L; Garfinkel, S; MacGregor, TR; Mao, Y; Norris, SH; Philip, E; Sabo, JP; Valdez, H; Wruck, JM, 2007
)
0.77
" Tipranavir is metabolized by cytochrome P450 (CYP) 3A and, when combined with ritonavir in vitro, causes inhibition of CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A in addition to induction of glucuronidase and the drug transporter P-glycoprotein."( Mechanisms of pharmacokinetic and pharmacodynamic drug interactions associated with ritonavir-enhanced tipranavir.
Kashuba, AD; Vourvahis, M, 2007
)
0.79
"Our objective was to identify possible differences in protease inhibitor plasma concentrations between and within three protease inhibitor regimens (indinavir, saquinavir and lopinavir all in combination with low-dose ritonavir) and to relate these differences to safety and efficacy."( Pharmacokinetics of two randomized trials evaluating the safety and efficacy of indinavir, saquinavir and lopinavir in combination with low-dose ritonavir: the MaxCmin1 and 2 trials.
Cahn, P; Castagna, A; Clumeck, N; Dragsted, UB; Fox, Z; Gerstoft, J; Justesen, US; Losso, M; Lundgren, JD; Obel, N; Pedersen, C; Peters, B, 2007
)
0.73
"7-fold in combination with lopinavir/ritonavir."( Drug/Drug interaction between lopinavir/ritonavir and rosuvastatin in healthy volunteers.
Flynn, DM; Gerber, JG; Hoody, DW; Kiser, JJ; Predhomme, JA; Wolfe, P, 2008
)
0.89
"51 examined the pharmacokinetic profile, safety, and efficacy of RTV-boosted tipranavir (TPV/r), alone and in combination with comparator PIs (CPIs) in 315 triple-class-experienced, HIV-infected patients."( Pharmacokinetics, safety, and efficacy of tipranavir boosted with ritonavir alone or in combination with other boosted protease inhibitors as part of optimized combination antiretroviral therapy in highly treatment-experienced patients (BI Study 1182.51).
Arestéh, K; Blick, G; Johnson, M; Katlama, C; Lazzarin, A; Leith, JG; MacGregor, TR; Meier, U; Pierone, G; Walmsley, SL, 2008
)
0.58
"The effects of omeprazole on indinavir when administered alone or in combination with ritonavir were evaluated."( Effect of omeprazole on the plasma concentrations of indinavir when administered alone and in combination with ritonavir.
Donovan, BJ; Hollowell, SB; Kashuba, AD; Min, SS; Raasch, RH; Rezk, NL; Rublein, JC; Smith, PC; Tallman, MN; Tappouni, HL; Theodore, D; Tien, HC, 2008
)
0.78
" After seven days, the single-dose pharmacokinetic profile of an 800-mg dose of indinavir alone or in combination with 200 mg of ritonavir was evaluated."( Effect of omeprazole on the plasma concentrations of indinavir when administered alone and in combination with ritonavir.
Donovan, BJ; Hollowell, SB; Kashuba, AD; Min, SS; Raasch, RH; Rezk, NL; Rublein, JC; Smith, PC; Tallman, MN; Tappouni, HL; Theodore, D; Tien, HC, 2008
)
0.76
" The addition of 200 mg of ritonavir to 800 mg of indinavir in combination with 40 mg of omeprazole significantly increased the mean indinavir AUC from 30."( Effect of omeprazole on the plasma concentrations of indinavir when administered alone and in combination with ritonavir.
Donovan, BJ; Hollowell, SB; Kashuba, AD; Min, SS; Raasch, RH; Rezk, NL; Rublein, JC; Smith, PC; Tallman, MN; Tappouni, HL; Theodore, D; Tien, HC, 2008
)
0.85
"HIV-1-infected men enrolled in the Monark randomized trial were eligible for the present study after 48 weeks of a first-line lopinavir/ritonavir alone or in combination with zidovudine and lamivudine."( Absence of HIV-1 shedding in male genital tract after 1 year of first-line lopinavir/ritonavir alone or in combination with zidovudine/lamivudine.
Bresson, JL; Chaix, ML; Cohen-Codar, I; Delfraissy, JF; Galimand, J; Ghosn, J; Girard, PM; Peytavin, G; Raffi, F; Rouzioux, C, 2008
)
0.77
"In vitro assessment of drug candidates' affinity for multi-drug resistance proteins is of crucial importance for the prediction of in vivo pharmacokinetics and drug-drug interactions."( Characterization of substrates and inhibitors for the in vitro assessment of Bcrp mediated drug-drug interactions.
Gnoth, MJ; Grieshop, B; Ickenroth, K; Muenster, U, 2008
)
0.35
" In order to not overlook potential drug-drug interactions when testing drug candidates for inhibitory potential towards Bcrp, distinct Bcrp probe substrates should be used."( Characterization of substrates and inhibitors for the in vitro assessment of Bcrp mediated drug-drug interactions.
Gnoth, MJ; Grieshop, B; Ickenroth, K; Muenster, U, 2008
)
0.35
" Patients with HIV may also have simultaneous chronic medical conditions, resulting in the possibility of complex drug-drug interactions."( Possible antiretroviral therapy-warfarin drug interaction.
Fulco, PP; Higginson, RT; Zingone, MM, 2008
)
0.35
"To characterize the cytochrome P450 enzyme(s) responsible for the N-dealkylation of maraviroc in vitro, and predict the extent of clinical drug-drug interactions (DDIs)."( Maraviroc: in vitro assessment of drug-drug interaction potential.
Collins, C; Dickins, M; Hyland, R; Jones, B; Jones, H, 2008
)
0.35
"In this open-label, international non-inferiority study, 883 antiretroviral-naive, HIV-1-infected patients were randomly assigned to receive atazanavir/ritonavir 300/100 mg once daily (n=440) or lopinavir/ritonavir 400/100 mg twice daily (n=443), in combination with fixed-dose tenofovir/emtricitabine 300/200 mg once daily."( Once-daily atazanavir/ritonavir versus twice-daily lopinavir/ritonavir, each in combination with tenofovir and emtricitabine, for management of antiretroviral-naive HIV-1-infected patients: 48 week efficacy and safety results of the CASTLE study.
Andrade-Villanueva, J; Chetchotisakd, P; Corral, J; David, N; Echevarria, J; Mancini, M; McGrath, D; Molina, JM; Moyle, G; Percival, L; Thiry, A; Yang, R, 2008
)
0.86
" This prospective, open-label, 24-week, single-arm trial assessed the efficacy and safety of enfuvirtide (90 mg injected subcutaneously twice daily) in combination with darunavir-ritonavir (600/100 mg administered orally twice daily) in triple-antiretroviral-class-experienced adults failing their current regimen."( Safety and efficacy of enfuvirtide in combination with darunavir-ritonavir and an optimized background regimen in treatment-experienced human immunodeficiency virus-infected patients: the below the level of quantification study.
DeJesus, E; Gathe, JC; Gottlieb, MS; Greenberg, ML; Guittari, CJ; Zolopa, AR, 2008
)
0.78
" In study arm 1, 20 subjects received saquinavir/ritonavir treatment alone for 14 days, followed in combination with ketoconazole treatment for 14 days."( Drug-drug interaction study of ketoconazole and ritonavir-boosted saquinavir.
Bour, F; Kaeser, B; Schmitt, C; Zandt, H; Zhang, X; Zwanziger, E, 2009
)
0.86
"A total of 191 patients were randomized 2:2:2:1 to one of three APL dosing regimens or to lamivudine (3TC)/zidovudine (ZDV) twice daily (bid), each in combination with lopinavir/ritonavir (LPV/r) 400 mg/100 mg bid."( Antiviral activity and safety of aplaviroc, a CCR5 antagonist, in combination with lopinavir/ritonavir in HIV-infected, therapy-naïve patients: results of the EPIC study (CCR100136).
Adkison, KK; Berger, D; Bonny, T; Cimoch, P; Lamarca, A; Lazzarin, A; Madison, SJ; McCarty, D; Millard, J; Nichols, WG; Salvato, P; Smaill, FM; Teofilo, E; Yeni, P, 2009
)
0.77
"This 96-week, open-label, randomized study assessed changes in body composition in treatment-naive patients infected with human immunodeficiency virus type 1 who were treated with either atazanavir or ritonavir-boosted atazanavir, in combination with stavudine and lamivudine."( Changes in body composition with ritonavir-boosted and unboosted atazanavir treatment in combination with Lamivudine and Stavudine: a 96-week randomized, controlled study.
Mathew, M; McComsey, G; McGrath, D; Rightmire, A; Wirtz, V; Yang, R, 2009
)
0.82
"Concomitant use of immunosuppressive agents and antiretroviral drugs may lead to complex drug-drug interactions."( Drug-drug interaction in a kidney transplant recipient receiving HIV salvage therapy and tacrolimus.
Battegay, M; Marzolini, C; Mayr, M; Mertz, D, 2009
)
0.35
"The KLEAN study extension assessed the long-term efficacy and safety of fosamprenavir-ritonavir (FPV/r) and lopinavir-ritonavir (LPV/r), both administered with abacavir/lamivudine (ABC/3TC) fixed dose combination, over 144 weeks."( Long-term efficacy and safety of fosamprenavir plus ritonavir versus lopinavir/ritonavir in combination with abacavir/lamivudine over 144 weeks.
Baril, JG; Duiculescu, D; Estrada, V; Lim, ML; Logue, K; Pharo, C; Plettenberg, A; Pulido, F; Schewe, K; Vavro, C; Yau, L,
)
0.61
"The findings of the KLEAN study extension (48 to 144 weeks) support durable viral suppression with both FPV/r and LPV/r treatment regimens when used in combination with ABC/3TC irrespective of viral load at baseline."( Long-term efficacy and safety of fosamprenavir plus ritonavir versus lopinavir/ritonavir in combination with abacavir/lamivudine over 144 weeks.
Baril, JG; Duiculescu, D; Estrada, V; Lim, ML; Logue, K; Pharo, C; Plettenberg, A; Pulido, F; Schewe, K; Vavro, C; Yau, L,
)
0.38
"Clinicians caring for patients infected with the human immunodeficiency virus (HIV) and diagnosed with psychiatric comorbidities must be aware of potential drug-drug interactions, particularly with protease inhibitor-based antiretroviral therapy."( Clinically significant adverse events from a drug interaction between quetiapine and atazanavir-ritonavir in two patients.
McCoy, C; Pollack, TM; Stead, W, 2009
)
0.57
"Similar virological efficacy was observed for efavirenz and lopinavir/r, when administered with Kivexa in antiretroviral-naïve patients, while immunological improvement was slightly superior for efavirenz."( Similar antiviral efficacy and tolerability between efavirenz and lopinavir/ritonavir, administered with abacavir/lamivudine (Kivexa), in antiretroviral-naïve patients: a 48-week, multicentre, randomized study (Lake Study).
Bravo, I; Carosi, G; Clotet, B; del Arco, A; Echeverría, P; Gálvez, J; Gómez, JL; López, JC; López-Blazquez, R; Mariño, A; Moreno, A; Negredo, E; Ocampo, A; Pedrol, E; Pérez-Alvarez, N; Portilla, J; Prieto, A; Rubio, R; Viladés, C, 2010
)
0.59
"International, multicenter, open-label, 96-week noninferiority randomized trial of atazanavir/ritonavir 300/100 mg once daily vs lopinavir/ritonavir 400/100 mg twice daily, each in combination with fixed-dose tenofovir/emtricitabine 300/200 mg once daily, in antiretroviral-naive, HIV-1-infected patients."( Once-daily atazanavir/ritonavir compared with twice-daily lopinavir/ritonavir, each in combination with tenofovir and emtricitabine, for management of antiretroviral-naive HIV-1-infected patients: 96-week efficacy and safety results of the CASTLE study.
Absalon, J; Andrade-Villanueva, J; Chetchotisakd, P; Corral, J; David, N; Echevarria, J; Lataillade, M; Mancini, M; McGrath, D; Molina, JM; Moyle, G; Percival, L; Wirtz, V; Yang, R, 2010
)
0.89
" Because OATP2B1 exhibits an increasing number of drug substrates, including several statins, alterations of its function by PIs could result in clinically significant drug-drug interactions in the intestine."( pH dependence of organic anion-transporting polypeptide 2B1 in Caco-2 cells: potential role in antiretroviral drug oral bioavailability and drug-drug interactions.
Bendayan, R; Kis, O; Ramaswamy, M; Zastre, JA, 2010
)
0.36
"Increasing the ritonavir dose to achieve a lopinavir/ritonavir ratio of 1:1 when given in combination with rifampicin-based TB treatment did not completely compensate for the enhancement of lopinavir CL/F caused by rifampicin."( Population pharmacokinetics of lopinavir in combination with rifampicin-based antitubercular treatment in HIV-infected South African children.
Elsherbiny, D; Maartens, G; McIlleron, H; Ren, Y; Simonsson, US, 2010
)
0.71
" In period 2, after a 4-week washout, the same dose and regimen of narlaprevir was administered in combination with PEG-IFN-α-2b for 14 days."( Antiviral activity of narlaprevir combined with ritonavir and pegylated interferon in chronic hepatitis C patients.
Bergmann, JF; de Bruijne, J; de Knegt, RJ; Hughes, EA; Janssen, HL; Li, J; Molenkamp, R; Reesink, HW; Schinkel, J; Tong, X; Treitel, MA; van Lier, JJ; van Vliet, AA; Weegink, CJ, 2010
)
0.62
"The aim of this review is to discuss the effect of pharmacokinetic drug-drug interactions (DDIs) in the antiretroviral treatment of HIV infection."( Drug-drug interactions in the treatment of HIV infection: focus on pharmacokinetic enhancement through CYP3A inhibition.
Josephson, F, 2010
)
0.36
"The aim of this open-label, fixed-sequence study was to investigate the potential of Echinacea purpurea, a commonly used botanical supplement, to interact with the boosted protease inhibitor darunavir-ritonavir."( Herb-drug interaction between Echinacea purpurea and darunavir-ritonavir in HIV-infected patients.
Barbanoj, MJ; Cedeño, S; Clotet, B; Miranda, C; Moltó, J; Negredo, E; Valle, M, 2011
)
0.8
" No adjustment of the saquinavir-ritonavir dose (1,000/100 mg) BID is required when the drugs are administered in combination with rifabutin."( Pharmacokinetic interaction study of ritonavir-boosted saquinavir in combination with rifabutin in healthy subjects.
Fettner, S; Rowell, L; Salgo, M; Zhang, X; Zwanziger, E, 2011
)
0.92
" It is by this mechanism that most HIV protease inhibitors, non-nucleoside reverse transcriptase inhibitors and maraviroc often interact with other medications."( Approaches for understanding and predicting drug interactions in human immunodeficiency virus-infected patients.
Alvarez, E; de Andrés, S; Jiménez-Nácher, I; Morello, J; Rodriguez-Nóvoa, S; Soriano, V, 2011
)
0.37
"Conflicting drug-drug interaction (DDI) studies with the HIV protease inhibitors (PIs) suggest net induction or inhibition of intestinal or hepatic CYP3A."( Complex drug interactions of HIV protease inhibitors 1: inactivation, induction, and inhibition of cytochrome P450 3A by ritonavir or nelfinavir.
Collier, AC; Kharasch, ED; Kirby, BJ; Thummel, KE; Unadkat, JD; Whittington, D, 2011
)
0.58
" nevirapine on the same background, in naïve HIV-1-infected patients) study compared prospectively ritonavir-boosted atazanavir (ATZ/r) 300 mg/100 mg once daily (qd) with immediate release nevirapine (NVP) 200 mg twice daily or 400 mg qd, each combined with fixed-dose tenofovir 300 mg/emtricitabine 200 mg qd in 569 ARV-naïve HIV-1-infected patients."( Lipid profiles for nevirapine vs. atazanavir/ritonavir, both combined with tenofovir disoproxil fumarate and emtricitabine over 48 weeks, in treatment-naïve HIV-1-infected patients (the ARTEN study).
Andrade-Villanueva, J; Cairns, V; Clotet, B; de Rossi, L; Domingo, P; Gellermann, HJ; Podzamczer, D; Reiss, P; Rockstroh, JK; Soriano, V; Taylor, S, 2011
)
0.85
" This finding suggests a potential significant drug-drug interaction between these two drugs."( Ritonavir inhibits the two main prasugrel bioactivation pathways in vitro: a potential drug-drug interaction in HIV patients.
Ancrenaz, V; Bosilkovska, M; Daali, Y; Dayer, P; Desmeules, J, 2011
)
1.81
"ARTEN is a randomized, open-label, non-inferiority trial that compares nevirapine (NVP) 200 mg twice daily or 400 mg once daily to atazanavir/ritonavir (ATZ/r) 300 mg/100 mg once daily, each combined with fixed-dose tenofovir disoproxil fumarate (TDF) 300 mg/emtricitabine (FTC) 200 mg once daily, in antiretroviral-naive HIV-1 patients with CD4(+) T-cell counts <400 (men) and <250 cells/mm(3) (women)."( Nevirapine versus atazanavir/ritonavir, each combined with tenofovir disoproxil fumarate/emtricitabine, in antiretroviral-naive HIV-1 patients: the ARTEN Trial.
Andrade-Villanueva, J; Antunes, F; Arastéh, K; de Rossi, L; Di Perri, G; Domingo, P; Gellermann, H; Lutz, T; Migrone, H; Opravil, M; Podzamczer, D; Soriano, V; Taylor, S, 2011
)
0.86
"NVP demonstrated at week 48 non-inferior antiviral efficacy compared with ATZ/r when given along with TDF/FTC, despite more drug-related discontinuations with NVP than ATZ/r."( Nevirapine versus atazanavir/ritonavir, each combined with tenofovir disoproxil fumarate/emtricitabine, in antiretroviral-naive HIV-1 patients: the ARTEN Trial.
Andrade-Villanueva, J; Antunes, F; Arastéh, K; de Rossi, L; Di Perri, G; Domingo, P; Gellermann, H; Lutz, T; Migrone, H; Opravil, M; Podzamczer, D; Soriano, V; Taylor, S, 2011
)
0.66
"Treatment of HIV/tuberculosis (TB) co-infected patients is complex due to drug-drug interactions for these chronic diseases."( Determination of rifabutin dosing regimen when administered in combination with ritonavir-boosted atazanavir.
Arikan, D; Bertz, R; Coumbis, J; Farajallah, A; Stonier, M; Wu, Y; Xu, X; Zhang, J; Zhu, L, 2011
)
0.6
" The study was stopped because subjects experienced more severe declines in neutrophil counts when rifabutin was given with atazanavir/ritonavir than alone."( Determination of rifabutin dosing regimen when administered in combination with ritonavir-boosted atazanavir.
Arikan, D; Bertz, R; Coumbis, J; Farajallah, A; Stonier, M; Wu, Y; Xu, X; Zhang, J; Zhu, L, 2011
)
0.8
" The lopinavir/ritonavir dose of 500/125 mg bid administered with efavirenz most closely approximates the pharmacokinetic exposure of lopinavir/ritonavir 400/100 mg bid administered alone."( Pharmacokinetics and safety of the lopinavir/ritonavir tablet 500/125 mg twice daily coadministered with efavirenz in healthy adult participants.
Awni, W; Bernstein, B; Causemaker, SJ; Chiu, YL; Klein, CE; Ng, J, 2012
)
0.99
"Efficacy results were consistent with the ARTEN study demonstrating that NVP was non-inferior to ATV/r when taken in combination with TDF/FTC."( A randomised comparison of safety and efficacy of nevirapine vs. atazanavir/ritonavir combined with tenofovir/emtricitabine in treatment-naïve patients.
Bhatti, L; Conner, C; Dejesus, E; Mills, A; Storfer, S, 2011
)
0.6
" The aim of this study was to develop an integrated population pharmacokinetic model accounting for the drug-drug interactions between lopinavir, ritonavir and rifampicin, and to evaluate optimal doses of lopinavir/ritonavir when co-administered with rifampicin."( Model-based approach to dose optimization of lopinavir/ritonavir when co-administered with rifampicin.
Decloedt, E; Denti, P; Karlsson, MO; Maartens, G; McIlleron, H; Simonsson, US; Zhang, C, 2012
)
0.83
"The model describes the drug-drug interactions between lopinavir, ritonavir and rifampicin in adults."( Model-based approach to dose optimization of lopinavir/ritonavir when co-administered with rifampicin.
Decloedt, E; Denti, P; Karlsson, MO; Maartens, G; McIlleron, H; Simonsson, US; Zhang, C, 2012
)
0.86
"As part of a larger clinical drug-drug interaction (DDI) study aimed at in vitro to in vivo prediction of HIV protease inhibitor metabolic and transporter-based DDIs, we measured the inductive (staggered administration) and inductive plus inhibitory (simultaneously administered) effect of multiple dose ritonavir (RTV), nelfinavir (NFV), or rifampin (RIF) on the pharmacokinetics of the P-glycoprotein probe, digoxin (DIG), when administered simultaneously or staggered with the protease inhibitors or RIF."( Complex drug interactions of the HIV protease inhibitors 3: effect of simultaneous or staggered dosing of digoxin and ritonavir, nelfinavir, rifampin, or bupropion.
Collier, AC; Kharasch, ED; Kirby, BJ; Thummel, KE; Unadkat, JD; Whittington, D, 2012
)
0.76
" Mean plasma concentration-time profiles for atazanavir, tenofovir disoproxil fumarate/emtricitabine (TDF/FTC), darunavir (DRV, administered with ritonavir [RTV]), and drospirenone/ethinylestradiol were similar following co-administration of GSK2248761."( Drug interaction profile for GSK2248761, a next generation non-nucleoside reverse transcriptase inhibitor.
de Serres, M; Gould, E; Johnson, M; Kim, J; Lou, Y; Mayers, D; Pietropaolo, K; Piscitelli, S; White, S; Zhou, XJ, 2012
)
0.58
"A multiple dose, parallel group study was conducted to assess for a drug-drug interaction between the pyronaridine/artesunate (PA) combination antimalarial and ritonavir."( Drug-drug interaction analysis of pyronaridine/artesunate and ritonavir in healthy volunteers.
Borghini-Fuhrer, I; Duparc, S; Fleckenstein, L; Jung, D; Lopez-Lazaro, L; Methaneethorn, J; Morris, CA; Pokorny, R; Shin, CS, 2012
)
0.82
"The hepatic organic anion transporting polypeptides (OATPs) influence the pharmacokinetics of several drug classes and are involved in many clinical drug-drug interactions."( Classification of inhibitors of hepatic organic anion transporting polypeptides (OATPs): influence of protein expression on drug-drug interactions.
Artursson, P; Haglund, U; Karlgren, M; Kimoto, E; Lai, Y; Norinder, U; Vildhede, A; Wisniewski, JR, 2012
)
0.38
" Ritonavir, an HIV-1 protease inhibitor (PI) and potent CYP3A inhibitor, is used as a pharmacokinetic enhancer at subtherapeutic doses in combination with other HIV PIs."( Impact of low-dose ritonavir on danoprevir pharmacokinetics: results of computer-based simulations and a clinical drug-drug interaction study.
Blotner, S; Chen, Y; Fretland, J; Haznedar, JO; Reddy, MB; Smith, P; Tran, JQ, 2012
)
1.62
" Following results from this drug-drug interaction (DDI) model, a crossover study was performed in healthy volunteers to investigate the effects of acute and repeat dosing of low-dose ritonavir on danoprevir single-dose pharmacokinetics."( Impact of low-dose ritonavir on danoprevir pharmacokinetics: results of computer-based simulations and a clinical drug-drug interaction study.
Blotner, S; Chen, Y; Fretland, J; Haznedar, JO; Reddy, MB; Smith, P; Tran, JQ, 2012
)
0.9
"To explore the durability of three first-line tenofovir/emtricitabine-based regimens in combination with atazanavir/ritonavir, efavirenz or lopinavir/ritonavir in HIV-1-infected patients."( Duration of first-line antiretroviral therapy with tenofovir and emtricitabine combined with atazanavir/ritonavir, efavirenz or lopinavir/ritonavir in the Italian ARCA cohort.
Borghi, V; Capetti, A; Cicconi, P; Di Biagio, A; Di Giambenedetto, S; Francisci, D; Giacometti, A; Giannarelli, D; Maggiolo, F; Monno, L; Penco, G; Prinapori, R; Sterrantino, G; Zoncada, A, 2013
)
0.81
"Interspecies differences in drug metabolism have made it difficult to use preclinical animal testing data to predict the drug metabolites or potential drug-drug interactions (DDIs) that will occur in humans."( Using chimeric mice with humanized livers to predict human drug metabolism and a drug-drug interaction.
Angus, P; Chen, Y; Elazar, M; Fitch, WL; Glenn, J; Hu, Y; Idilman, R; Liu, M; Murphy, B; Nakamura, M; Nishimura, T; Nomura, T; Peltz, G; Pham, E; Stedman, C; Suemizu, H; Wu, M; Yurdaydin, C; Zheng, M, 2013
)
0.39
"We evaluated the safety and efficacy of the coformulation of ABC/3TC administered with DRV/r in treatment-naïve and treatment-experienced patients."( Abacavir/lamivudine fixed-dose combination with ritonavir-boosted darunavir: a safe and efficacious regimen for HIV therapy.
Boissonnault, M; Dion, H; Gallant, S; Lavoie, S; Legault, D; Longpré, D; Machouf, N; Nguyen, VK; Thomas, R; Trottier, B; Vézina, S,
)
0.39
" The drug combination appears to be generally safe and well tolerated."( Abacavir/lamivudine fixed-dose combination with ritonavir-boosted darunavir: a safe and efficacious regimen for HIV therapy.
Boissonnault, M; Dion, H; Gallant, S; Lavoie, S; Legault, D; Longpré, D; Machouf, N; Nguyen, VK; Thomas, R; Trottier, B; Vézina, S,
)
0.39
"Patients administered with atazanavir/ritonavir (300/100 mg, once daily), darunavir/ritonavir [600/100 mg, twice daily (darunavir-600) and 800/100 mg, once daily (darunavir-800)], lopinavir/ritonavir (400/100 mg, twice daily) and tipranavir/ritonavir (500/200 mg, twice daily) were considered."( Intracellular accumulation of ritonavir combined with different protease inhibitors and correlations between concentrations in plasma and peripheral blood mononuclear cells.
Agati, S; Baietto, L; Bonora, S; Calcagno, A; Cusato, J; D'Avolio, A; Di Perri, G; Larovere, G; Sciandra, M; Siccardi, M; Simiele, M; Tettoni, M; Trentini, L, 2013
)
0.95
" Telaprevir is an NS3/4A protease inhibitor approved for the treatment of chronic HCV genotype 1 infection in adults in combination with pegylated interferon and ribavirin."( Review of drug interactions with telaprevir and antiretrovirals.
Beumont, M; Garg, V; Kauffman, RS; van Heeswijk, RP, 2013
)
0.39
"Drugs most likely to interact with combined oral contraceptives, transdermal and implant contraceptives include protease inhibitors, the NNRTIs efavirenz and nevirapine, and cobicistat-boosted elvitegravir."( Drug interactions between antiretrovirals and hormonal contraceptives.
Hills-Nieminen, C; Tseng, A, 2013
)
0.39
" As adult studies cannot reliably predict their magnitude in children, drug-drug interactions should be evaluated in paediatric patient populations."( Model-based evaluation of the pharmacokinetic differences between adults and children for lopinavir and ritonavir in combination with rifampicin.
Decloedt, EH; Denti, P; Karlsson, MO; McIlleron, H; Ren, Y; Zhang, C, 2013
)
0.6
" Although antidepressants are prescribed to a significant proportion of patients treated with antiretrovirals, there are limited clinical data on drug-drug interactions."( Prediction of drug-drug interactions between various antidepressants and efavirenz or boosted protease inhibitors using a physiologically based pharmacokinetic modelling approach.
Almond, L; Back, D; Khoo, S; Kirov, A; Marzolini, C; Owen, A; Seden, K; Siccardi, M, 2013
)
0.39
" Pharmacokinetics and drug-drug interaction were simulated using the full physiologically based pharmacokinetic model implemented in the Simcyp™ ADME simulator."( Prediction of drug-drug interactions between various antidepressants and efavirenz or boosted protease inhibitors using a physiologically based pharmacokinetic modelling approach.
Almond, L; Back, D; Khoo, S; Kirov, A; Marzolini, C; Owen, A; Seden, K; Siccardi, M, 2013
)
0.39
"Simulated pharmacokinetics and drug-drug interactions were in concordance with available clinical data."( Prediction of drug-drug interactions between various antidepressants and efavirenz or boosted protease inhibitors using a physiologically based pharmacokinetic modelling approach.
Almond, L; Back, D; Khoo, S; Kirov, A; Marzolini, C; Owen, A; Seden, K; Siccardi, M, 2013
)
0.39
" These findings indicate that IVIVE is a useful tool for predicting drug-drug interactions and designing prospective clinical trials, giving insight into the variability of exposure, sample size and time-dependent induction or inhibition."( Prediction of drug-drug interactions between various antidepressants and efavirenz or boosted protease inhibitors using a physiologically based pharmacokinetic modelling approach.
Almond, L; Back, D; Khoo, S; Kirov, A; Marzolini, C; Owen, A; Seden, K; Siccardi, M, 2013
)
0.39
"Danoprevir (RG7227) is a potent macrocyclic inhibitor of the hepatitis C virus NS3/4A protease, which is currently in development in combination with low-dose ritonavir for the treatment of chronic hepatitis C infection."( Pharmacokinetics of a three-way drug interaction between danoprevir, ritonavir and the organic anion transporting polypeptide (OATP) inhibitor ciclosporin.
Asthappan, J; Brennan, BJ; Goelzer, P; Morcos, PN; Moreira, SA; Navarro, MT; Smith, PF; Weigl, P, 2013
)
0.82
" In the first period, subjects were randomized to receive either a single oral dose of danoprevir 100 mg in combination with ritonavir 100 mg or a single oral dose of ciclosporin 100 mg."( Pharmacokinetics of a three-way drug interaction between danoprevir, ritonavir and the organic anion transporting polypeptide (OATP) inhibitor ciclosporin.
Asthappan, J; Brennan, BJ; Goelzer, P; Morcos, PN; Moreira, SA; Navarro, MT; Smith, PF; Weigl, P, 2013
)
0.83
"A significant drug-drug interaction was observed between ciclosporin and danoprevir/ritonavir, leading to substantial increases in exposure to danoprevir and a lesser impact on exposure to ritonavir."( Pharmacokinetics of a three-way drug interaction between danoprevir, ritonavir and the organic anion transporting polypeptide (OATP) inhibitor ciclosporin.
Asthappan, J; Brennan, BJ; Goelzer, P; Morcos, PN; Moreira, SA; Navarro, MT; Smith, PF; Weigl, P, 2013
)
0.85
"The present study aims to determine if an in vivo rat model of drug-drug interaction (DDI) could be useful to discriminate a sensitive (buspirone) from a 'non-sensitive' (verapamil) CYP3A substrate, using ketoconazole and ritonavir as perpetrator drugs."( Assessment of CYP3A-mediated drug-drug interaction potential for victim drugs using an in vivo rat model.
Bellavance, E; Bourg, S; Duan, J; Garneau, M; Ribadeneira, MD; Rioux, N, 2013
)
0.57
" TMC310911 was generally safe and tolerable when administered with or without ritonavir."( Safety and pharmacokinetics of the HIV-1 protease inhibitor TMC310911 coadministered with ritonavir in healthy participants: results from 2 phase 1 studies.
Dierynck, I; Hoetelmans, RM; Jacquemyn, B; Marien, K; Meyvisch, P; Simmen, K; Smyej, I; Verloes, R, 2014
)
0.85
" As afatinib is not a relevant modulator or substrate of cytochrome P450 enzymes, the drug-drug interaction potential is considered to be low."( Pharmacokinetic drug interactions of afatinib with rifampicin and ritonavir.
Bertulis, J; Brand, T; Gansser, D; Giessmann, T; Hocke, J; Jungnik, A; Marzin, K; Stopfer, P; Wind, S, 2014
)
0.64
" Finally, observed/predicted drug-drug interactions between antiretrovirals and antifungals are summarized along with clinical recommendations."( Clinically relevant drug-drug interactions between antiretrovirals and antifungals.
Mitra, AK; Pal, D; Patel, M; Paturi, DK; Vadlapatla, RK, 2014
)
0.4
" Posaconazole is contraindicated in combination with either efavirenz or fosamprenavir."( Clinically relevant drug-drug interactions between antiretrovirals and antifungals.
Mitra, AK; Pal, D; Patel, M; Paturi, DK; Vadlapatla, RK, 2014
)
0.4
" The objective of this study was to compare changes in body composition between ritonavir-boosted atazanavir (ATV/r) and ritonavir-boosted lopinavir (LPV/r) over 96 weeks using data from a substudy of CASTLE, which compared once-daily ATV/r with twice-daily LPV/r, both in combination with tenofovir disoproxil fumarate/emtricitabine in treatment-naïve patients with HIV-1 infection."( Comparison of body composition changes between atazanavir/ritonavir and lopinavir/ritonavir each in combination with tenofovir/emtricitabine in antiretroviral-naïve patients with HIV-1 infection.
DeGrosky, M; Farajallah, A; Hardy, H; McGrath, D; Moyle, GJ, 2014
)
0.87
"We report an unusual case of pulmonary aspergillosis in a patient with AIDS and we demonstrated the drug-drug interactions between voriconazole, darunavir/ritonavir and tenofovir/emtricitabine."( Drug interactions between voriconazole, darunavir/ritonavir and tenofovir/emtricitabine in an HIV-infected patient treated for Aspergillus candidus lung abscess.
Becker, A; Desprez, S; Froidure, M; Gagneux, M; Huguet, D; Leduc, D; Legout, L; Sifaoui, F; Vignoli, P, 2015
)
0.87
"We conducted an open-label, steady-state pharmacokinetic (PK) study of drug-drug interactions between depot medroxyprogesterone acetate (DMPA) and twice-daily lopinavir (LPV) plus low-dose ritonavir (RTV) (LPV/r) among 24 HIV-infected women and compared the results to those for HIV-infected women receiving DMPA while on no antiretroviral therapy or on nucleosides only (n = 14 subjects from the control arm of AIDS Clinical Trials Group [ACTG] study 5093)."( Depot medroxyprogesterone acetate in combination with a twice-daily lopinavir-ritonavir-based regimen in HIV-infected women showed effective contraception and a lack of clinically significant interactions, with good safety and tolerability: results of the
Aweeka, F; Cohn, SE; Cramer, Y; Klingman, KL; Livingston, E; Luque, AE; Park, JG; Watts, DH; Weinberg, A, 2015
)
0.84
" Thirteen studies were conducted to characterize drug-drug interactions for the 3D regimen of OBV, PTV/r, and DSV and various medications in healthy volunteers to inform dosing recommendations in HCV-infected patients."( Drug-drug interaction profile of the all-oral anti-hepatitis C virus regimen of paritaprevir/ritonavir, ombitasvir, and dasabuvir.
Awni, WM; Badri, PS; Coakley, EP; Dutta, S; Hu, B; Khatri, A; Menon, RM; Podsadecki, TJ; Polepally, AR; Wang, H; Wang, T; Zha, J, 2015
)
0.64
"Mechanism-based drug-drug interactions were evaluated for gemfibrozil, ketoconazole, carbamazepine, warfarin, omeprazole, digoxin, pravastatin, and rosuvastatin."( Drug-drug interaction profile of the all-oral anti-hepatitis C virus regimen of paritaprevir/ritonavir, ombitasvir, and dasabuvir.
Awni, WM; Badri, PS; Coakley, EP; Dutta, S; Hu, B; Khatri, A; Menon, RM; Podsadecki, TJ; Polepally, AR; Wang, H; Wang, T; Zha, J, 2015
)
0.64
"The regulatory prohibition of ketoconazole as a CYP3A index inhibitor in drug-drug interaction (DDI) studies has compelled consideration of alternative inhibitors."( Ritonavir is the best alternative to ketoconazole as an index inhibitor of cytochrome P450-3A in drug-drug interaction studies.
Greenblatt, DJ; Harmatz, JS, 2015
)
1.86
" These predictions were in good agreement with clinical single dose drug-drug interaction studies, but not with reports of imatinib interactions at steady-state."( Human hepatocyte assessment of imatinib drug-drug interactions - complexities in clinical translation.
Beumer, JH; Christner, SM; Kiesel, BF; Parise, RA; Pillai, VC; Rudek, MA; Venkataramanan, R, 2015
)
0.42
"International, randomized double-blind active-controlled trial to evaluate the efficacy and safety of COBI vs ritonavir (RTV) as a pharmacoenhancer of atazanavir in combination with emtricitabine/tenofovir disoproxil fumarate in HIV treatment-naive patients followed through week 144."( Brief Report: Cobicistat Compared With Ritonavir as a Pharmacoenhancer for Atazanavir in Combination With Emtricitabine/Tenofovir Disoproxil Fumarate: Week 144 Results.
Abram, ME; Andrade-Villanueva, JF; Antunes, F; Arastéh, K; Cao, H; Chetchotisakd, P; DeJesus, E; Fehr, J; Gallant, JE; Koenig, E; Liu, HC; Rizzardini, G; Szwarcberg, J, 2015
)
0.9
" Drug-drug interactions were evaluated in healthy volunteers to develop dosing recommendations for HCV-infected subjects."( Drug Interactions with the Direct-Acting Antiviral Combination of Ombitasvir and Paritaprevir-Ritonavir.
Awni, WM; Badri, PS; Chiu, YL; Dutta, S; Khatri, A; Menon, RM; Podsadecki, TJ; Polepally, AR; Wang, H; Zha, J, 2016
)
0.65
" A drug-drug interaction (DDI) study was conducted to guide dosing recommendations for UDCA and GCR when coadministered with the 2D regimen."( Drug Interactions Between Hepatoprotective Agents Ursodeoxycholic Acid or Glycyrrhizin and Ombitasvir/Paritaprevir/Ritonavir in Healthy Japanese Subjects.
Alves, K; Badri, PS; Ding, B; Dutta, S; Menon, RM; Redman, R; Rodrigues, L; Uchiyama, N; Zha, J, 2015
)
0.63
" This phase 1, drug-drug interaction, open-label, multiple-dose study enrolled 32 healthy subjects to receive the 3D or 2D regimen in combination with sofosbuvir."( Drug-Drug Interactions between Sofosbuvir and Ombitasvir-Paritaprevir-Ritonavir with or without Dasabuvir.
Awni, WM; Cohen, D; Ding, B; Dutta, S; King, JR; Menon, RM; Podsadecki, TJ, 2016
)
0.67
" However, evening atazanavir plus ritonavir and lopinavir/ritonavir regimens are not recommended in combination with the 3D regimen."( Evaluation of Drug-Drug Interactions Between Hepatitis C Antiviral Agents Ombitasvir, Paritaprevir/Ritonavir, and Dasabuvir and HIV-1 Protease Inhibitors.
Awni, W; Dutta, S; Khatri, A; Menon, R; Podsadecki, T; Trinh, R; Wang, H, 2016
)
0.93
" Single-dose drug-drug interaction studies found no significant interactions with nevirapine or lopinavir/ritonavir, but these findings could be misleading, especially because of bedaquiline's long terminal t1/2."( Drug-drug interactions between bedaquiline and the antiretrovirals lopinavir/ritonavir and nevirapine in HIV-infected patients with drug-resistant TB.
Conradie, F; Hughes, J; Maartens, G; McIlleron, H; Pandie, M; Siwendu, S; Variava, E; Wiesner, L, 2016
)
0.88
" Drug-drug interaction (DDI) studies of the 3D regimen and commonly used medications were conducted in healthy volunteers to provide information on coadministering these medications with or without dose adjustments."( Drug-Drug Interactions Between the Anti-Hepatitis C Virus 3D Regimen of Ombitasvir, Paritaprevir/Ritonavir, and Dasabuvir and Eight Commonly Used Medications in Healthy Volunteers.
Chiu, YL; Ding, B; Dumas, EO; Khatri, A; King, JR; Menon, RM; Podsadecki, TJ; Polepally, AR; Shuster, DL, 2016
)
0.65
" While studies have demonstrated that boosting with either cobicistat or ritonavir results in comparable plasma exposure of the target antiretroviral agent, a better understanding of drug-drug interactions between cobicistat- and ritonavir-boosted antiretrovirals and other medications will inform treatment decisions in HIV-infected patients."( Drug Interactions with Cobicistat- or Ritonavir-Boosted Elvitegravir.
Custodio, JM; McNicholl, I; Nguyen, T; Piontkowsky, D; Szwarcberg, J,
)
0.63
" Drug-drug interactions between 2D (ombitasvir/paritaprevir/ritonavir) or 3D (ombitasvir/paritaprevir/ritonavir and dasabuvir) regimens and omeprazole, a CYP2C19 substrate and acid-reducing agent, were evaluated in 24 healthy volunteers."( Drug-Drug Interaction of Omeprazole With the HCV Direct-Acting Antiviral Agents Paritaprevir/Ritonavir and Ombitasvir With and Without Dasabuvir.
Awni, WM; Dutta, S; Hu, B; Menon, RM; Podsadecki, TJ; Polepally, AR, 2016
)
0.9
" However, digoxin is rarely used in HIV patients; hence, digoxin toxicity due to drug-drug interaction is not widely recognised."( Life-threatening digoxin toxicity due to drug-drug interactions in an HIV-positive man.
Heatley, MK; Roberts, B; Yoganathan, K, 2017
)
0.46
" These data indicate that the 3-direct-acting-antiviral regimen can be administered with dolutegravir or abacavir plus lamivudine without dose adjustment."( Drug-Drug Interaction between the Direct-Acting Antiviral Regimen of Ombitasvir-Paritaprevir-Ritonavir plus Dasabuvir and the HIV Antiretroviral Agent Dolutegravir or Abacavir plus Lamivudine.
Khatri, A; Menon, R; Podsadecki, T; Trinh, R; Zhao, W, 2016
)
0.65
"Ritonavir and cobicistat, used as pharmacokinetic enhancers in combination with some antiretrovirals (ARVs) for the treatment of HIV, are potent inhibitors of the CYP3A4 isoenzyme."( Iatrogenic Cushing's syndrome due to drug interaction between glucocorticoids and the ritonavir or cobicistat containing HIV therapies.
Baldeweg, SE; Boffito, M; Elliot, ER; Jain, LR; Marshall, NJ; Theodoraki, A; Waters, LJ, 2016
)
2.1
" Safe and efficacious antiviral regimens resulting in minimal to no drug-drug interactions (DDIs) with antiretrovirals are needed to ensure that patients coinfected with HCV and the human immunodeficiency virus (HIV) achieve 12-week sustained virologic response rates similar to HCV-monoinfected patients."( Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir: Drug Interactions With Antiretroviral Agents and Drugs forSubstance Abuse.
King, JR; Menon, RM, 2017
)
0.76
" To the best of our knowledge, this is the first report of severe hypokalemia and proximal tubular renal dysfunction as a result of a possible drug-drug interaction between vinblastine, tenofovir and ritonavir-boosted atazanavir."( Severe hypokalemia due to a possible drug-drug interaction between vinblastine and antiretrovirals in a HIV-infected patient with Hodgkin's lymphoma.
Arcondo, F; Cordova, E; Morganti, L; Odzak, A; Rodriguez, C; Silva, M; Zylberman, M, 2017
)
0.64
" This commentary outlines the US Food and Drug Administration (FDA) interdisciplinary review team's scientific perspective to address the potential for a significant drug-drug interaction (DDI) between clopidogrel and VIEKIRA PAK."( Utilizing PBPK Modeling to Evaluate the Potential of a Significant Drug-Drug Interaction Between Clopidogrel and Dasabuvir: A Scientific Perspective.
Arya, V; Mishra, P; Reynolds, KS; Younis, IR; Zhao, P, 2017
)
0.46
"To assess drug-drug interaction (DDI) potential for the three direct-acting antiviral (3D) regimen of ombitasvir, dasabuvir, and paritaprevir, in vitro studies profiled drug-metabolizing enzyme and transporter interactions."( Mechanisms and Predictions of Drug-Drug Interactions of the Hepatitis C Virus Three Direct-Acting Antiviral Regimen: Paritaprevir/Ritonavir, Ombitasvir, and Dasabuvir.
Bow, DAJ; de Morais, SM; Fischer, V; Kavetskaia, O; Liu, J; Nijsen, MJMA; Shebley, M; Sydor, J, 2017
)
0.66
"No drug-drug interaction study has been conducted to date for the combination of ombitasvir, paritaprevir/ritonavir, dasabuvir (3D), and mycophenolic acid (MPA)."( Managing Drug-Drug Interaction Between Ombitasvir, Paritaprevir/Ritonavir, Dasabuvir, and Mycophenolate Mofetil.
Bellissant, E; Ben Ali, Z; Boglione-Kerrien, C; Guyader, D; Jezequel, C; Lemaitre, F; Tron, C; Verdier, MC, 2017
)
0.91
" We describe the pharmacokinetics of and potential drug-drug interactions between lopinavir-ritonavir and drugs routinely used for MDR-TB treatment in HIV-infected children."( Pharmacokinetics and Drug-Drug Interactions of Lopinavir-Ritonavir Administered with First- and Second-Line Antituberculosis Drugs in HIV-Infected Children Treated for Multidrug-Resistant Tuberculosis.
de Kock, M; Denti, P; Garcia-Prats, AJ; Hesseling, AC; McIlleron, H; Norman, J; Schaaf, HS; Tikiso, T; van der Laan, LE; Wiesner, L; Winckler, J, 2018
)
0.95
" DTG in combination with one to two NRTIs was as efficient as PI/r in individuals with pre-existing NRTI mutations in this setting."( Effect of dolutegravir in combination with Nucleoside Reverse Transcriptase Inhibitors (NRTIs) on people living with HIV who have pre-existing NRTI mutations.
Carlander, C; Flamholc, L; Gisslén, M; Hejdeman, B; Sönnerborg, A; Sörstedt, E; Svedhem, V; Yilmaz, A, 2018
)
0.48
" A single subcutaneous (SC) injection of this first-generation formulation of drug combination nanoparticles (DcNPs), named TLC-ART101, provided persistent ARV levels in macaque lymph node mononuclear cells (LNMCs) for at least 1 week, and in peripheral blood mononuclear cells (PBMCs) and plasma for at least 2 weeks, demonstrating long-acting pharmacokinetics for all three drugs."( Mechanism-based pharmacokinetic (MBPK) models describe the complex plasma kinetics of three antiretrovirals delivered by a long-acting anti-HIV drug combination nanoparticle formulation.
Collier, AC; Collins, C; Ho, RJY; Kinman, L; Koehn, J; Kraft, JC; McConnachie, LA; Shen, DD; Sun, J, 2018
)
0.48
"Ritonavir is one of several ketoconazole alternatives used to evaluate strong CYP3A4 inhibition potential in clinical drug-drug interaction (DDI) studies."( Verification of a physiologically based pharmacokinetic model of ritonavir to estimate drug-drug interaction potential of CYP3A4 substrates.
He, H; Heimbach, T; Huth, F; Umehara, KI; Won, CS, 2018
)
2.16
" Hepatic clearance and drug-drug interactions (DDI) were estimated by in vitro in vivo extrapolation (IVIVE) based on parameters obtained from in vitro experiments."( Application of physiologically based pharmacokinetic modeling to the prediction of drug-drug and drug-disease interactions for rivaroxaban.
Ge, W; Jiang, Q; Xu, R, 2018
)
0.48
" When a combination therapy with interferon-free, direct-acting antivirals is used in patients post-transplantation, consideration of drug-drug interactions with and monitoring CyA are of vital importance."( Ombitasvir-Paritaprevir-Ritonavir Therapy in a Kidney Transplant Recipient With Chronic Hepatitis C Virus Genotype 1 Infection: A Case Report on the Importance of Considering Drug-Drug Interactions and Monitoring Cyclosporine Levels.
Hashimoto, S; Hayashi, K; Nakagawa, Y; Saito, K; Takahashi, K; Takamura, M; Takeuchi, S; Tanabe, Y; Tasaki, M; Terai, S; Tomita, Y; Yamagiwa, S; Yoshida, T, 2018
)
0.79
"We present the case of a 58-year-old HIV-infected patient with adrenal insufficiency after local injection of triamcinolone, most likely due to drug-drug interaction with his ritonavir-boosted antiretroviral therapy (ART)."( Adrenal insufficiency due to ritonavir-triamcinolone drug-drug interaction without preceding Cushing's syndrome.
Heldwein, S; Jaeger, H; Noe, S, 2018
)
0.97
" Treatment in the post-transplantation setting may be complicated by significant drug-drug interactions between antiviral agents and standard immune suppressive treatment regimens."( Ombitasvir/paritaprevir/ritonavir plus dasabuvir regimen may be used safely in combination with sirolimus for the treatment of chronic hepatitis C.
Dolman, GE; Gelson, WT; Selby, P, 2018
)
0.79
" This case presents the combination of tenofovir disoproxil in combination with a protease inhibitor as a new potential dual treatment regimen."( Dual antiretroviral therapy with tenofovir (TDF) and darunavir/ritonavir (DRV/RTV) in an HIV-1 positive patient: a case report, review, and meta-analysis of the literature on dual treatment strategies using protease inhibitors in combination with an NRTI.
Höring, S; Löffler, B; Pletz, MW; Rößler, S; Schleenvoigt, BT; Weis, S, 2018
)
0.72
"The coformulated lopinavir/ritonavir significantly reduces quinine concentration in healthy volunteers due to potential drug-drug interactions (DDIs)."( Physiologically-Based Pharmacokinetic Modeling for Optimal Dosage Prediction of Quinine Coadministered With Ritonavir-Boosted Lopinavir.
Karbwang, J; Na-Bangchang, K; Rajoli, RKR; Saeheng, T; Siccardi, M, 2020
)
1.07
"TLC-ART101 is a long-acting triple-HIV drug combination of lopinavir-ritonavir-tenofovir in one nanosuspension intended for subcutaneous injection."( Integration of Computational and Experimental Approaches to Elucidate Mechanisms of First-Pass Lymphatic Drug Sequestration and Long-Acting Pharmacokinetics of the Injectable Triple-HIV Drug Combination TLC-ART 101.
Collier, AC; Ho, RJY; Kinman, L; Koehn, J; Lane, S; Lee, W; McConnachie, LA; Perazzolo, S; Shen, DD; Shireman, LM, 2020
)
0.79
"Treating malaria in HIV-coinfected individuals should consider potential drug-drug interactions."( An Individual Participant Data Population Pharmacokinetic Meta-analysis of Drug-Drug Interactions between Lumefantrine and Commonly Used Antiretroviral Treatment.
Aweeka, FT; Barnes, KI; Byakika-Kibwika, P; Bygbjerg, IC; Chijioke-Nwauche, I; Denti, P; Francis, J; Hoglund, RM; Khoo, SH; Kredo, T; Lamorde, M; Lemnge, MM; Merry, C; Nyagonde, N; Parikh, S; Scarsi, KK; Sutherland, CJ; Tarning, J; Vestergaard, LS; Walimbwa, SI; Workman, L, 2020
)
0.56
"Ribociclib is approved in combination with endocrine therapy as initial endocrine-based therapy for HR-positive and HER2-negative advanced breast cancer."( Ribociclib Drug-Drug Interactions: Clinical Evaluations and Physiologically-Based Pharmacokinetic Modeling to Guide Drug Labeling.
Chakraborty, A; Dhuria, SV; Elmeliegy, M; He, H; Heimbach, T; Huth, F; Ji, Y; Miller, M; Samant, TS; Schiller, H; Umehara, K, 2020
)
0.56
"D), both in combination with lopinavir/ritonavir (LPV/r), in adult patients with human immunodeficiency virus (HIV) and tuberculosis (TB) co-infection."( Pharmacokinetic study of two different rifabutin doses co-administered with lopinavir/ritonavir in African HIV and tuberculosis co-infected adult patients.
Cisse, K; Compaoré, TR; Diagbouga, S; Kouanda, S; Matteelli, A; Ouedraogo, HG; Regazzi, M; Roggi, A; Sangare, L; Simporé, J; Sulis, G; Tarnagda, G; Villani, P, 2020
)
1.05
"This study confirmed that the 150 mg dose of rifabutin ingested EOD in combination with LPV/r is inadequate and could lead to selection of rifamycin-resistant mycobacteria."( Pharmacokinetic study of two different rifabutin doses co-administered with lopinavir/ritonavir in African HIV and tuberculosis co-infected adult patients.
Cisse, K; Compaoré, TR; Diagbouga, S; Kouanda, S; Matteelli, A; Ouedraogo, HG; Regazzi, M; Roggi, A; Sangare, L; Simporé, J; Sulis, G; Tarnagda, G; Villani, P, 2020
)
0.78
" We confirmed that ATV inhibits SARS-CoV-2 replication, alone or in combination with ritonavir (RTV) in Vero cells and a human pulmonary epithelial cell line."( Atazanavir, Alone or in Combination with Ritonavir, Inhibits SARS-CoV-2 Replication and Proinflammatory Cytokine Production.
Alves, CR; Bozza, FA; Bozza, PT; Cardoso Soares, V; Carels, N; da Silva Gomes Dias, S; de Freitas, CS; Ferreira, AC; Fintelman-Rodrigues, N; Matos, AR; Mattos, M; Miranda, MD; Ribeiro Lima, C; Sacramento, CQ; Siqueira, MM; Souza da Silva, F; Souza, TML; Temerozo, JR, 2020
)
1.05
" This potent, time-dependent interference of major hepatic drug-metabolizing enzymes by ritonavir leads to several clinically important drug-drug interactions."( Lopinavir-Ritonavir in SARS-CoV-2 Infection and Drug-Drug Interactions with Cardioactive Medications.
Agarwal, S; Agarwal, SK, 2021
)
1.25
" In this study, 37 patients were treated with up to twice-daily 30-mg paclitaxel combined with twice-daily 100-mg ritonavir (MP5/r 30-30/100-100) in 9 dose levels."( A Phase 1 Dose-Escalation Study of Low-Dose Metronomic Treatment With Novel Oral Paclitaxel Formulations in Combination With Ritonavir in Patients With Advanced Solid Tumors.
Beijnen, JH; Burylo, AM; Damoiseaux, D; de Weger, VA; Hendrikx, JJMA; Huitema, ADR; Marchetti, S; Mergui-Roelvink, M; Moes, JJ; Nuijen, B; Rosing, H; Sawicki, E; Stuurman, FE; Vermunt, MAC, 2021
)
1.04
" Here, a scalable, gravity-driven microfluidic system featuring 3D microtissues (MTs) that represent different organs for the prediction of drug-drug interactions is used."( Predicting Metabolism-Related Drug-Drug Interactions Using a Microphysiological Multitissue System.
Bonanini, F; Frey, O; Hierlemann, A; Hoelting, L; Lohasz, C; Renggli, K, 2020
)
0.56
"Patients hospitalised with severe acute respiratory syndrome coronavirus 2 [SARS-CoV-2; coronavirus 2019 disease (COVID-19)] infection are frequently older with co-morbidities and receiving polypharmacy, all of which are known risk factors for drug-drug interactions (DDIs)."( Drug-Drug Interactions and Prescription Appropriateness in Patients with COVID-19: A Retrospective Analysis from a Reference Hospital in Northern Italy.
Antinori, S; Bradanini, L; Cattaneo, D; Covizzi, A; Gervasoni, C; Giacomelli, A; Maggioni, AP; Oreni, L; Pasina, L; Ridolfo, A; Schiuma, M, 2020
)
0.56
"The impact of drug-drug interactions (DDI) between ritonavir-boosted lopinavir (LPV-r) to treat patients with coronavirus disease 2019 (COVID-19) and commonly used drugs in clinical practice is not well-known."( High rate of major drug-drug interactions of lopinavir-ritonavir for COVID-19 treatment.
Contreras-Macias, E; Corma, A; Fernández-Fuertes, M; González-Serna, A; Gutierrez-Pizarraya, A; Lao-Dominguez, FA; Macías, J; Morillo-Verdugo, R; Pineda, JA; Pinilla, A; Real, LM; Trigo, M, 2020
)
1.06
" The objective of this study was to analyse the potential drug-drug interactions (pDDIs) derived from the medication used in COVID-19 patients in the first pandemic wave and to evaluate the real consequences of such interactions in clinical practice."( Real-world prevalence and consequences of potential drug-drug interactions in the first-wave COVID-19 treatments.
Álvarez-Payero, M; Casanova-Martínez, C; Castro-Núñez, I; García-Beloso, N; González-Costas, S; Inaraja-Bobo, MT; Leboreiro-Enríquez, B; López-López, A; Lorenzo-Lorenzo, K; Martínez-López-de-Castro, N; Martínez-Reglero, C; Otero-Millán, L; Paradela-Carreiro, A; Pérez-Landeiro, A; Piñeiro-Corrales, G; Regueira-Arcay, AM; Robles-Torres, D; Samartín-Ucha, M; Vázquez-López, C; Yaiza Romero-Ventosa, E, 2021
)
0.62
"This study aimed to evaluate the antiviral efficacy of lopinavir-ritonavir alone or combined with arbidol in the treatment of hospitalized patients with common coronavirus disease-19 (COVID-19)."( Lopinavir-ritonavir alone or combined with arbidol in the treatment of 73 hospitalized patients with COVID-19: A pilot retrospective study.
Lan, X; Shao, C; Wu, Z; Xu, Y; Zeng, X, 2021
)
1.26
"No benefit was observed in the antiviral effect of lopinavir-ritonavir combined with arbidol compared with lopinavir-ritonavir alone in the hospitalized patients with COVID-19."( Lopinavir-ritonavir alone or combined with arbidol in the treatment of 73 hospitalized patients with COVID-19: A pilot retrospective study.
Lan, X; Shao, C; Wu, Z; Xu, Y; Zeng, X, 2021
)
1.27
" Drug-drug interaction (DDI) studies were only conducted for strong inhibitors and inducers, leading to some uncertainty whether moderate perpetrators or multiple drug associations can be safely coadministered with bictegravir."( Physiologically-Based Pharmacokinetic Modeling to Support the Clinical Management of Drug-Drug Interactions With Bictegravir.
Battegay, M; Marzolini, C; Stader, F, 2021
)
0.62
" Due to suspicion of a potential drug-drug interaction, both eplerenone and ARVs were interrupted."( Severe hyperkalaemia due to a potential drug-drug interaction between eplerenone and antiretrovirals in a HIV-positive patient after a myocardial infarction.
Bono, L; Cordova, E; Garibaldi, F; Rodriguez, C, 2021
)
0.62
" The aim of this study was to establish an insect model for pharmacokinetic and drug-drug interaction studies to develop sustainable endectocides for vector control."( The pharmacokinetics and drug-drug interactions of ivermectin in Aedes aegypti mosquitoes.
Chaccour, C; Duthaler, U; Hammann, F; Hofer, L; Krähenbühl, S; Maia, M; Müller, P; Weber, M, 2021
)
0.62
" However, management of acute seizures in patients with COVID-19 as well as management of PWE and COVID-19 needs to consider potential drug-drug interactions between antiseizure drugs and candidate drugs currently assessed as therapeutic options for COVID-19."( Management of COVID-19 in patients with seizures: Mechanisms of action of potential COVID-19 drug treatments and consideration for potential drug-drug interactions with anti-seizure medications.
Chandra, PP; Jain, S; Potschka, H; Tripathi, M; Vohora, D, 2021
)
0.62
"Primary aim was to assess prevalence and severity of potential and real drug-drug interactions (DDIs) among therapies for COVID-19 and concomitant medications in hospitalized patients with confirmed SARS-CoV-2 infection."( Drug-drug interactions between treatment specific pharmacotherapy and concomitant medication in patients with COVID-19 in the first wave in Spain.
Cantudo-Cuenca, MD; Contreras-Macías, E; Fernández-Fuertes, M; Gutiérrez-Pizarraya, A; Lao-Domínguez, FA; Macías, J; Morillo-Verdugo, R; Pineda, JA; Pinilla-Fernández, A; Rincón, P, 2021
)
0.62
" As medroxyprogesterone acetate is a cytochrome P450 (CYP3A4) substrate, drug-drug interactions (DDIs) with antiretroviral or antituberculosis treatment may lead to subtherapeutic medroxyprogesterone acetate concentrations (< 0."( A Semimechanistic Pharmacokinetic Model for Depot Medroxyprogesterone Acetate and Drug-Drug Interactions With Antiretroviral and Antituberculosis Treatment.
Cohn, SE; Denti, P; Dooley, KE; Firnhaber, C; Francis, J; Godfrey, C; Kendall, MA; McIlleron, H; Mngqibisa, R; Wu, X, 2021
)
0.62
"The aim of this study was to simulate the drug-drug interaction (DDI) between ritonavir-boosted atazanavir (ATV/r) and rifampicin (RIF) using physiologically based pharmacokinetic (PBPK) modelling, and to predict suitable dose adjustments for ATV/r for the treatment of people living with HIV (PLWH) co-infected with tuberculosis."( Predicting Drug-Drug Interactions between Rifampicin and Ritonavir-Boosted Atazanavir Using PBPK Modelling.
Bunglawala, F; Cottura, N; Denti, P; Fabrega, F; Howarth, A; Kinvig, H; Lloyd, A; Montanha, MC; Siccardi, M; Waitt, C, 2022
)
1.19
"The aim of the study is to provide an overview of Drug-drug Interactions (DDIs) and adverse effects caused by drugs used in SARS-CoV-2 infection during the first epidemic wave."( [Treatments for SARS-CoV-2 infection: A retrospective study of drug-drug interactions and safety].
Benomar, A; Clou, E; Debrix, I; Dubois, A; Fansi Ndengoue, D; Féral, A; Michot, J; Pain, JB, 2022
)
0.72
" Potential drug-drug interactions may occur, leading to iatrogenic Cushing syndrome or adrenal insufficiency."( Antiretroviral therapies and corticosteroids: Drug-drug interactions.
Cadet, MJ, 2021
)
0.62
"Although the combination with PrOD significantly affects the pharmacokinetics of CsA, it is effective and safe with regular monitoring of the CsA blood concentrations and appropriate CsA dose adjustment."( Drug-Drug Interactions With Cyclosporine in the Anti-Hepatitis C Viral PrOD Combination Regimen of Paritaprevir/Ritonavir-Ombitasvir and Dasabuvir in Organ Transplant Recipients With Severe Hepatic Fibrosis or Cirrhosis.
Chang, YL; Chou, YC; Hsu, CC; Huang, YH; Huang, YY; Loong, CC; Wu, TH, 2022
)
0.93
" Following a 2 × 2 factorial design and stratified by site and screening HIV-1 RNA concentration, participants were randomly assigned (1:1:1:1) to receive a 96-week regimen containing either dolutegravir (50 mg once daily) or ritonavir-boosted darunavir (800 mg of darunavir plus 100 mg of ritonavir once daily) in combination with either tenofovir (300 mg once daily) plus lamivudine (300 mg once daily) or zidovudine (300 mg twice daily) plus lamivudine (150 mg twice daily)."( Efficacy and safety of dolutegravir or darunavir in combination with lamivudine plus either zidovudine or tenofovir for second-line treatment of HIV infection (NADIA): week 96 results from a prospective, multicentre, open-label, factorial, randomised, non
Asienzo, J; Ategeka, G; Balyegisawa, A; Borok, M; Castelnuovo, B; Hoppe, A; Kaimal, A; Kambugu, A; Kiragga, A; Kityo, C; Lugemwa, A; Mirembe, G; Mugerwa, H; Musaazi, J; Odongpiny, ELA; Paton, NI; Siika, A; Walimbwa, S, 2022
)
0.91
"Although rifampin drug-drug interaction (DDI) studies are routinely conducted, there have been instances of liver function test (LFT) elevations, warranting further evaluation."( A literature review of liver function test elevations in rifampin drug-drug interaction studies.
Chen, J; Ibrahim, SM; Pithavala, YK; Vourvahis, M, 2022
)
0.72
"Due to health authority warnings and the recommended limited use of ketoconazole as a model inhibitor of cytochrome P450 (CYP) 3A4 in clinical drug-drug interaction (DDI) studies, there is a need to search for alternatives."( A Mechanistic Absorption and Disposition Model of Ritonavir to Predict Exposure and Drug-Drug Interaction Potential of CYP3A4/5 and CYP2D6 Substrates.
Arora, S; Gardner, I; Jamei, M; Kilford, PJ; Pansari, A; Turner, DB, 2022
)
0.97
"The current model, which incorporates formulation characteristics and mechanistic disposition parameters, can be used to assess the DDI potential of CYP3A4/5 and CYP2D6 substrates administered with a twice-daily dose of 100 mg of ritonavir for 14 days."( A Mechanistic Absorption and Disposition Model of Ritonavir to Predict Exposure and Drug-Drug Interaction Potential of CYP3A4/5 and CYP2D6 Substrates.
Arora, S; Gardner, I; Jamei, M; Kilford, PJ; Pansari, A; Turner, DB, 2022
)
1.16
" Thus nirmatrelvir/ritonavir, even given as a short treatment course, has a high potential to cause harm from drug-drug interactions (DDIs) with other drugs metabolized through this pathway."( Recommendations for the Management of Drug-Drug Interactions Between the COVID-19 Antiviral Nirmatrelvir/Ritonavir (Paxlovid) and Comedications.
Back, DJ; Boffito, M; Boyle, A; Burger, D; Flexner, C; Gibbons, S; Khoo, S; Kuritzkes, DR; Marra, F; Marzolini, C; Pozniak, A; Waters, L, 2022
)
1.26
" New antiviral medications against this disease have not been properly tested yet, and their efficiency, side effects, and drug-drug interactions are not entirely known."( Paxlovid (Nirmatelvir/Ritonavir) and Tacrolimus Drug-Drug Interaction in a Kidney Transplant Patient with SARS-2-CoV infection: A Case Report.
Cameron, A; Prikis, M,
)
0.45
" This combination with a well-known pharmacokinetic enhancer leads to a high risk for drug-drug interactions in a polymedicated elected population for treatment."( Management of drug-drug interactions with nirmatrelvir/ritonavir in patients treated for Covid-19: Guidelines from the French Society of Pharmacology and Therapeutics (SFPT).
Bouchet, S; Grégoire, M; Lemaitre, F; Monchaud, C; Polard, E; Saint-Salvi, B,
)
0.38
" In complex situations, clinicians are advised to contact their pharmacology department to obtain specific recommendations on the management of drug-drug interactions with nirmatrelvir/ritonavir."( Management of drug-drug interactions with nirmatrelvir/ritonavir in patients treated for Covid-19: Guidelines from the French Society of Pharmacology and Therapeutics (SFPT).
Bouchet, S; Grégoire, M; Lemaitre, F; Monchaud, C; Polard, E; Saint-Salvi, B,
)
0.57
"These recommendations intend to be a help for clinicians willing to prescribe nirmatrelvir/ritonavir and to prevent drug-drug interactions leading to adverse drug reactions or loss of efficacy."( Management of drug-drug interactions with nirmatrelvir/ritonavir in patients treated for Covid-19: Guidelines from the French Society of Pharmacology and Therapeutics (SFPT).
Bouchet, S; Grégoire, M; Lemaitre, F; Monchaud, C; Polard, E; Saint-Salvi, B,
)
0.6
" Ritonavir, contained in NMV/r, is known to have significant drug-drug interactions (DDI) with several drugs frequently used by the elderly."( Assessing the proportion of the Danish population at risk of clinically significant drug-drug interactions with new oral antivirals for early treatment of COVID-19.
Larsen, CS, 2022
)
1.63
"We estimated the size of the Danish population at risk of significant DDI with antiviral COVID-19 treatment using the number of claimed prescriptions for drugs predicted to interact with NMV/r in Denmark in 2020."( Assessing the proportion of the Danish population at risk of clinically significant drug-drug interactions with new oral antivirals for early treatment of COVID-19.
Larsen, CS, 2022
)
0.72
"Danish prescription data demonstrate the extensive use of drugs likely to interact with NMV/r."( Assessing the proportion of the Danish population at risk of clinically significant drug-drug interactions with new oral antivirals for early treatment of COVID-19.
Larsen, CS, 2022
)
0.72
" This combination of nirmatrelvir and ritonavir can mediate significant and complex drug-drug interactions (DDIs), primarily due to the ritonavir component."( Therapeutic Drug Monitoring and Dosage Adjustments of Immunosuppressive Drugs When Combined With Nirmatrelvir/Ritonavir in Patients With COVID-19.
Bergan, S; Brunet, M; Budde, K; De Winter, BCM; Elens, L; Hesselink, DA; Johnson-Davis, KL; Langman, LJ; Lawson, R; Lemaitre, F; Masuda, S; Moes, DJAR; Noceti, O; Pattanaik, S; Pawinski, T; Van Gelder, T; Venkataramanan, R, 2023
)
1.39
" FTR was administered with and without ritonavir (RTV), a protease inhibitor previously shown to boost TMR exposures."( Pharmacokinetics, metabolism and excretion of radiolabeled fostemsavir administered with or without ritonavir in healthy male subjects.
Ackerman, P; Gorycki, P; Magee, M; Miao, X; Moore, K, 2022
)
1.21
" Co-administration of NMVr with medications commonly used to manage cardiovascular conditions can potentially cause significant drug-drug interactions and may lead to severe adverse effects."( Cardiovascular Drug Interactions With Nirmatrelvir/Ritonavir in Patients With COVID-19: JACC Review Topic of the Week.
Abraham, GM; Abraham, S; Asnani, A; Dani, SS; Ganatra, S; Grossman, J; Lech, T; Martin, DT; McQuillen, DP; Neilan, TG; Nohria, A; Saji, AM; Sax, PE; Shah, J, 2022
)
0.97
"This case highlights the strong and important drug-drug interaction between tacrolimus and nirmatrelvir/ritonavir leading to toxic levels of tacrolimus."( Tacrolimus Drug-Drug Interaction with Nirmatrelvir/Ritonavir (Paxlovid™) Managed with Phenytoin.
Carroll, E; McCabe, D; Sindelar, M, 2023
)
1.38
" However, the use of NMV/r is complicated by significant drug-drug interactions (DDIs) with frequently prescribed medications."( Drug interactions between common dermatological medications and the oral anti-COVID-19 agents nirmatrelvir-ritonavir and molnupiravir.
Huang, X; Oon, HH; Quah, KSE; Renia, L, 2022
)
0.93
"To review the drug-drug interactions between tacrolimus and lopinavir/ritonavir in 23 patients who received solid organ transplant during the first wave of COVID-19 and to determine the efficacy as well as safety of prednisone monotherapy."( Drug-drug interactions of ritonavir-boosted SARS-CoV-2 protease inhibitors in solid organ transplant recipients: experience from the initial use of lopinavir-ritonavir.
Ambrosioni, J; Arranz, N; Bodro, M; Brunet, M; Castel, MÁ; Cofan, F; Crespo, G; Diekmann, F; Farrero, M; Forner, A; Gonzalez-García, R; LLigoña, A; Marcos, MÁ; Miró, JM; Moreno, A; Rodríguez-García, M; Roma, JR; Ruiz, P; Soy, D; Tuset, M, 2023
)
1.44
"This Teachable Moment discusses drug-drug interactions with nirmatrelvir and ritonavir (Paxlovid) treatment for COVID-19 and considerations when prescribing."( Safely Prescribing Nirmatrelvir and Ritonavir-Avoiding Drug-Drug Interactions.
Culas, R; Nath, S, 2023
)
1.41
"Previous use of a mechanistic static model to accurately quantify the increased rosuvastatin exposure due to drug-drug interaction (DDI) with coadministered atazanavir underpredicted the magnitude of area under the plasma concentration-time curve ratio (AUCR) based on inhibition of breast cancer resistance protein (BCRP) and organic anion transporting polypeptide (OATP) 1B1."( Mechanistic in vitro studies indicate that the clinical drug-drug interactions between protease inhibitors and rosuvastatin are driven by inhibition of intestinal BCRP and hepatic OATP1B1 with minimal contribution from OATP1B3, NTCP and OAT3.
Atkinson, H; Coghlan, H; Edgerton, J; Elsby, R; Hodgson, D; Outteridge, S, 2023
)
0.91
"The evolving epidemiology and treatment landscape of COVID-19 necessitates research into potential drug-drug interactions (pDDIs) from the use of new treatments for COVID-19, particularly those that contain ritonavir, a potent inhibitor of the cytochrome P350 3A4 (CYP3A4) metabolic pathway."( Prevalence of Potential Drug-drug Interactions With Ritonavir-containing COVID-19 Therapy in the United States: An Analysis of the National Health and Nutrition Examination Survey.
Brown, J; Brzozowski, K; Igho-Osagie, E; Jin, H; Puenpatom, A; Williams, MG, 2023
)
1.35
"Co-administration of Bruton's tyrosine kinase (BTK) inhibitors with nirmatrelvir/ritonavir is challenging because of potential drug-drug interactions (DDIs)."( Drug-drug interactions and dose management of BTK inhibitors when initiating nirmatrelvir/ritonavir (paxlovid) based on physiologically-based pharmacokinetic models.
Chen, L; Chen, W; Li, C; Li, L, 2023
)
1.36

Bioavailability

Coadministration of ritonavir significantly enhanced the apparent oral bioavailability of docetaxel. In HIV+ children on lopinavir/ritonavir, bioavailability was reduced by 32% [median (IQR) steady-state Cmax’s].

ExcerptReferenceRelevance
" These findings demonstrate that high oral bioavailability can be achieved in humans with peptidomimetic inhibitors of HIV protease."( ABT-538 is a potent inhibitor of human immunodeficiency virus protease and has high oral bioavailability in humans.
Denissen, JF; Fino, L; Flentge, CA; Green, BE; Kempf, DJ; Kong, XP; Marsh, KC; McDonald, E; Park, CH; Vasavanonda, S, 1995
)
0.29
" Ritonavir has good oral bioavailability, and may increase the bioavailability of other protease inhibitors including saquinavir, nelfinavir, indinavir and VX-478."( Ritonavir.
Faulds, D; Lea, AP, 1996
)
2.65
" Ritonavir was well absorbed at all dose levels, and plasma concentrations reached a peak 2 to 4 hours after a dose."( A phase I/II study of the protease inhibitor ritonavir in children with human immunodeficiency virus infection.
Aker, D; Balis, FM; Brouwers, P; Edgerly, M; Heath-Chiozzi, M; Higham, C; Hsu, A; Jarosinski, P; Katz, TT; Mueller, BU; Nelson, RP; Pizzo, PA; Pizzuti, D; Saulis, R; Sei, S; Serchuck, L; Sleasman, J; Steinberg, SM; Whitcup, SM; Wood, LV; Zeichner, S; Zuckerman, J, 1998
)
1.47
" Beginning with the moderately potent and orally bioavailable inhibitor A-80987, systematic investigation of peripheral (P3 and P2') heterocyclic groups designed to decrease the rate of hepatic metabolism provided analogues with improved pharmacokinetic properties after oral dosing in rats."( Discovery of ritonavir, a potent inhibitor of HIV protease with high oral bioavailability and clinical efficacy.
Betebenner, D; Fino, L; Flentge, CA; Green, BE; Kati, WM; Kempf, DJ; Marsh, KC; McDonald, E; Molla, A; Norbeck, DW; Patterson, J; Plattner, JJ; Ruiz, L; Saldivar, A; Sham, HL; Vasavanonda, S; Wideburg, NE; Zhao, C, 1998
)
0.67
"Ritonavir is a potent, orally bioavailable inhibitor of HIV-1 protease."( Randomised placebo-controlled trial of ritonavir in advanced HIV-1 disease. The Advanced HIV Disease Ritonavir Study Group.
Cameron, DW; Cohen, C; Danner, S; Heath-Chiozzi, M; Henry, D; Kravcik, S; Leonard, J; Maurath, C; Potthoff, A; Rode, R; Sun, E, 1998
)
2.01
"To investigate in vitro the mechanisms involved in the gastro-intestinal absorption of the HIV protease inhibitor, saquinavir mesylate (Invirase), whose oral bioavailability is low, variable, and significantly increased by co-administration with ritonavir, also an HIV protease inhibitor but with higher oral bioavailability."( Active apical secretory efflux of the HIV protease inhibitors saquinavir and ritonavir in Caco-2 cell monolayers.
Alex, R; Alsenz, J; Steffen, H, 1998
)
0.71
" Together with sensitivity to gutwall metabolism by cytochrome P-450 3A, this may partially account for the low and variable oral bioavailability of saquinavir in clinical studies and for its increased bioavailability after co-administration with ritonavir."( Active apical secretory efflux of the HIV protease inhibitors saquinavir and ritonavir in Caco-2 cell monolayers.
Alex, R; Alsenz, J; Steffen, H, 1998
)
0.71
" Ritonavir may have enhanced rifabutin bioavailability by reducing either intestinal of hepatic metabolism of both."( The effect of multiple doses of ritonavir on the pharmacokinetics of rifabutin.
Cato, A; Cavanaugh, J; Granneman, R; Hsu, A; Leonard, J; Shi, H, 1998
)
1.49
" Given the limited bioavailability of saquinavir given in the hard gelatin capsule formulation, this drug interaction is expected to have implications in the use of protease inhibitors in the management of human immunodeficiency virus infection."( Pharmacokinetic interactions between two human immunodeficiency virus protease inhibitors, ritonavir and saquinavir.
Baroldi, P; Brown, F; Cao, G; Carothers, L; el-Shourbagy, T; Erdman, K; Granneman, GR; Hsu, A; Leonard, JM; Sun, E, 1998
)
0.52
" Metabolic inactivation of CYP3A by ritonavir explains the improved bioavailability and pharmacokinetics of ritonavir and the sustained elevation of blood levels of other, concomitantly administered, substrates of CYP3A."( Metabolism of the human immunodeficiency virus protease inhibitors indinavir and ritonavir by human intestinal microsomes and expressed cytochrome P4503A4/3A5: mechanism-based inactivation of cytochrome P4503A by ritonavir.
Gangl, E; Gerber, N; Iatsimirskaia, E; Koudriakova, T; Marinina, J; Orza, D; Storozhuk, E; Utkin, I; Vouros, P, 1998
)
0.8
" Using this approach several very potent (IC90 11 nM) and orally bioavailable (F% 93-100%) compounds were discovered (21, 22)."( Nonsymmetric P2/P2' cyclic urea HIV protease inhibitors. Structure-activity relationship, bioavailability, and resistance profile of monoindazole-substituted P2 analogues.
Bacheler, LT; Cordova, B; De Lucca, GV; Erickson-Viitanen, S; Garber, S; Kim, UT; Klabe, RM; Ko, SS; Lam, GN; Liang, J; Logue, KA; Trainor, GL; Wright, MR, 1998
)
0.3
" Ritonavir is a potent, orally bioavailable HIV protease inhibitor with demonstrated impact on surrogate endpoints, AIDS-defining disease, and mortality."( A randomized trial of the effect of ritonavir in maintaining quality of life in advanced HIV disease. Advanced HIV Disease Ritonavir Study Group.
Cohen, C; Jiang, P; Nabulsi, A; Revicki, DA; Sarocco, PW, 1998
)
1.49
" The drug transporting protein, P-glycoprotein (P-gp), which is highly expressed in the intestinal mucosa, could be responsible for the low oral bioavailability of these and other drugs which are substrates for this transporter."( Interaction of anti-HIV protease inhibitors with the multidrug transporter P-glycoprotein (P-gp) in human cultured cells.
Blaschke, TF; Duran, GE; Man, MC; Sikic, BI; Washington, CB, 1998
)
0.3
" The ultimate success of DMP 850 and DMP 851 in clinical trials might depend on achieving or exceeding the oral bioavailability seen in dog."( Design and selection of DMP 850 and DMP 851: the next generation of cyclic urea HIV protease inhibitors.
Anderson, PS; Bacheler, LT; Chang, CH; Cordova, B; Erickson-Viitanen, S; Garber, S; Johnson, BL; Klabe, RM; Ko, SS; Lam, PY; Li, R; Reid, C; Rodgers, JD; Ru, Y; Seitz, SP; Trainor, GL; Wang, H; Wright, MR, 1998
)
0.3
" The poor bioavailability of the current saquinavir formulation also leads to risk of interactions."( The choice of HIV protease inhibitor: indinavir is currently the best option.
, 1999
)
0.3
"The low oral bioavailability of the HIV protease inhibitor (HPI) saquinavir is dramatically increased by coadministration of the HPI ritonavir."( P-glycoprotein limits oral availability, brain, and fetal penetration of saquinavir even with high doses of ritonavir.
Beijnen, JH; Hoetelmans, RM; Huisman, MT; Schinkel, AH; Smit, JW; Wiltshire, HR, 2001
)
0.73
" However, two pharmacokinetic problems have been identified: suboptimal oral bioavailability of peptidic inhibitors; and reduced cellular uptake of inhibitor that has become bound to alpha-acid glycoprotein."( Update on HIV protease inhibitors.
Vella, S, 1995
)
0.29
"A study of patients taking ritonavir and methadone indicates that ritonavir lowers bioavailability of methadone by 30 to 40 percent."( Ritonavir (Norvir) and methadone.
, 1998
)
2.04
" Toward this end, we previously identified a 4-hydroxy-5,6-dihydropyrone lead compound (CI-1029, 1) which possesses excellent activity against the protease enzyme, good antiviral efficacy in cellular assays, and promising bioavailability in several animal species."( 4-Hydroxy-5,6-dihydropyrones as inhibitors of HIV protease: the effect of heterocyclic substituents at C-6 on antiviral potency and pharmacokinetic parameters.
Brodfuehrer, J; Domagala, J; Gajda, C; Gracheck, SJ; Hagen, SE; Holler, T; Hupe, D; Lovdahl, M; Lunney, EA; Nouhan, C; Pavlovsky, A; Saunders, J; Tait, BD; Urumov, A; VanderRoest, S; Wise, E; Zeikus, E; Zeikus, G, 2001
)
0.31
" The coadministration of the HIV-1 protease inhibitor ritonavir increased the bioavailability of (R)-(-)-PPO464, having little effect on its plasma and brain elimination rates."( The stereoselective targeting of a specific enzyme-substrate complex is the molecular mechanism for the synergic inhibition of HIV-1 reverse transcriptase by (R)-(-)-PPO464: a novel generation of nonnucleoside inhibitors.
Baldanti, F; Bergamini, A; Caccia, S; Campiani, G; Fiorini, I; Galletti, B; Giorgi, G; Guiso, G; Hubscher, U; Locatelli, GA; Maga, G; Muck, S; Nacci, V; Paolucci, S; Ramunno, A; Spadari, S; Zavattoni, M, 2001
)
0.56
" The absence of marked central opioid effects has been attributed to its low bioavailability and its poor penetration of the blood-brain barrier, both of which might be altered by ritonavir, a potent P-glycoprotein and cytochrome P4503A inhibitor."( Ritonavir increases loperamide plasma concentrations without evidence for P-glycoprotein involvement.
Aderjan, R; Burhenne, J; Ding, R; Ganssmann, B; Haefeli, WE; Klingmann, A; Mikus, G; Tayrouz, Y, 2001
)
1.95
" Chronic use of Saint John's wort (SJW) has been shown to lower the bioavailability for a variety of co-administered drugs including indinavir, cyclosporin, and digoxin."( Saint John's wort: an in vitro analysis of P-glycoprotein induction due to extended exposure.
Greenblatt, DJ; Perloff, MD; Shader, RI; Störmer, E; von Moltke, LL, 2001
)
0.31
" Similar in vivo phenomena may occur during anti-HIV drug therapy, explaining potential decrements in therapeutic efficacy due to decreases in bioavailability or alterations in drug distribution."( Ritonavir induces P-glycoprotein expression, multidrug resistance-associated protein (MRP1) expression, and drug transporter-mediated activity in a human intestinal cell line.
Greenblatt, DJ; Marchand, JE; Perloff, MD; Von Moltke, LL, 2001
)
1.75
"The liver had a high intrinsic capacity for clearing VL because the absolute bioavailability (BA) of VL was 21."( Differentiation of gut and hepatic first-pass effect of drugs: 1. Studies of verapamil in ported dogs.
Kunta, JR; Lee, HS; Lee, YH; Perry, BA; Sinko, PJ; Sutyak, JP, 2001
)
0.31
" Inhibitors of CYP3A4 such as other protease inhibitors will substantially increase the bioavailability of saquinavir."( CYP3A4-mediated hepatic metabolism of the HIV-1 protease inhibitor saquinavir in vitro.
Back, DJ; Eagling, VA; Whitcombe, IW; Wiltshire, H, 2002
)
0.31
" After having recalled some thermodynamic considerations which characterize the polymorphism, we will illustrate succinctly the impact of these phenomena on some important aspects of the pharmaceutical development such as the dissolution, the bioavailability and the stability."( [Crystallization and solid state properties of molecules of pharmaceutical interest].
Bauer, M, 2002
)
0.31
"Emergence of human immunodeficiency virus type-1 (HIV-1) genotypic drug resistance is generally attributed to noncompliance, poorly absorbed drugs, or drug-to-drug interaction."( In vivo emergence of drug-resistant mutations at less than 50 HIV-1 RNA copies/mL that are maintained at viral rebound in longitudinal plasma samples from human immunodeficiency virus type-1-infected patients on highly active antiretroviral therapy.
Benetti, MS; Bortolozzi, RL; Campodonico, ME; Chernoff, D; Dileanis, J; Elbeik, T; Fay, FF; Fay, OH; Hoo, BS; Marlowe, N; Ng, VL; Petrauskene, O; Smith, L,
)
0.13
" We have now explored the mechanisms responsible for the low oral bioavailability of docetaxel, a structurally related taxane drug."( Low systemic exposure of oral docetaxel in mice resulting from extensive first-pass metabolism is boosted by ritonavir.
Bardelmeijer, HA; Beijnen, JH; Buckle, T; Huisman, MT; Ouwehand, M; Schellens, JH; van Tellingen, O, 2002
)
0.53
" R given simultaneously with S decreases S hepatic intrinsic clearance almost 50-fold relative to that predicted for S given alone and increases its gut bioavailability 90% (but decreases its rate of absorption 40%) relative to when N is given simultaneously; 2)."( Model-based analysis of the pharmacokinetic interactions between ritonavir, nelfinavir, and saquinavir after simultaneous and staggered oral administration.
Blaschke, TF; Flexner, C; Lu, JF; Rosenkranz, SL; Sheiner, LB, 2002
)
0.55
"Various drug transporters of the ATP-binding cassette (ABC) family restrict the oral bioavailability and cellular, brain, testis, cerebrospinal fluid and fetal penetration of substrate drugs."( Multidrug resistance protein 2 (MRP2) transports HIV protease inhibitors, and transport can be enhanced by other drugs.
Beijnen, JH; Crommentuyn, KM; Huisman, MT; Schinkel, AH; Smit, JW; Wiltshire, HR; Zelcer, N, 2002
)
0.31
" Although the systemic pharmacokinetics of MRK-1 in rats and monkeys were linear, the oral bioavailability increased with an increase in dose from 2 to 10 mg/kg."( Pharmacokinetics and interactions of a novel antagonist of chemokine receptor 5 (CCR5) with ritonavir in rats and monkeys: role of CYP3A and P-glycoprotein.
Baillie, TA; Chen, Q; Chiu, SH; Didolkar, V; Franklin, RB; Iliff, SA; Kumar, S; Kwei, GY; Lin, JH; Pearson, PG; Poon, GK; Wang, RW; Wang, Y; Yamazaki, M, 2003
)
0.54
" In 1998, a lower energy, more stable polymorph (form II) appeared, causing slowed dissolution of the marketed dosage form and compromising the oral bioavailability of the drug."( Elucidation of crystal form diversity of the HIV protease inhibitor ritonavir by high-throughput crystallization.
Almarsson, O; Cima, MJ; Levinson, D; Morissette, SL; Soukasene, S, 2003
)
0.55
"The absolute bioavailability of lopinavir coformulated with ritonavir in humans has not yet been established."( Lopinavir/ritonavir: a review of its use in the management of HIV infection.
Cvetkovic, RS; Goa, KL, 2003
)
0.96
" As the oral bioavailability of both didanosine and lopinavir/ritonavir is significantly affected by concurrent food ingestion, didanosine should be administered 1 hour before or 2 hours after lopinavir/ritonavir has been taken with food."( Lopinavir/ritonavir: a review of its use in the management of HIV infection.
Cvetkovic, RS; Goa, KL, 2003
)
0.96
" As a potent inhibitor of this enzyme, ritonavir can increase the bioavailability and half-life of coadministered protease inhibitors."( A review of low-dose ritonavir in protease inhibitor combination therapy.
Cameron, DW; Cooper, CL; Gallicano, K; van Heeswijk, RP, 2003
)
0.91
" The results of this study are in agreement with the clinical observation that a high-calorie protein meal significantly reduces the oral bioavailability of indinavir in man, accompanying a pH elevation in the stomach."( In-vitro crystallization of indinavir in the presence of ritonavir and as a function of pH.
Fleisher, D; Heimbach, T; Li, LY; Rodríguez-Hornedo, N, 2003
)
0.56
" This study showed that ethanol-intake decreases the bioavailability of SQV after oral administration alone or with RTV."( Pharmacokinetic characterization of a human immunodeficiency virus protease inhibitor, saquinavir, during ethanol intake in rats.
Kageyama, M; Kimura, K; Kishida, T; Kuwahara, T; Shibata, N; Shirasaka, T; Takada, K; Toh, J; Yoshikawa, Y, 2003
)
0.32
" As a result, the bioavailability of the boosted protease inhibitor is increased and improved penetration into HIV reservoirs may be achieved."( Pharmacological and therapeutic properties of ritonavir-boosted protease inhibitor therapy in HIV-infected patients.
Petruschke, RA; Zeldin, RK, 2004
)
0.58
" A crossover design was used to evaluate the oral bioavailability of amorphous dispersions relative to crystalline drug in beagle dogs."( Ritonavir-PEG 8000 amorphous solid dispersions: in vitro and in vivo evaluations.
Everitt, EA; Fort, JJ; Krill, SL; Law, D; Marsh, KC; Qiu, Y; Schmitt, EA; Wang, W, 2004
)
1.77
" This property of RTV is responsible for regulating the oral bioavailability of drugs that are mediated by CYP3A and Pgp."( Effect of chronic administration of ritonavir on function of cytochrome P450 3A and P-glycoprotein in rats.
Fukushima, H; Ito, Y; Kageyama, M; Namiki, H; Shibata, N; Takada, K; Tanaka, A; Terasaka, S; Togawa, T, 2005
)
0.6
" Clearance, volume of distribution and absorption rate constant were 10."( Development and validation of a population pharmacokinetic model for ritonavir used as a booster or as an antiviral agent in HIV-1-infected patients.
Beijnen, JH; Crommentuyn, KM; Huitema, AD; Kappelhoff, BS; Mairuhu, AT; Meenhorst, PL; Mulder, JW; van Gorp, EC, 2005
)
0.56
" The interindividual variability value of the Vm could explain, at least in part, the variability in absorption rate constants observed."( Use of nonlinear mixed effect modeling for the intestinal absorption data: application to ritonavir in the rat.
Casabó, VG; Lledó-García, R; Máñez-Castillejo, FJ; Merino-Sanjuán, M; Muñoz, MJ; Nácher, A, 2005
)
0.55
" Values for the clearance, volume of distribution and the absorption rate constant were 46."( Population pharmacokinetics of indinavir alone and in combination with ritonavir in HIV-1-infected patients.
Beijnen, JH; Huitema, AD; Kappelhoff, BS; Meenhorst, PL; Mulder, JW; Prins, JM; Sankatsing, SU; Van Gorp, EC, 2005
)
0.56
" The model parameters volume of distribution and absorption rate constant were 61."( Population pharmacokinetics of lopinavir in combination with ritonavir in HIV-1-infected patients.
Beijnen, JH; Crommentuyn, KM; Huitema, AD; Kappelhoff, BS; Mairuhu, AT; Meenhorst, PL; Mulder, JW; van Gorp, EC, 2005
)
0.57
"9 L), absorption rate constant (0."( No significant influence of saquinavir hard-gel capsule administration on pharmacokinetics of lopinavir in combination with ritonavir: a population approach.
Allavena, C; Dailly, E; Gagnieu, MC; Jolliet, P; Raffi, F, 2005
)
0.54
" The relative bioavailability of FCT SOV versus HC SQV was calculated as the ratio of the respective estimated mean saquinavir AUC0-alpha and Cmax."( Saquinavir 500 mg film-coated tablets demonstrate bioequivalence to saquinavir 200 mg hard capsules when boosted with twice-daily ritonavir in healthy volunteers.
Bittner, B; Grange, S; Holmes, B; Riek, M, 2005
)
0.53
" The fexofenadine AUC(infinity) was increased by lopinavir/ritonavir, likely due to increased bioavailability secondary to P-glycoprotein inhibition."( Time-dependent interaction between lopinavir/ritonavir and fexofenadine.
Bourbeau, M; Cameron, DW; Campbell, P; Chauhan, BM; Foster, BC; Seguin, I; van Heeswijk, RP, 2006
)
0.84
"Cyclic ureas form a perspective class of non-peptidic HIV-1 protease inhibitors with major bioavailability problems."( P-glycoprotein effects of cyclic urea HIV protease inhibitor DMP 323 in competitional absorption studies.
Gyémánt, N; Hilgeroth, A; Molnár, J; Richter, M, 2006
)
0.33
"The aim of this study is to investigate in vivo the oral bioavailability of ritonavir and to evaluate the pharmacokinetic model that best describes the plasma concentration behavior after oral and intravenous administration."( Bioavailability and pharmacokinetic model for ritonavir in the rat.
Casabó, VG; Lledó-García, R; Merino-Sanjuán, M; Nácher, A; Prats-García, L, 2007
)
0.83
"Lopinavir, an HIV protease inhibitor, is coformulated with ritonavir to enhance the bioavailability and pharmacokinetics of lopinavir."( The tablet formulation of lopinavir/ritonavir provides similar bioavailability to the soft-gelatin capsule formulation with less pharmacokinetic variability and diminished food effect.
Awni, W; Breitenbach, J; Brun, SC; Chiu, YL; Doan, T; Hanna, GJ; Heuser, RS; Klein, CE; Morris, JB; Zhu, T, 2007
)
0.86
" Capravirine was well absorbed in rats but only moderately so in dogs."( Evaluation of capravirine as a CYP3A probe substrate: in vitro and in vivo metabolism of capravirine in rats and dogs.
Bu, HZ; Kang, P; Pool, WF; Shetty, BV; Wu, EY; Zhao, P, 2007
)
0.34
" Coadministration with small doses of the strong CYP3A4 inhibitor ritonavir results in an increase in darunavir bioavailability from 37% to 82%."( Clinical pharmacokinetics of darunavir.
Arastéh, K; Rittweger, M, 2007
)
0.58
" A population analysis done in NONMEM with a time-dependent covariate confirmed that ritonavir did not influence the clearance or bioavailability of imatinib."( Influence of CYP3A4 inhibition on the steady-state pharmacokinetics of imatinib.
Baker, SD; Gelderblom, H; Guchelaar, HJ; Karlsson, MO; Li, J; Nortier, JW; Ouwerkerk, J; Sparreboom, A; van Erp, NP; Zhao, M, 2007
)
0.56
"This investigation was carried out to evaluate the bioavailability of a new single fixed-dose combination formulation of lopinavir and ritonavir, relative to reference product, Kaletra (133."( A bioequivalence study comparing two formulations of lopinavir/ritonavir capsules.
Gurule, S; Khuroo, AH; Lao, VK; Mishra, S; Monif, T; Raghuvanshi, R; Thudi, NR; Tippabhotla, SK, 2008
)
0.79
" The results indicated that lopinavir bioavailability was not affected by the coadministration of omeprazole or ranitidine."( Effects of acid-reducing agents on the pharmacokinetics of lopinavir/ritonavir and ritonavir-boosted atazanavir.
Beck, K; Cai, Y; Causemaker, SJ; Chiu, YL; Doan, T; Esslinger, HU; Hanna, GJ; King, KR; Klein, CE; Podsadecki, TJ, 2008
)
0.58
"Atazanavir (ATV) is clinically coadministered with low-dose ritonavir (RTV), which boosts the oral bioavailability (BA) of ATV by inhibiting cytochrome P450 (CYP) 3A, and P-glycoprotein (Pgp) via the same metabolic pathway; however, it is well known that in the chronic phase, the inhibition effect of RTV on Pgp and CYP3A becomes an induction effect."( Long-term pharmacokinetic efficacy and safety of low-dose ritonavir as a booster and atazanavir pharmaceutical formulation based on solid dispersion system in rats.
Fukushima, K; Haraya, K; Ito, Y; Sugioka, N; Takada, K; Terasaka, S, 2008
)
0.83
" In conclusion, TAT-conjugated NPs enhanced the CNS bioavailability of the encapsulated PI and maintained therapeutic drug levels in the brain for a sustained period that could be effective in reducing the viral load in the CNS, which acts as a reservoir for the replicating HIV-1 virus."( TAT-conjugated nanoparticles for the CNS delivery of anti-HIV drugs.
Horning, JL; Labhasetwar, V; Rao, KS; Reddy, MK, 2008
)
0.35
" This observed nonlinearity in CL/F resulted from the increased bioavailability attributed to a saturated absorption and/or elimination process at higher doses."( Pharmacokinetic and safety evaluation of BILR 355, a second-generation nonnucleoside reverse transcriptase inhibitor, in healthy volunteers.
Hattox, S; Huang, F; Koenen-Bergmann, M; Macgregor, TR; Ring, A; Robinson, P, 2008
)
0.35
" The decrease in elvucitabine bioavailability when elvucitabine was coadministered with ritonavir may be due to ritonavir's inhibiting influx gut transporters."( Effect of a single dose of ritonavir on the pharmacokinetic behavior of elvucitabine, a nucleoside reverse transcriptase inhibitor, administered in healthy volunteers.
Colucci, P; Ducharme, MP; Pottage, JC; Robison, H; Turgeon, J, 2009
)
0.87
" Consequently, the coadministration of elvitegravir with the protease inhibitor ritonavir (a substantial CYP3A4 inhibitor) results in significantly enhanced bioavailability and a longer half-life than with elvitegravir alone, allowing for the once-daily dosing of elvitegravir."( Elvitegravir, an oral HIV integrase inhibitor, for the potential treatment of HIV infection.
Klibanov, OM, 2009
)
0.58
" Methadone bioavailability was unchanged, despite inhibition of gastrointestinal P-glycoprotein activity, suggesting that this transporter does not limit methadone intestinal absorption."( Methadone pharmacokinetics are independent of cytochrome P4503A (CYP3A) activity and gastrointestinal drug transport: insights from methadone interactions with ritonavir/indinavir.
Bedynek, PS; Hoffer, C; Kharasch, ED; Walker, A; Whittington, D, 2009
)
0.55
"In this study, we explored the bioavailability in dogs and chemical potency of generic ritonavir and lopinavir/ritonavir tablet products manufactured by various pharmaceutical companies."( Bioavailability of generic ritonavir and lopinavir/ritonavir tablet products in a dog model.
Garren, KW; Marsh, K; Morris, JB; Rahim, S, 2010
)
0.88
" Bioavailability was unchanged despite significant inhibition of intestinal P-glycoprotein."( Mechanism of ritonavir changes in methadone pharmacokinetics and pharmacodynamics: I. Evidence against CYP3A mediation of methadone clearance.
Bedynek, PS; Hoffer, C; Kharasch, ED; Park, S; Walker, A; Whittington, D, 2008
)
0.72
"51 to 0); and increased bioavailability (from 37 to 95%)."( Mechanism of ritonavir changes in methadone pharmacokinetics and pharmacodynamics: II. Ritonavir effects on CYP3A and P-glycoprotein activities.
Bedynek, PS; Hoffer, C; Kharasch, ED; Walker, A; Whittington, D, 2008
)
0.72
" Modification of the core regiochemistry and stereochemistry significantly affected the potency, metabolic stability, and oral bioavailability of the inhibitors, as did the variation of a pendent arylmethyl P3 group."( 2-Pyridyl P1'-substituted symmetry-based human immunodeficiency virus protease inhibitors (A-792611 and A-790742) with potential for convenient dosing and reduced side effects.
Chen, HJ; Colletti, L; Degoey, DA; Dekhtyar, T; Flentge, CA; Flosi, WJ; Grampovnik, DJ; Kati, WM; Kempf, DJ; Klein, LL; Mamo, M; Marsh, KC; Mo, H; Molla, A; Morfitt, DC; Nguyen, B; Randolph, JT; Schmidt, JM; Stoll, V; Swanson, SJ; Yeung, CM, 2009
)
0.35
" The apparent oral bioavailability of docetaxel combined with ritonavir simultaneously or ritonavir given 60 minutes before docetaxel was 131% +/- 90% and 161% +/- 91%, respectively."( Coadministration of ritonavir strongly enhances the apparent oral bioavailability of docetaxel in patients with solid tumors.
Beijnen, JH; Huitema, A; Jansen, RS; Keessen, M; Oostendorp, RL; Rosing, H; Schellens, JH; Ter Heine, R, 2009
)
0.92
"Coadministration of ritonavir significantly enhanced the apparent oral bioavailability of docetaxel."( Coadministration of ritonavir strongly enhances the apparent oral bioavailability of docetaxel in patients with solid tumors.
Beijnen, JH; Huitema, A; Jansen, RS; Keessen, M; Oostendorp, RL; Rosing, H; Schellens, JH; Ter Heine, R, 2009
)
1
" Low bioavailability and extensive first-pass metabolism make benzimidazoles prone to pharmacokinetic drug interactions."( Effect of ritonavir on the pharmacokinetics of the benzimidazoles albendazole and mebendazole: an interaction study in healthy volunteers.
Corti, N; Heck, A; Jetter, A; Pauli-Magnus, C; Rentsch, K; Stieger, B; Zingg, W, 2009
)
0.76
" The ritonavir dose-dependence of boosting effects did not correlate with their bioavailability or their effects on ritonavir plasma levels."( How much ritonavir is needed to boost protease inhibitors? Systematic review of 17 dose-ranging pharmacokinetic trials.
Boffito, M; Hill, A; Sawyer, W; van der Lugt, J, 2009
)
1.28
"Oral bioavailability (F) is a product of fraction absorbed (Fa), fraction escaping gut-wall elimination (Fg), and fraction escaping hepatic elimination (Fh)."( Physicochemical space for optimum oral bioavailability: contribution of human intestinal absorption and first-pass elimination.
Chang, G; El-Kattan, A; Miller, HR; Obach, RS; Rotter, C; Steyn, SJ; Troutman, MD; Varma, MV, 2010
)
0.36
" Oral bioavailability of 10 mg of ritonavir in solid dispersion prepared by solvent evaporation (SE1) and melt method (MM1) was compared with pure drug after oral administration of solid dispersion and pure drug to Albino Wistar rats of either sex."( Solid dispersion as an approach for bioavailability enhancement of poorly water-soluble drug ritonavir.
Ahuja, A; Ali, J; Ali, M; Baboota, S; Kumar, A; Sinha, S, 2010
)
0.86
" The results indicated that incorporation of antioxidants in this new SMEDDS not only significantly reduced ritonavir-induced ER stress activation, ROS production and apoptosis in intestinal epithelial cells and macrophages, but also improved the solubility, stability and bioavailability of ritonavir, and significantly reduced ritonavir-induced disruption of intestinal barrier function in vivo."( Development of a novel self-microemulsifying drug delivery system for reducing HIV protease inhibitor-induced intestinal epithelial barrier dysfunction.
Jin, F; Lei, B; Studer, EJ; Wang, G; Wang, X; Wang, Y; Wen, C; Zha, W; Zhang, L; Zhou, H, 2010
)
0.57
" Meltrex) have proven to be a promising formulation tool for poorly water-soluble and poorly bioavailable drugs."( In situ formation of nanoparticles upon dispersion of melt extrudate formulations in aqueous medium assessed by asymmetrical flow field-flow fractionation.
Brandl, M; Fricker, G; Hölig, P; Hupfeld, S; Kanzer, J; Mägerlein, M; Tho, I; Vasskog, T, 2010
)
0.36
"Docetaxel has a low oral bioavailability due to affinity for P-glycoprotein and cytochrome P450 (CYP) 3A4 enzymes."( Population pharmacokinetics of intravenously and orally administered docetaxel with or without co-administration of ritonavir in patients with advanced cancer.
Beijnen, JH; Huitema, AD; Koolen, SL; Oostendorp, RL; Schellens, JH, 2010
)
0.57
"Gut bioavailability of docetaxel increased approximately two-fold from 19 to 39% (CV 13%) with ritonavir co-administration."( Population pharmacokinetics of intravenously and orally administered docetaxel with or without co-administration of ritonavir in patients with advanced cancer.
Beijnen, JH; Huitema, AD; Koolen, SL; Oostendorp, RL; Schellens, JH, 2010
)
0.79
" Relative bioavailability of ritonavir was low, when compared with other ritonavir regimens."( Intracellular and plasma steady-state pharmacokinetics of raltegravir, darunavir, etravirine and ritonavir in heavily pre-treated HIV-infected patients.
Beijnen, JH; Huitema, AD; Mulder, JW; Ter Heine, R; van Gorp, EC; Wagenaar, JF, 2010
)
0.87
" We also evaluated whether ritonavir increases lopinavir oral bioavailability by inhibition of CYP3A, ABCB1 and/or ABCC2."( Effects of cytochrome P450 3A (CYP3A) and the drug transporters P-glycoprotein (MDR1/ABCB1) and MRP2 (ABCC2) on the pharmacokinetics of lopinavir.
Beijnen, JH; Huitema, AD; Rooswinkel, RW; Schinkel, AH; ter Heine, R; van der Kruijssen, CM; van Waterschoot, RA; Wagenaar, E, 2010
)
0.66
" Both intestinal and hepatic CYP3A activity contributed importantly to low oral bioavailability of lopinavir."( Effects of cytochrome P450 3A (CYP3A) and the drug transporters P-glycoprotein (MDR1/ABCB1) and MRP2 (ABCC2) on the pharmacokinetics of lopinavir.
Beijnen, JH; Huitema, AD; Rooswinkel, RW; Schinkel, AH; ter Heine, R; van der Kruijssen, CM; van Waterschoot, RA; Wagenaar, E, 2010
)
0.36
" Based on this study, coadministration of investigated compounds with garlic supplements could result in significant in vivo modification of hepatic transport-enzyme interplay, possibly leading to further bioavailability change."( The influence of aged garlic extract on the uptake of saquinavir and darunavir into HepG2 cells and rat liver slices.
Berginc, K; Kristl, A; Trontelj, J, 2010
)
0.36
" It also demonstrated that low oral bioavailability of saquinavir is due mainly to intestinal rather than to hepatic first-pass metabolism."( A pharmacokinetic model for evaluating the impact of hepatic and intestinal first-pass loss of saquinavir in the rat.
Casabó, VG; Lledó-García, R; Merino-Sanjuán, M; Nácher, A, 2011
)
0.37
" The selective CYP3A4 inhibitors, ketoconazole, troleandomycin and ritonavir demonstrated significant inhibitory effects on CMDCK intestinal metabolism, which suggested that co-administration of CMDCK with potent CYP3A inhibitors, such as ritonavir, might decrease its intestinal metabolic clearance and subsequently improve its bioavailability in body."( [Metabolism of 3-cyanomethyl-4-methyl-DCK, a new anti-HIV candidate, in human intestinal microsomes].
Cui, SL; Deng, JT; Kong, WL; Li, H; Tian, XT; Wen, YY; Xie, L; Zhuang, XM, 2010
)
0.6
"0%), and the apparent volume of distribution and absorption rate constant were 55."( Sequential population pharmacokinetic modeling of lopinavir and ritonavir in healthy volunteers and assessment of different dosing strategies.
Aarons, L; Back, D; Boffito, M; Davies, G; Dickinson, L; Else, L; Khoo, S; Moyle, G; Pozniak, A; von Hentig, N, 2011
)
0.61
" Because docetaxel has a very low permeability and a very low aqueous solubility (biopharmaceutical classification system class IV), a pharmacokinetic booster was combined with a newly developed solid dispersion formulation to improve the oral bioavailability of docetaxel."( Pharmaceutical development and preliminary clinical testing of an oral solid dispersion formulation of docetaxel (ModraDoc001).
Beijnen, JH; Huitema, AD; Koolen, SL; Moes, JJ; Nuijen, B; Schellens, JH, 2011
)
0.37
" LPV alone suffers the poor bioavailability due to its rapid and extensive metabolism."( CYP3A4-mediated lopinavir bioactivation and its inhibition by ritonavir.
Li, F; Lu, J; Ma, X, 2012
)
0.62
" Rifampicin reduced the oral bioavailability of lopinavir and ritonavir by 20% and 45% respectively, and it increased their clearance by 71% and 36% respectively."( Model-based approach to dose optimization of lopinavir/ritonavir when co-administered with rifampicin.
Decloedt, E; Denti, P; Karlsson, MO; Maartens, G; McIlleron, H; Simonsson, US; Zhang, C, 2012
)
0.87
" The higher trough concentrations observed in the morning were explained by both higher bioavailability with the evening meal and lower clearance overnight."( Model-based approach to dose optimization of lopinavir/ritonavir when co-administered with rifampicin.
Decloedt, E; Denti, P; Karlsson, MO; Maartens, G; McIlleron, H; Simonsson, US; Zhang, C, 2012
)
0.63
" Antitubercular treatment reduced the oral bioavailability of lopinavir by 77% in children receiving twice usual lopinavir/ritonavir doses and increased ritonavir clearance by 50%."( Population pharmacokinetics of lopinavir and ritonavir in combination with rifampicin-based antitubercular treatment in HIV-infected children.
Denti, P; Karlsson, MO; McIlleron, H; Ren, Y; Simonsson, US; van der Walt, JS; Zhang, C, 2012
)
0.85
"Lopinavir (LPV), a newer HIV protease inhibitor, has poor bioavailability being a substrate of both cytochrome P450 3A enzyme system (CYP3A) and permeability-glycoprotein (P-gp)."( Effect of grapefruit juice and ritonavir on pharmacokinetics of lopinavir in Wistar rats.
Aditya, N; Ravi, PR; Srivani, S; Thakur, R; Vats, R, 2012
)
0.66
" Genetic association analysis was performed with PLINK using the relative bioavailability as the phenotype."( Population pharmacokinetic analysis and pharmacogenetics of raltegravir in HIV-positive and healthy individuals.
Aouri, M; Arab-Alameddine, M; Boffito, M; Buclin, T; Cavassini, M; Csajka, C; Decosterd, LA; di Iulio, J; Fayet-Mello, A; Günthard, HF; Lubomirov, R; Neely, M; Owen, A; Rentsch, K; Rotger, M; Telenti, A, 2012
)
0.38
"Eltrombopag is an orally bioavailable thrombopoietin receptor agonist that is approved for the treatment of chronic idiopathic thrombocytopenic purpura."( Assessment of the pharmacokinetic interaction between eltrombopag and lopinavir-ritonavir in healthy adult subjects.
McLean, HB; Park, JW; Pendry, C; Peng, B; Theodore, D; Wire, MB, 2012
)
0.61
" Their bioavailability may be impaired because they are substrates of CYP3A4 and several transporters, including P-glycoprotein."( Boosting of HIV protease inhibitors by ritonavir in the intestine: the relative role of cytochrome P450 and P-glycoprotein inhibition based on Caco-2 monolayers versus in situ intestinal perfusion in mice.
Annaert, P; Augustijns, P; Holmstock, N, 2012
)
0.65
"  Ritonavir dramatically increases the bioavailability of a variety of concurrently administered drugs by inhibition of metabolic enzymes and drug transporters."( Analysis of the pharmacokinetic boosting effects of ritonavir on oral bioavailability of drugs in mice.
Banba, H; Takayama, K; Takeda-Morishita, M; Tomaru, A, 2013
)
1.36
" Unlike paclitaxel, docetaxel is extensively metabolized by CYP3A4 and its oral bioavailability can be enhanced in mice and humans by coadministration of the potent CYP3A inhibitor ritonavir."( P-glycoprotein and cytochrome P450 3A act together in restricting the oral bioavailability of paclitaxel.
Beijnen, JH; Hendrikx, JJ; Lagas, JS; Rosing, H; Schellens, JH; Schinkel, AH, 2013
)
0.58
"The aim of the present study was to prepare surface-stabilized nanoparticles (NPs) for oral bioavailability enhancement of lopinavir (LPN), a Biopharmaceutics Classification System class II antiretroviral drug that possesses low oral bioavailability due to its poor aqueous solubility and extensive metabolism by liver microsomal enzymes."( Surface-stabilized lopinavir nanoparticles enhance oral bioavailability without coadministration of ritonavir.
Jain, AK; Jain, S; Mahajan, RR; Sharma, JM, 2013
)
0.61
"11-fold enhancement in bioavailability in comparison to free LPN with ritonavir (conventional formulation)."( Surface-stabilized lopinavir nanoparticles enhance oral bioavailability without coadministration of ritonavir.
Jain, AK; Jain, S; Mahajan, RR; Sharma, JM, 2013
)
0.84
"The bioavailability of lopinavir was reduced by 25% in adults whereas children on antituberculosis treatment experienced a 59% reduction, an effect that was moderated by the dose of ritonavir."( Model-based evaluation of the pharmacokinetic differences between adults and children for lopinavir and ritonavir in combination with rifampicin.
Decloedt, EH; Denti, P; Karlsson, MO; McIlleron, H; Ren, Y; Zhang, C, 2013
)
0.8
" The present study is aimed to enhance the dissolution and oral bioavailability of water-insoluble RTV using the Solid Self-Microemulsifying Drug Delivery System (S-SMEDDS)."( Solid self-microemulsifying drug delivery system of ritonavir.
Deshmukh, A; Kulkarni, S, 2014
)
0.65
"To enhance the dissolution and oral bioavailability of water-insoluble RTV using the S-SMEDDS."( Solid self-microemulsifying drug delivery system of ritonavir.
Deshmukh, A; Kulkarni, S, 2014
)
0.65
" Relative bioavailability of RTV was determined in male Wistar rats and pharmacokinetic parameters were calculated by the comparison of optimized S-SMEDDS versus aqueous suspension of RTV."( Solid self-microemulsifying drug delivery system of ritonavir.
Deshmukh, A; Kulkarni, S, 2014
)
0.65
"S-SMEDDS tablet of RTV was formulated successfully by adsorbing optimized L-SMEDDS of RTV on Neusilin-US2(®) as a potential carrier with enhanced solubility and relative oral bioavailability compared to pure RTV by twofolds."( Solid self-microemulsifying drug delivery system of ritonavir.
Deshmukh, A; Kulkarni, S, 2014
)
0.65
" The oral bioavailability of CMDCK was increased from 15% of the control group to 45% of the RTV concomitant group (2."( Investigation of the pharmacokinetic interaction between ritonavir and CMDCK, a new non-nucleoside reverse transcriptase inhibitor.
Li, H; Shen, GL; Yuan, M; Zhuang, XM, 2013
)
0.64
" However, when this glucocorticoid agent is given to patients receiving the HIV protease inhibitor (PI) ritonavir (RTV),inhibition of their shared cytochrome P450 3A4 degradation pathway leads to an increased bioavailability of triamcinolone, with subsequent heightening and prolongation of the glucocorticoid serum levels."( Iatrogenic Cushing syndrome and secondary adrenal insufficiency related to concomitant triamcinolone and ritonavir administration: a case report and review.
Bush, LM; Schroeder, JR; Song, Y,
)
0.56
"Danoprevir is a potent, highly selective, macrocyclic, orally bioavailable inhibitor of the hepatitis C virus protease, and a substrate of cytochrome P450 (CYP) 3A."( Two-way interaction study between ritonavir boosted danoprevir, a potent HCV protease inhibitor, and ketoconazole in healthy subjects.
Brennan, BJ; Chang, L; Chung, D; Morcos, PN; Navarro, M; Smith, PF; Tran, JQ, 2014
)
0.68
" The principal aim of this study was to compare bioavailability of two doses of rifabutin (150 mg three times per week and 150 mg daily) in patients with HIV-associated tuberculosis who initiated lopinavir/ritonavir-based antiretroviral therapy in Vietnam."( Randomised pharmacokinetic trial of rifabutin with lopinavir/ritonavir-antiretroviral therapy in patients with HIV-associated tuberculosis in Vietnam.
Barrail-Tran, A; Borand, L; Connolly, C; Duc, NH; Dung, NH; Harries, AD; Lagarde, D; Lan, NN; Lan, NT; Laureillard, D; Lien, TT; Lienhardt, C; Pym, A; Quillet, C; Taburet, AM; Thu, NT, 2014
)
0.83
" The objective of this animal study was to determine whether pre-treatment with antibiotics affects the intestinal bioavailability of Atazanavir (ATV) and Ritonavir (RTV)."( Metronidazole or Cotrimoxazole therapy is associated with a decrease in intestinal bioavailability of common antiretroviral drugs.
Desjeux, JF; Dossou-Yovo, F; Eto, B; Limas-Nzouzi, N; Mamadou, G; Miantezila, J; Soudy, ID, 2014
)
0.6
"This study compared the bioavailability of two candidate fixed-dose combinations (FDCs: G003 and G004) of darunavir/cobicistat 800/150 mg with that of darunavir 800 mg and ritonavir 100 mg coadministered as single agents."( Pharmacokinetics of darunavir in fixed-dose combination with cobicistat compared with coadministration of darunavir and ritonavir as single agents in healthy volunteers.
Hillewaert, V; Hoetelmans, RM; Iterbeke, K; Kakuda, TN; Opsomer, M; Petrovic, R; Timmers, M; Van De Casteele, T, 2014
)
0.8
" Significant increase in oral bioavailability of LPV from LPV SLNs (5 folds) and LPV/RTV coformulation (3."( A hybrid design to optimize preparation of lopinavir loaded solid lipid nanoparticles and comparative pharmacokinetic evaluation with marketed lopinavir/ritonavir coformulation.
Dalal, V; Murthy, AN; Ravi, PR; Vats, R, 2014
)
0.6
"SLNs enhanced oral bioavailability and improved distribution profile of LPV to HIV reservoirs and hence could be better alternative to LPV/RTV coformulation."( A hybrid design to optimize preparation of lopinavir loaded solid lipid nanoparticles and comparative pharmacokinetic evaluation with marketed lopinavir/ritonavir coformulation.
Dalal, V; Murthy, AN; Ravi, PR; Vats, R, 2014
)
0.6
" The resulting low oral bioavailability can be boosted by co-administration of P-gp or CYP3A4 inhibitors."( Oral co-administration of elacridar and ritonavir enhances plasma levels of oral paclitaxel and docetaxel without affecting relative brain accumulation.
Beijnen, JH; Hendrikx, JJ; Lagas, JS; Rosing, H; Schellens, JH; Schinkel, AH; Wagenaar, E, 2014
)
0.67
"The EFV-based HAART regimen was associated with a reduction in the bioavailability of ENG, which showed decreases of 63."( Effect of antiretroviral therapy including lopinavir/ritonavir or efavirenz on etonogestrel-releasing implant pharmacokinetics in HIV-positive women.
Amaral, E; Bahamondes, L; Bahamondes, MV; Brito, MB; de Souza, RM; Duarte, G; Ferriani, RA; Quintana, SM; Rocha Prandini, TR; Scaranari, C; Vieira, CS, 2014
)
0.65
"The coadministration of EFV decreased the bioavailability of ENG released from the implant, which could impair contraceptive efficacy."( Effect of antiretroviral therapy including lopinavir/ritonavir or efavirenz on etonogestrel-releasing implant pharmacokinetics in HIV-positive women.
Amaral, E; Bahamondes, L; Bahamondes, MV; Brito, MB; de Souza, RM; Duarte, G; Ferriani, RA; Quintana, SM; Rocha Prandini, TR; Scaranari, C; Vieira, CS, 2014
)
0.65
"An open-label comparative bioavailability (randomized crossover) study compared a novel twice-daily minitab sprinkle formulation (40 mg/10 mg, Cipla Pharmaceuticals) versus innovator syrup in HIV-infected Ugandan infants aged 3 to <12 months (cohort A) and children aged 1-4 years (cohort B) and versus Cipla tablets (100/25 mg) in children aged 4 to <13 years (cohort C)."( The pharmacokinetics and acceptability of lopinavir/ritonavir minitab sprinkles, tablets, and syrups in african HIV-infected children.
Burger, D; Fillekes, Q; Gibb, DM; Keishanyu, R; Kekitiinwa, A; Kendall, L; Lallemant, M; Musiime, V; Namuddu, R; Opilo, W; Walker, AS; Young, N, 2014
)
0.65
"To assess the relative oral bioavailability (NCT01052883) and bioequivalence (NCT01308658) of a darunavir 800-mg tablet vs."( Bioavailability and bioequivalence of a darunavir 800-mg tablet formulation compared with the 400-mg tablet formulation.
Hillewaert, V; Hoetelmans, RM; Kakuda, TN; Leopold, L; Timmers, M; Tomaka, FL; Van De Casteele, T, 2014
)
0.4
"00%, except bioavailability AUC(0-∞) (fed and fasted conditions)."( Bioavailability and bioequivalence of a darunavir 800-mg tablet formulation compared with the 400-mg tablet formulation.
Hillewaert, V; Hoetelmans, RM; Kakuda, TN; Leopold, L; Timmers, M; Tomaka, FL; Van De Casteele, T, 2014
)
0.4
" Protease inhibitors (PIs), an important component of the antiretroviral armada, were historically associated with poor oral bioavailability and high pill burden."( Pharmacokinetic enhancers in HIV therapeutics.
Acosta, EP; Delille, C; Larson, KB; Otofokun, I; Wang, K, 2014
)
0.4
" Concentrations of Arga-L® at 12 h after intake were considerably lower than those of Kaletra®, revealing very low oral bioavailability of generic lopinavir and ritonavir (<10%) compared to the brand-name drug."( [Lack of bioavailability of generic lopinavir/ritonavir not prequalified by WHO marketed in Africa (Congo Brazzaville)].
Camara, S; Goudjo, A; Guillard, E; Peytavin, G; Vasse, M; Zucman, D, 2015
)
0.87
" Significant covariates in the model included RTV coadministration on clearance, fed status on bioavailability for the oral suspension, body weight on clearance and volume terms, black race on clearance, and age on clearance."( Population pharmacokinetic modeling and simulation of amprenavir following fosamprenavir/ritonavir administration for dose optimization in HIV infected pediatric patients.
Barbour, AM; Gibiansky, L; Wire, MB, 2014
)
0.62
" Absolute bioavailability of danoprevir 100 mg was low (1."( Characterization of the transmembrane transport and absolute bioavailability of the HCV protease inhibitor danoprevir.
Asthappan, J; Brennan, BJ; Funk, C; Goelzer, P; Morcos, PN; Moreira, S; Poirier, A; Portmann, R; Smith, PF, 2015
)
0.42
" However, when this glucocorticoid agent is given to patients receiving the HIV protease inhibitor (PI) ritonavir (RTV),inhibition of their shared cytochrome P450 3A4 degradation pathway leads to an increased bioavailability of triamcinolone, with subsequent heightening and prolongation of the glucocorticoid serum levels."( Iatrogenic Cushing syndrome and secondary adrenal insufficiency related to concomitant triamcinolone and ritonavir administration: a case report and review.
Bush, LM; Schroeder, JR; Song, Y,
)
0.56
" FA receptor-targeted poloxamer 407 nanocrystals, containing ritonavir-boosted atazanavir (ATV/r), significantly increased drug bioavailability and PD by five and 100 times, respectively."( Pharmacodynamics of long-acting folic acid-receptor targeted ritonavir-boosted atazanavir nanoformulations.
Bade, AN; Baldridge, HM; Balkundi, SS; Dash, PK; Dimitrov, DS; Feng, Y; Gendelman, HE; Gorantla, S; Hilaire, JR; Kendrick, LM; Liu, XM; McMillan, JM; Poluektova, LY; Puligujja, P; Wang, Y; Ying, T; Zhang, G, 2015
)
0.9
" In vivo oral bioavailability study was conducted for 1:2 binary amorphous form by using pure RTV as a control."( Fabrication, solid state characterization and bioavailability assessment of stable binary amorphous phases of Ritonavir with Quercetin.
Bhat, K; Dengale, SJ; Gautham Shenoy, G; Hussen, SS; Krishna, BS; Musmade, PB, 2015
)
0.63
" Bioavailability comparisons were assessed by the 90% CIs for the geometric least-squares mean ratios."( A novel ritonavir paediatric powder formulation is bioequivalent to ritonavir oral solution with a similar food effect.
Chiu, YL; Klein, CE; Nilius, AM; Salem, AH; Valdes, JM, 2015
)
0.85
"To evaluate the effect of ritonavir (RTV) co-administration on the bioavailability of an amorphous dispersion of acetyl-11-keto-beta-boswellic acid (AKBA) and to develop a pharmaceutically acceptable AKBA-RTV combination tablet."( Bioavailability enhancement of a BCS IV compound via an amorphous combination product containing ritonavir.
Brough, C; Cisneros, M; Ellenberger, DJ; Keen, JM; McGinity, JW; Miller, DA; Williams, RO, 2016
)
0.95
"A pharmacokinetic (PK) study in rats was conducted to evaluate the influence of RTV co-administration on the oral bioavailability of an AKBA amorphous dispersion."( Bioavailability enhancement of a BCS IV compound via an amorphous combination product containing ritonavir.
Brough, C; Cisneros, M; Ellenberger, DJ; Keen, JM; McGinity, JW; Miller, DA; Williams, RO, 2016
)
0.65
" The AKBA-RTV combination tablets yielded a substantial increase in AKBA's bioavailability in dogs."( Bioavailability enhancement of a BCS IV compound via an amorphous combination product containing ritonavir.
Brough, C; Cisneros, M; Ellenberger, DJ; Keen, JM; McGinity, JW; Miller, DA; Williams, RO, 2016
)
0.65
" Consequently, AKBA's oral bioavailability is maximized by administration from a supersaturating formulation in conjunction with a CYP3A inhibitor."( Bioavailability enhancement of a BCS IV compound via an amorphous combination product containing ritonavir.
Brough, C; Cisneros, M; Ellenberger, DJ; Keen, JM; McGinity, JW; Miller, DA; Williams, RO, 2016
)
0.65
" LPV bioavailability is increased by co-formulated ritonavir (r), which may enhance the interaction of INH on LPV."( The pharmacokinetics of lopinavir/ritonavir when given with isoniazid in South African HIV-infected individuals.
Decloedt, EH; Maartens, G; McIlleron, H; van der Walt, JS; Wiesner, L, 2015
)
0.95
" Therapeutic drug monitoring (TDM) was performed as unboosted DRV has a bioavailability of 37% and the virologic and immunologic response has only been demonstrated with ritonavirboosted DRV."( The use of unboosted darunavir in the setting of ritonavir intolerance: three case reports.
Fulco, PP; Gomes, D; Nixon, D; Smith, K, 2016
)
0.88
" Simulations with apparent oral clearance increased by 71% and 36% and relative bioavailability decreased by 20% and 45% for darunavir and ritonavir, respectively, were performed for +rifampicin, 600 mg once daily (n = 1000)."( Simulation of the impact of rifampicin on once-daily darunavir/ritonavir pharmacokinetics and dose adjustment strategies: a population pharmacokinetic approach.
Back, D; Boffito, M; Dickinson, L; Khoo, S; Siccardi, M; Winston, A, 2016
)
0.88
"56-fold increase in bioavailability and significantly increased LPV concentrations in tested tissues, especially in HIV sanctuary sites, as compared to the commercial LPV/RTV tablet (Kaletra®) in rats."( Development and in vivo evaluation of child-friendly lopinavir/ritonavir pediatric granules utilizing novel in situ self-assembly nanoparticles.
Dong, X; Guo, S; Li, D; Penzak, S; Pham, K, 2016
)
0.67
"Water-soluble prodrug strategy is a practical alternative for improving the drug bioavailability of sparingly-soluble drugs with reduced drug efficacy."( Novel prodrugs with a spontaneous cleavable guanidine moiety.
Hamada, Y, 2016
)
0.43
" ombitasvir and dasabuvir) on paritaprevir bioavailability were included in the model."( Dose- and Formulation-Dependent Non-Linear Pharmacokinetic Model of Paritaprevir, a Protease Inhibitor for the Treatment of Hepatitis C Virus Infection: Combined Analysis from 12 Phase I Studies.
Awni, WM; Beck, D; Dutta, S; Khatri, A; Liu, W; Menon, RM; Mensing, S; Polepally, AR, 2016
)
0.43
" Paritaprevir bioavailability was formulation- and dose-dependent, and increased supraproportionally."( Dose- and Formulation-Dependent Non-Linear Pharmacokinetic Model of Paritaprevir, a Protease Inhibitor for the Treatment of Hepatitis C Virus Infection: Combined Analysis from 12 Phase I Studies.
Awni, WM; Beck, D; Dutta, S; Khatri, A; Liu, W; Menon, RM; Mensing, S; Polepally, AR, 2016
)
0.43
"Acid-reducing agents (ARAs) and proton-pump inhibitors (PPIs) that increase gastric pH can alter the bioavailability of antiviral drugs, particularly relevant in patients with advanced liver disease caused by chronic hepatitis C virus (HCV) infection seeking therapy."( Safety and Efficacy of Ombitasvir/Paritaprevir/Ritonavir Plus Dasabuvir With or Without Ribavirin in HCV-Infected Patients Taking Concomitant Acid-Reducing Agents.
Asselah, T; Bennett, M; Forns, X; Liu, L; Moller, J; Pedrosa, M; Planas Vila, R; Reau, N; Rustgi, V; Shiffman, ML, 2016
)
0.69
" We also used rats to test the bioavailability of RTV ISNP granules."( Novel in situ self-assembly nanoparticles for formulating a poorly water-soluble drug in oral solid granules, improving stability, palatability, and bioavailability.
Dong, X; Guo, S; Li, D; Penzak, SR; Pham, K, 2016
)
0.43
"Cytochrome P450 3A4 (CYP3A4) is the dominant P450 enzyme involved in human drug metabolism, and its inhibition may result in adverse interactions or, conversely, favorably reduce the systemic elimination rates of poorly bioavailable drugs."( N-Heterocyclic Carbene Capture by Cytochrome P450 3A4.
Hackett, JC; Jennings, GK; Lyons, CE; Ritchie, CM; Shock, LS, 2016
)
0.43
"This 2-part, phase 1, open-label, randomized, crossover study (NCT00752310) assessed ritonavir-boosted darunavir bioavailability (oral suspension vs."( Pharmacokinetics of darunavir after administration of an oral suspension with low-dose ritonavir and with or without food.
De Paepe, E; Hoetelmans, RM; Kakuda, TN; Lavreys, L; Sekar, V; Stevens, T; Vangeneugden, T; Vanstockem, M, 2014
)
0.85
" Co-administration with some antiretroviral therapies (ART) changes the bioavailability of the etonogestrel (ENG)-releasing contraceptive implant, possibly affecting the bleeding pattern."( Bleeding patterns of HIV-infected women using an etonogestrel-releasing contraceptive implant and efavirenz-based or lopinavir/ritonavir-based antiretroviral therapy.
Amaral, E; Bahamondes, L; Bahamondes, MV; Brito, MB; Duarte, G; Ferriani, RA; Prandini, TR; Quintana, SM; Ragazini, CS; Vieira, CS, 2016
)
0.64
" Encasement of MABC into poloxamer nanoparticles extended drug bioavailability for 2 weeks."( Development and characterization of a long-acting nanoformulated abacavir prodrug.
Alnouti, Y; Edagwa, B; Gautam, N; Gendelman, HE; Hilaire, J; McMillan, J; Palandri, D; Singh, D, 2016
)
0.43
" P-gp inhibitors could serve as helpful tools to enhance the oral bioavailability of those substances."( The application of P-gp inhibiting phospholipids as novel oral bioavailability enhancers - An in vitro and in vivo comparison.
Burhenne, J; Fricker, G; Mylius, P; Schubert, R; Weinheimer, M, 2017
)
0.46
"Cell membrane permeability is an important determinant for oral absorption and bioavailability of a drug molecule."( Highly predictive and interpretable models for PAMPA permeability.
Jadhav, A; Kerns, E; Nguyen, K; Shah, P; Sun, H; Xu, X; Yan, Z; Yu, KR, 2017
)
0.46
"Although the prevalence of alcohol consumption is higher in HIV+ people than general public, limited information is available on how alcohol affects the metabolism and bioavailability of darunavir (DRV)."( Influence of Ethanol on Darunavir Hepatic Clearance and Intracellular PK/PD in HIV-Infected Monocytes, and CYP3A4-Darunavir Interactions Using Inhibition and in Silico Binding Studies.
Cory, TJ; Gong, Y; Kumar, S; Li, J; Li, W; Meibohm, B; Midde, NM, 2017
)
0.46
"The AUC for the liposomes were found to be much higher in the spleen and thymus compared to that in the plasma which indicated that the developed formulations enhance the bioavailability and target specificity compared to that of the pure drug thereby enhancing bioavailability at target site."( Development and in vivo evaluation of functionalized ritonavir proliposomes for lymphatic targeting.
Ahammed, V; Narayan, R; Nayak, UY; Nayak, Y; Paul, J; Roy, B; Shavi, GV, 2017
)
0.7
"Lopinavir is a BCS Class IV drug exhibiting poor bioavailability due to P-gp efflux and limited permeation."( Improvement of Oral Bioavailability of Lopinavir Without Co-administration of Ritonavir Using Microspheres of Thiolated Xyloglucan.
Bhalekar, MR; Kadam, AA; Madgulkar, AR, 2018
)
0.71
" We hypothesized that the presence of multiple drugs would reduce crystallization tendency, thereby providing stable, supersaturating formulations for bioavailability enhancement."( Multidrug, Anti-HIV Amorphous Solid Dispersions: Nature and Mechanisms of Impacts of Drugs on Each Other's Solution Concentrations.
Arca, HÇ; Dahal, D; Edgar, KJ; Mosquera-Giraldo, LI; Taylor, LS, 2017
)
0.46
" MDR-TB treatment did not have a significant effect on the bioavailability, clearance, or absorption rate constants of lopinavir or ritonavir."( Pharmacokinetics and Drug-Drug Interactions of Lopinavir-Ritonavir Administered with First- and Second-Line Antituberculosis Drugs in HIV-Infected Children Treated for Multidrug-Resistant Tuberculosis.
de Kock, M; Denti, P; Garcia-Prats, AJ; Hesseling, AC; McIlleron, H; Norman, J; Schaaf, HS; Tikiso, T; van der Laan, LE; Wiesner, L; Winckler, J, 2018
)
0.93
"Objective Since the majority of direct-acting antivirals (DAAs) that are used in the treatment of hepatitis C virus (HCV) infection are mainly metabolized by CYP3A4, it is hypothesized that inter-individual differences in CYP3A4 activity may be associated with the bioavailability of these agents."( Differences in the Serum 4β-hydroxycholesterol Levels of Patients with Chronic Hepatitis C Virus (HCV) Infection: A Possible Impact on the Efficacy and Safety of Interferon (IFN)-free Treatment.
Hirayama, T; Honda, A; Ikegami, T; Kohjima, M; Matsuzaki, Y; Miyazaki, T; Nakamuta, M; Yara, SI, 2018
)
0.48
" Agreement was also obtained between the RTV concentrations in octanol at 3 h from these generic drug products and their corresponding relative bioavailability in dogs."( In vitro characterization of ritonavir formulations and correlation to in vivo performance in dogs.
Gao, P; Krakow, S; Rosenberg, J; Shi, Y; Xu, H, 2018
)
0.77
"Amorphous solid dispersions (ASDs) are a promising formulation strategy to increase both the apparent aqueous solubility and bioavailability of poorly water-soluble drugs."( Mechanistic understanding of the phase behavior of supersaturated solutions of poorly water-soluble drugs.
Hall, SD; Mohutsky, MA; Posada, MM; Taylor, LS; Tres, F, 2018
)
0.48
"Nanonizationhas been extensively investigated to increase theoral bioavailability of hydrophobicdrugsin general andantiretrovirals(ARVs)used inthe therapy of the human immunodeficiency virus (HIV) infection in particular."( Nanoparticle-in-microparticle oral drug delivery system of a clinically relevant darunavir/ritonavir antiretroviral combination.
Arzi, RS; Ashkenazi, DL; Augustine, R; Shofti, R; Sosnik, A; Zlobin, V, 2018
)
0.7
"This work aimed to assess the safety, tolerability, pharmacokinetics (PK), and relative bioavailability of GSK2838232, an investigational HIV maturation inhibitor."( The safety, tolerability, and pharmacokinetic profile of GSK2838232, a novel 2nd generation HIV maturation inhibitor, as assessed in healthy subjects.
Baldwin, S; Burke, M; Davies, M; Gould, E; Jeffrey, J; Jewell, RC; Johns, BA; Johnson, M; Lou, Y; Peppercorn, A; Tenorio, AR; Xu, J, 2018
)
0.48
" The findings described herein are highly significant in aiding our understanding of ordered mesoporous silica as a supersaturating drug delivery system for bioavailability enhancement."( Supersaturation Potential of Ordered Mesoporous Silica Delivery Systems. Part 1: Dissolution Performance and Drug Membrane Transport Rates.
Dening, TJ; Taylor, LS, 2018
)
0.48
" Furthermore, the in vitro release and the in vivo pharmacokinetics analyses both showed that RTV SD formulations using Span 20 or HSPC as plasticizer possessed better release profiles and bioavailability over RTV bulk powder but showed equal physicochemical characteristics compared to RTV SD formulations without plasticizer."( Effect of plasticizers on manufacturing ritonavir/copovidone solid dispersions via hot-melt extrusion: Preformulation, physicochemical characterization, and pharmacokinetics in rats.
Cai, C; Ding, Z; Fan, Z; Gao, Y; Han, J; Liu, M; Xie, X; Zhang, H; Zhang, Q; Zhao, Y, 2019
)
0.78
" Pretomanid pharmacokinetic behavior was described by a one-compartment model that at a given dose was linear in its absorption and clearance processes but where the rate of absorption and extent of bioavailability changed with dose."( Population Pharmacokinetics of the Antituberculosis Agent Pretomanid.
Everitt, D; Nedelman, JR; Salinger, DH; Subramoney, V, 2019
)
0.51
"The ATP-binding cassette transporter P-glycoprotein (P-gp) is known to limit both brain penetration and oral bioavailability of many chemotherapy drugs."( A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
Ambudkar, SV; Brimacombe, KR; Chen, L; Gottesman, MM; Guha, R; Hall, MD; Klumpp-Thomas, C; Lee, OW; Lee, TD; Lusvarghi, S; Robey, RW; Shen, M; Tebase, BG, 2019
)
0.51
" The low bioavailability of docetaxel, however, hinders the application of oral docetaxel in the clinic."( A Population Pharmacokinetic Model of Oral Docetaxel Coadministered With Ritonavir to Support Early Clinical Development.
Beijnen, JH; de Weger, VA; Huitema, ADR; Janssen, JM; Nuijen, B; Sawicki, E; Schellens, JHM; van Hasselt, JGC; Yu, H, 2020
)
0.79
" For all 3 drugs, we found a high association with the nanoparticles in plasma (>87% lopinavir-ritonavir, 97% tenofovir), and an incomplete subcutaneous bioavailability (<29% lopinavir-ritonavir, 85% tenofovir)."( Integration of Computational and Experimental Approaches to Elucidate Mechanisms of First-Pass Lymphatic Drug Sequestration and Long-Acting Pharmacokinetics of the Injectable Triple-HIV Drug Combination TLC-ART 101.
Collier, AC; Ho, RJY; Kinman, L; Koehn, J; Lane, S; Lee, W; McConnachie, LA; Perazzolo, S; Shen, DD; Shireman, LM, 2020
)
0.78
" Extensive use of atazanavir (ATV) as antiretroviral and previous evidence suggesting its bioavailability within the respiratory tract prompted us to study this molecule against SARS-CoV-2."( Atazanavir, Alone or in Combination with Ritonavir, Inhibits SARS-CoV-2 Replication and Proinflammatory Cytokine Production.
Alves, CR; Bozza, FA; Bozza, PT; Cardoso Soares, V; Carels, N; da Silva Gomes Dias, S; de Freitas, CS; Ferreira, AC; Fintelman-Rodrigues, N; Matos, AR; Mattos, M; Miranda, MD; Ribeiro Lima, C; Sacramento, CQ; Siqueira, MM; Souza da Silva, F; Souza, TML; Temerozo, JR, 2020
)
0.82
"Human Immunodeficiency Virus (HIV) is a global health concern to which nanomedicine approaches provide opportunities to improve the bioavailability of existing drugs used to treat HIV."( Optimization of the synthetic parameters of lipid polymer hybrid nanoparticles dual loaded with darunavir and ritonavir for the treatment of HIV.
Cauldbeck, H; Elkateb, H; McDonald, T; Niezabitowska, E; Owen, A; Rannard, S; Tatham, LM, 2020
)
0.77
" The present research aims to develop a nanoparticle-based orodispersible pediatric drug delivery platform to improve oral bioavailability and taste of poorly water-soluble and unpalatable therapeutics."( Development of nanoparticle-based orodispersible palatable pediatric formulations.
Deng, Y; Shen, J; Shen, L; Yang, Y, 2021
)
0.62
" None of the tested CYP3A, ABCB1, ABCC2, and SLCO1B3 genotypes and diplotypes showed a significant relation with an altered bioavailability or clearance of either docetaxel or ritonavir."( Investigating the influence of relevant pharmacogenetic variants on the pharmacokinetics and pharmacodynamics of orally administered docetaxel combined with ritonavir.
Beijnen, JH; Dorlo, TPC; Huitema, ADR; Marchetti, S; Pluim, D; van Eijk, M, 2021
)
1.01
"Application of multi-scale modelling workflows to characterise polymorphism in ritonavir with regard to its stability, bioavailability and processing."( Molecular, Solid-State and Surface Structures of the Conformational Polymorphic Forms of Ritonavir in Relation to their Physicochemical Properties.
Docherty, R; Laing, S; Maloney, AGP; Roberts, KJ; Rosbottom, I; Sheikh, AY; Turner, TD; Wang, C; Yin, Q, 2021
)
1.07
" On the contrary, ritonavir co-administration given as pre-treatment significantly enhanced oral bioavailability of both the marketed and milk-based docetaxel formulations; an even more marked increase in drug exposure was observed when ritonavir was incorporated within the docetaxel milk-based formulation."( Enhancement of Docetaxel Absorption Using Ritonavir in an Oral Milk-Based Formulation.
Karampelas, T; Macheras, P; Soulele, K; Tamvakopoulos, C, 2021
)
1.22
" There was a delayed onset of symptoms in many cases, most likely due to the relatively lower systemic bioavailability of intranasal fluticasone."( Prescribing intranasal steroids in HIV-positive patients: systematic review of the literature.
McGilligan, JA; Mohammed, H; Richardson, D; Robinson, M; Seymour, N; Williams, D, 2021
)
0.62
" LPV/r-A tablets contain key excipients critical to ensuring acceptable bioavailability of lopinavir and ritonavir in humans."( Comparative Bioavailability of Two Formulations of Biopharmaceutical Classification System (BCS) Class IV Drugs: A Case Study of Lopinavir/Ritonavir.
Jarvis, MF; Morris, JB; Mostafa, NM; Munasinghe, WP; Saeed, AM; Schmidt, JM; Vallabh, BK, 2021
)
1.04
"Analytical characterizations of LPV/r-B tablets were performed and a clinical study was conducted to assess the relative bioavailability of Kalidavir® (LPV/r-B) 400/100 mg tablets relative to Kaletra® (LPV/r-A) 400/100 mg tablets under fasting conditions."( Comparative Bioavailability of Two Formulations of Biopharmaceutical Classification System (BCS) Class IV Drugs: A Case Study of Lopinavir/Ritonavir.
Jarvis, MF; Morris, JB; Mostafa, NM; Munasinghe, WP; Saeed, AM; Schmidt, JM; Vallabh, BK, 2021
)
0.82
" RTV SD exhibited enhanced dissolution manner, while the oral bioavailability of RTV SD was equivalent with the Reference Standard Norvir® but increased significantly compared to the ternary physical mixture."( Cellulose derivatives as effective recrystallization inhibitor for ternary ritonavir solid dispersions: In vitro-in vivo evaluation.
Guan, Q; Han, J; Jiang, X; Liu, M; Ma, Q; Wang, Z; Zhang, H; Zhao, Y, 2021
)
0.85
"The objective of the present work was to optimize ritonavir (RTV)-loaded nanostructured lipid carriers (NLCs) to improve bioavailability using a quality by design (QbD)-based technique."( Development of ritonavir-loaded nanostructured lipid carriers employing quality by design (QbD) as a tool: characterizations, permeability, and bioavailability studies.
Bari, SB; Gurumukhi, VC, 2022
)
1.33
" Here, we report the discovery and characterization of PF-07321332, an orally bioavailable SARS-CoV-2 main protease inhibitor with in vitro pan-human coronavirus antiviral activity and excellent off-target selectivity and in vivo safety profiles."( An oral SARS-CoV-2 M
Allerton, CMN; Anderson, AS; Aschenbrenner, L; Avery, M; Berritt, S; Boras, B; Cardin, RD; Carlo, A; Coffman, KJ; Dantonio, A; Di, L; Eng, H; Ferre, R; Gajiwala, KS; Gibson, SA; Greasley, SE; Hurst, BL; Kadar, EP; Kalgutkar, AS; Lee, J; Lee, JC; Liu, W; Mason, SW; Noell, S; Novak, JJ; Obach, RS; Ogilvie, K; Owen, DR; Patel, NC; Pettersson, M; Rai, DK; Reese, MR; Sammons, MF; Sathish, JG; Singh, RSP; Steppan, CM; Stewart, AE; Tuttle, JB; Updyke, L; Verhoest, PR; Wei, L; Yang, Q; Zhu, Y, 2021
)
0.62
" Molnupiravir is an orally bioavailable antiviral drug for use at home when a SARS-CoV-2 test is positive."( Editorial: Current Status of Oral Antiviral Drug Treatments for SARS-CoV-2 Infection in Non-Hospitalized Patients.
Parums, DV, 2022
)
0.72
" Additionally, an ∼85% correlation was obtained between PAMPA pH 5 permeability and in vivo oral bioavailability in mice and rats."( Using in vitro ADME data for lead compound selection: An emphasis on PAMPA pH 5 permeability and oral bioavailability.
Itkin, M; Kabir, M; Mathé, EA; Nguyễn, ÐT; Padilha, EC; Shah, P; Shinn, P; Siramshetty, V; Wang, AQ; Williams, J; Xu, X; Yu, KR; Zhao, T, 2022
)
0.72
" The corresponding oral bioavailability was moderate in rats (34%-50%) and low in monkeys (8."( Disposition of Nirmatrelvir, an Orally Bioavailable Inhibitor of SARS-CoV-2 3C-Like Protease, across Animals and Humans.
Boras, B; Dantonio, AL; Di, L; Eng, H; Kadar, EP; Kalgutkar, AS; Kimoto, E; Lin, J; Novak, JJ; Obach, RS; Patel, NC; Singh, RSP; Walker, GS, 2022
)
0.72
" Because of its lower bioavailability and severe side effects, presently, ritonavir is not being used as a PI."( Anti-oxidant Containing Nanostructured Lipid Carriers of Ritonavir: Development, Optimization, and In Vitro and In Vivo Evaluations.
Bhaskaran, NA; Jitta, SR; Kumar, L, 2022
)
1.2
" In HIV+ children on lopinavir/ritonavir, bioavailability was reduced by 32% [median (IQR) steady-state Cmax = 0."( Population pharmacokinetics of ethambutol in African children: a pooled analysis.
Abdelwahab, MT; Andrieux-Meyer, I; Bekker, A; Chabala, C; Cotton, MF; Davies, G; Denti, P; Hesseling, A; Lee, J; McIlleron, H; Rabie, H; Tikiso, T; Wiesner, L; Zar, HJ, 2022
)
1.01
" Oral bioavailability of CsA from liposomal formulations were almost comparable with that from a marketed product (Neoral)."( Liposomal Formulation for Oral Delivery of Cyclosporine A: Usefulness as a Semisolid-Dispersion System.
Asai, T; Kataoka, M; Minami, K; Oku, N; Takagi, T; Yamashita, S, 2022
)
0.72
"Paxlovid is a promising, orally bioavailable novel drug for SARS-CoV-2 with excellent safety profiles."( In Silico Evaluation of Paxlovid's Pharmacometrics for SARS-CoV-2: A Multiscale Approach.
Bartha, FA; Han, R; Juhász, N; Marzban, S; Röst, G, 2022
)
0.72
" The oral bioavailability of paclitaxel is improved through co-administration with ritonavir and application of a suitable pharmaceutical formulation, which addresses the dissolution-limited absorption of paclitaxel."( Development of a population pharmacokinetic/pharmacodynamic model for various oral paclitaxel formulations co-administered with ritonavir and thrombospondin-1 based on data from early phase clinical studies.
Beijnen, JH; de Weger, VA; Dorlo, TPC; Huitema, ADR; Nuijen, B; Sawicki, E; van Eijk, M; Yu, H, 2022
)
1.15
"Controlled inhibition of drug-metabolizing cytochrome P450 3A4 (CYP3A4) is utilized to boost bioavailability of anti-viral and immunosuppressant pharmaceuticals."( Interaction of CYP3A4 with Rationally Designed Ritonavir Analogues: Impact of Steric Constraints Imposed on the Heme-Ligating Group and the End-Pyridine Attachment.
Samuels, ER; Sevrioukova, IF, 2022
)
0.98
"Certain drugs inherently have unfavourable pharmacokinetic properties; for example, they are poorly absorbed or broken down too quickly in the liver."( [CYP3A inhibitors as a pharmacokinetic enhancer: pros and cons of drug interactions].
Burger, DM; Ter Heine, R; van Erp, PH, 2022
)
0.72
" However, several attempts were made by researchers in the past to enhance the oral bioavailability of Ritonavir."( Recent advances in nanoformulation development of Ritonavir, a key protease inhibitor used in the treatment of HIV-AIDS.
Bhaskaran, NA; Jitta, SR; Kumar, L; Marques, SM, 2022
)
1.19
"Most research on nanoformulation development for Ritonavir is mainly focused on enhancing the solubility and oral bioavailability of the drug."( Recent advances in nanoformulation development of Ritonavir, a key protease inhibitor used in the treatment of HIV-AIDS.
Bhaskaran, NA; Jitta, SR; Kumar, L; Marques, SM, 2022
)
1.23
"Ritonavir is the most potent cytochrome P450 (CYP) 3A4 inhibitor in clinical use and is often applied as a booster for drugs with low oral bioavailability due to CYP3A4-mediated biotransformation, as in the treatment of HIV (e."( The Mechanism-Based Inactivation of CYP3A4 by Ritonavir: What Mechanism?
Beijnen, JH; Loos, NHC; Schinkel, AH, 2022
)
2.42
" Nirmatrelvir, an orally bioavailable protease inhibitor that prevents SARS-CoV-2 replication by cleaving the two viral polyproteins, is packaged with ritonavir, a cytochrome P450 (CYP)3A4 inhibitor and pharmacokinetic boosting agent that increases nirmatrelvir concentrations."( Optimizing the use of Paxlovid in clinical practice.
McCarthy, MW, 2022
)
0.92
"Amorphous drugs are used to improve bioavailability of poorly water-soluble drugs."( Impact of Crystal Nuclei on Dissolution of Amorphous Drugs.
Yao, X; Yu, L; Zhang, GGZ, 2023
)
0.91
" The antiviral activity of lopinavir/ritonavir in vivo is mainly derived from lopinavir, while ritonavir improves the bioavailability of lopinavir."( Pharmacokinetics, Safety, and Bioequivalence of 2 Lopinavir/Ritonavir (200/50 mg) Tablets in Healthy Chinese Volunteers: Effect of Food on Absorption.
Bao, D; Fu, W; Hu, W; Lin, L; Liu, Y; Zhang, Q; Zhang, W; Zheng, L, 2023
)
1.43
"VV116 is a chemically-modified version of the antiviral remdesivir with oral bioavailability and potent activity against SARS-CoV-2."( VV116 as a potential treatment for COVID-19.
McCarthy, MW, 2023
)
0.91
"Solid lipid-based formulations (sLBFs) have the potential to increase the oral bioavailability of drugs with poor solubility in water, while counteracting some of the disadvantages of liquid LBFs."( Exploring the use of modified in vitro digestion assays for the evaluation of ritonavir loaded solid lipid-based formulations.
Andreadis, II; Bergström, CAS; Quodbach, J; Schulzen, A, 2023
)
1.14
" Age affected the bioavailability of rifampicin and isoniazid; at birth, children had 48."( Evaluating pediatric tuberculosis dosing guidelines: A model-based individual data pooled analysis.
Aarnoutse, R; Chabala, C; Cotton, MF; Denti, P; Galileya, LT; Gibb, D; Hesseling, A; Lee, J; McIlleron, H; Njahira Mukui, I; Rabie, H; Turkova, A; Wasmann, RE; Zar, H, 2023
)
0.91

Dosage Studied

Combination of indinavir and 100 mg ritonavir in twice daily dosing regimens significantly affects the pharmacokinetic profile. A Bayesian approach is proposed to fit this model to clinical data.

ExcerptRelevanceReference
"During the first 4 weeks, increases in CD4+ lymphocyte counts and reductions in the log number of copies of HIV-1 RNA per milliliter of plasma were similar among the four dosage groups, but in the three lower-dosage groups there was a return to base-line levels by 16 weeks."( A short-term study of the safety, pharmacokinetics, and efficacy of ritonavir, an inhibitor of HIV-1 protease. European-Australian Collaborative Ritonavir Study Group.
Bouza, E; Carr, A; Danner, SA; Gonzales, J; Gudiol, F; Lehman, LM; Leonard, JM; Pintado, V; Raventos, A; Rubio, R, 1995
)
0.53
"18 log in the four dosage groups."( A preliminary study of ritonavir, an inhibitor of HIV-1 protease, to treat HIV-1 infection.
Henry, D; Hsu, A; Hurley, AM; La Marca, A; Leonard, JM; Markowitz, M; Powderly, WG; Saag, M; Sattler, F; Valdes, JM, 1995
)
0.6
" Selecting the initial PI must be individualized, and factors to consider include proven activity, possible toxicities, dosing regimens, drug interactions, and costs."( HIV-1 protease inhibitors. A review for clinicians.
Deeks, SG; Holodniy, M; Kahn, JO; Smith, M, 1997
)
0.3
" After iv or oral dosing in either rats or dogs, > 92% of the dose was recovered in rat and dog feces and < or = 4% was recovered in the urine."( Metabolism and disposition of the HIV-1 protease inhibitor ritonavir (ABT-538) in rats, dogs, and humans.
Buko, AM; Denissen, JF; Grabowski, BA; Johnson, MK; Kempf, DJ; Surber, BW; Thomas, SB, 1997
)
0.54
" Concurrent therapy must be evaluated before treatment, as many agents are either contraindicated or require dosage modification."( Drug interaction potential with inhibitors of HIV protease.
Fisher, EJ; Polk, RE; Van Cleef, GF,
)
0.13
" Furthermore, adverse effects, resistance, dosage and administration, clinical pharmacokinetics, pharmacokinetic-pharmacodynamic relationships, and drug interactions are discussed."( Clinical pharmacology of HIV protease inhibitors: focus on saquinavir, indinavir, and ritonavir.
Beijnen, JH; Burger, DM; Hoetelmans, RM; Koks, CH; Meenhorst, PL; Mulder, JW, 1997
)
0.52
"The effects of fluconazole on the pharmacokinetics of the HIV protease inhibitor ritonavir were investigated after multiple dosing in an open-label study."( Evaluation of the effect of fluconazole on the pharmacokinetics of ritonavir.
Cao, G; Cato, A; Cavanaugh, J; Granneman, R; Hsu, A; Leonard, J, 1997
)
0.76
" Beginning with the moderately potent and orally bioavailable inhibitor A-80987, systematic investigation of peripheral (P3 and P2') heterocyclic groups designed to decrease the rate of hepatic metabolism provided analogues with improved pharmacokinetic properties after oral dosing in rats."( Discovery of ritonavir, a potent inhibitor of HIV protease with high oral bioavailability and clinical efficacy.
Betebenner, D; Fino, L; Flentge, CA; Green, BE; Kati, WM; Kempf, DJ; Marsh, KC; McDonald, E; Molla, A; Norbeck, DW; Patterson, J; Plattner, JJ; Ruiz, L; Saldivar, A; Sham, HL; Vasavanonda, S; Wideburg, NE; Zhao, C, 1998
)
0.67
"The pharmacology, pharmacokinetics, efficacy, adverse effects, drug interactions, and dosage and administration of protease inhibitors are reviewed."( Protease inhibitors for the treatment of human immunodeficiency virus infection.
Kakuda, TN; Piscitelli, SC; Struble, KA, 1998
)
0.3
" Administration of ritonavir with a reduced rifabutin dosage regimen (150 mg every Monday, Wednesday, and Friday) is being investigated."( The effect of multiple doses of ritonavir on the pharmacokinetics of rifabutin.
Cato, A; Cavanaugh, J; Granneman, R; Hsu, A; Leonard, J; Shi, H, 1998
)
0.91
" The lack of change in ritonavir pharmacokinetics suggests that dosage adjustment of ritonavir is unnecessary when it is administered concurrently with ZDV."( Multidose pharmacokinetics of ritonavir and zidovudine in human immunodeficiency virus-infected patients.
Cato, A; Granneman, R; Hsu, A; Leonard, J; Levy, B; Qian, J, 1998
)
0.9
" The relatively minor changes in ritonavir and ddI pharmacokinetics are probably not clinically relevant; therefore, dosage adjustment of either compound appears unnecessary when administered concurrently."( Pharmacokinetic interaction between ritonavir and didanosine when administered concurrently to HIV-infected patients.
Cato, A; Granneman, R; Hsu, A; Leonard, J; Piergies, AA; Qian, J; Vomvouras, S, 1998
)
0.86
") dosing of this combination."( Pharmacokinetic interaction between ritonavir and indinavir in healthy volunteers.
Berg, J; Cao, G; Carothers, L; Dennis, S; El-Shourbagy, T; Erdman, K; Granneman, GR; Hsu, A; Japour, A; Leonard, JM; Sun, E, 1998
)
0.58
" The warfarin dosage was almost doubled in order to maintain a therapeutic INR."( Potential interaction involving warfarin and ritonavir.
Cousins, ES; Knoell, KR; Young, TM, 1998
)
0.56
" Potentiation of warfarin effect and subsequent decrease in the warfarin dosage requirement was anticipated following ritonavir administration; however, the opposite occurred."( Potential interaction involving warfarin and ritonavir.
Cousins, ES; Knoell, KR; Young, TM, 1998
)
0.77
" Due to toxicity risks, co-administration is contraindicated for various interacting drugs, whereas dosage adjustments may only be required for others."( [HIV protease inhibitors: drug interactions].
Dubreuil, L; Gérard, Y; Maulin, L; Mouton, Y, 1999
)
0.3
"To explore the steady-state plasma pharmacokinetics of indinavir in twice daily dosing regimens with and without the co-administration of 100 mg ritonavir."( The steady-state plasma pharmacokinetics of indinavir alone and in combination with a low dose of ritonavir in twice daily dosing regimens in HIV-1-infected individuals.
Beijnen, JH; Hoetelmans, RM; Hsu, A; Lange, JM; Meenhorst, PL; Mulder, JW; Schreij, G; van Heeswijk, RP; Veldkamp, AI, 1999
)
0.72
"Significant differences were observed for indinavir pharmacokinetics between the dosing regimens indinavir 1200 mg twice daily alone and indinavir/ ritonavir 800/100 mg twice daily with respect to the mean trough concentration (0."( The steady-state plasma pharmacokinetics of indinavir alone and in combination with a low dose of ritonavir in twice daily dosing regimens in HIV-1-infected individuals.
Beijnen, JH; Hoetelmans, RM; Hsu, A; Lange, JM; Meenhorst, PL; Mulder, JW; Schreij, G; van Heeswijk, RP; Veldkamp, AI, 1999
)
0.72
"Combination of indinavir and 100 mg ritonavir in twice daily dosing regimens significantly affects the pharmacokinetic profile of indinavir."( The steady-state plasma pharmacokinetics of indinavir alone and in combination with a low dose of ritonavir in twice daily dosing regimens in HIV-1-infected individuals.
Beijnen, JH; Hoetelmans, RM; Hsu, A; Lange, JM; Meenhorst, PL; Mulder, JW; Schreij, G; van Heeswijk, RP; Veldkamp, AI, 1999
)
0.8
" Ritonavir was administered with the following escalation dosing scheme: 300, 400, 500 mg twice a day for 3, 4, and 5 days, respectively, then the full dose of 600 mg twice a day."( The relationship between ritonavir plasma levels and side-effects: implications for therapeutic drug monitoring.
Bassetti, D; Bassetti, M; Casazza, R; Cruciani, M; De Pascalis, C; Di Biagio, A; Gatti, G; Vella, S, 1999
)
1.52
"To investigate the steady-state pharmacokinetics of a once-daily dosing regimen of saquinavir soft gelatin capsules in combination with a low dose of ritonavir in HIV-1-infected individuals."( Once-daily dosing of saquinavir and low-dose ritonavir in HIV-1-infected individuals: a pharmacokinetic pilot study.
Beijnen, JH; Hoetelmans, RM; Lange, JM; Meenhorst, PL; Mulder, JW; van Heeswijk, RP; Veldkamp, AI, 2000
)
0.77
"Steady-state pharmacokinetics of saquinavir and ritonavir were assessed during a 24 h dosing interval after 2 weeks of continued therapy (day 14)."( Once-daily dosing of saquinavir and low-dose ritonavir in HIV-1-infected individuals: a pharmacokinetic pilot study.
Beijnen, JH; Hoetelmans, RM; Lange, JM; Meenhorst, PL; Mulder, JW; van Heeswijk, RP; Veldkamp, AI, 2000
)
0.82
"This pharmacokinetic study indicates that the combination of 1600 mg of saquinavir (soft gelatin capsules) and 200 mg of ritonavir (liquid formulation) in a once-daily dosing regimen generally results in therapeutic plasma concentrations of saquinavir."( Once-daily dosing of saquinavir and low-dose ritonavir in HIV-1-infected individuals: a pharmacokinetic pilot study.
Beijnen, JH; Hoetelmans, RM; Lange, JM; Meenhorst, PL; Mulder, JW; van Heeswijk, RP; Veldkamp, AI, 2000
)
0.77
" After discontinuing RTV and reducing the dosage of CBZ, the serum concentration of CBZ returned to the optimal range, symptoms subsided, and liver function returned to baseline."( Potential interaction between ritonavir and carbamazepine.
Feldman, MD; Fujii, T; Kato, Y; Kayser, SR; Mizoguchi, N; Takata, N; Ueda, K, 2000
)
0.6
"Human immunodeficiency virus type 1 (HIV-1) protease inhibitors have dramatically improved treatment options for HIV infection, but frequent dosing may impact adherence to highly active antiretroviral treatment regimens (HAART)."( Safety and pharmacokinetics of once-daily regimens of soft-gel capsule saquinavir plus minidose ritonavir in human immunodeficiency virus-negative adults.
Buss, N; Ehrensing, E; Gizzi, N; Kilby, JM; Oo, C; Saag, MS; Sfakianos, G; Siemon-Hryczyk, P, 2000
)
0.53
" Trial designs include comparisons between the various licensed protease inhibitors, comparisons of protease inhibitors to other classes of potent antiretroviral drugs, investigations with new protease inhibitors, investigations of protease inhibitor-related toxicities and attempts at simplifying current dosing regimens."( Ongoing trials in HIV protease inhibitors.
Tavel, JA, 2000
)
0.31
" All patients received RTV at a dosage of 400 mg twice daily."( Safety, tolerability, and antiretroviral effects of ritonavir-nelfinavir combination therapy administered for 48 weeks.
Apuzzo, L; Deetz, C; Eshleman, SH; Fields, C; Flexner, C; Gallant, JE; Heath-Chiozzi, M; Jackson, JB; Lewis, RH; Raines, CP; Sun, E, 2000
)
0.56
" At 4-week intervals, plasma levels were measured and dosage of IND/RIT switched to 1000/100 mg daily and then 800/200 mg daily."( Dose-finding study of once-daily indinavir/ritonavir plus zidovudine and lamivudine in HIV-infected patients.
Arnaiz, JA; Blanco, JL; Carné, X; Codina, C; Cruceta, A; García-Viejo, MA; Gatell, JM; Giner, V; Mallolas, J; Martínez, E; Pumarola, T; Sarasa, M; Soriano, A; Soy, D; Tuset, M, 2000
)
0.57
" The objective of our study was to find a once-daily dosing regimen of a HIV-protease inhibitor, indinavir (IDV), by combining it with ritonavir (RTV)."( Dose-finding study of a once-daily indinavir/ritonavir regimen.
Burger, DM; Hekster, YA; Hugen, PW; Koopmans, PP; Lange, JM; Reiss, P; Stek, M; ter Hofstede, HJ, 2000
)
0.77
" However, since protease inhibitors are high clearance drugs, free drug concentration will likely remain unaffected in the presence of elevated AGP during chronic oral dosing although there will be an increase in total plasma drug concentration."( Effect of alpha1-acid glycoprotein on the intracellular accumulation of the HIV protease inhibitors saquinavir, ritonavir and indinavir in vitro.
Back, DJ; Hoggard, PG; Jones, K; Khoo, S; Maher, B, 2001
)
0.52
" Plasma trough levels of nelfinavir (NFV) and saquinavir (SQV), in a twice daily dosing regimen, were above the protein-corrected IC(95) in most patients despite the addition of an enzymatic inducer such as nevirapine, and peak levels were 2- and 5-fold increased with respect to standard doses."( Efficacy, tolerance, and pharmacokinetics of the combination of stavudine, nevirapine, nelfinavir, and saquinavir as salvage regimen after ritonavir or indinavir failure.
Antela, A; Casado, JL; Dehertogh, P; Dronda, F; Hertogs, K; Martí-Belda, P; Moreno, S; Sabido, R, 2001
)
0.51
" Blood samples were collected over the daytime 12-hour dosing interval of the protease inhibitors at baseline (period 1, day 0) and after 10 days of coadministration of 200 mg (n = 6) or 400 mg (n = 6) of ketoconazole once daily (period 2, day 10)."( Effect of ketoconazole on ritonavir and saquinavir concentrations in plasma and cerebrospinal fluid from patients infected with human immunodeficiency virus.
Cameron, DW; Gallicano, K; Khaliq, Y; Kravcik, S; Venance, S, 2000
)
0.61
"Indinavir + ritonavir regimens show improved pharmacokinetic properties, allowing twice-daily dosing with food."( A retrospective, cohort-based survey of patients using twice-daily indinavir + ritonavir combinations: pharmacokinetics, safety, and efficacy.
Aarnoutse, RE; Blok, WL; Burger, DM; Dieleman, JP; Hugen, PW; Lange, JM; Meenhorst, PL; Mulder, JW; Prins, JM; Reiss, P; ten Veen, JH; van der Meer, JT; van der Poll, T, 2001
)
0.92
"Demographics, dosage regimens, genotype data, viral RNA and CD4+ lymphocyte counts, adverse drug events (ADEs), laboratory tests, and compliance were evaluated over 3 years."( The safety and antiviral effect of protease inhibitors in children.
Koranyi, KI; Nahata, MC; Temple, ME, 2001
)
0.31
" In addition, preliminary data suggests the possibility of once daily dosing of ritonavir and saquinavir, which would be expected to increase compliance and allow for direct observed therapy."( Pharmacology and clinical experience with saquinavir.
Kravcik, S, 2001
)
0.54
" When given in combination with indinavir (IDV) it increases the IDV trough concentrations (Cmin) allowing a lower IDV dosage in a twice a day regimen, independently of meals."( [Indinavir-ritonavir combination: pharmacologic results and tolerance in patients infected by HIV].
Bani-Sadr, F; Bernard, L; de Truchis, P; Melchior, JC; Perré, P; Perronne, C; Peytavin, G, 2001
)
0.7
" However, patients must continue taking the drugs once they begin therapy; stopping the treatment or reducing the dosage can lead to resistance."( Freedom of choice.
Vazquez, E,
)
0.13
" Researchers are now attempting to find the proper dosing strategy--one that is strong enough to suppress viral replication and prevent resistance while being mild enough to be tolerated."( Ritonavir plus saquinavir: two trials with different results.
Mascolini, M, 1996
)
1.74
" Several dosage variations were tested."( Saquinavir plus ritonavir reduce viral load by 99.9 percent.
Vazquez, E,
)
0.48
" Dosage regimens are as follows: saquinavir, 3 capsules every 8 hours with food; ritonavir, 6 capsules every 12 hours with food; and indinavir, 2 capsules every 8 hours on an empty stomach."( A patient's guide to protease inhibitors.
Elperin, A; Sax, P, 1996
)
0.52
" Several dosage combinations were studied, and some viral load analyses showed reductions of 99."( Combined protease results continue to hold up.
McGuire, S,
)
0.13
" Situations to avoid include using dosing schedules that do not sufficiently suppress the virus, waiting for viral load to return to pre-treatment levels before switching drugs, and not switching drugs to at least two new potent compounds when changing combination therapy."( Update on antivirals.
, 1997
)
0.3
" Addjusting the methadone dosage may be necessary when ritonavir is used."( Ritonavir (Norvir) and methadone.
, 1998
)
1.99
" The Conference revealed two trends in protease inhibitor use: the increased simultaneous use of two protease inhibitors (PIs) and the switch to twice-daily dosing for some drugs."( Combinations of protease inhibitors.
, 1998
)
0.3
" Delavirdine, the company's FDA-approved NNRTI, has suffered due to the lack of trial data demonstrating a solid anti-HIV effect and due to an inconvenient dosing schedule."( Efavirenz's complications.
, 1998
)
0.3
" Studies suggest that if a woman is taking both antiretroviral drugs and birth control pills she should discuss increasing the birth control dosage and/or using other means of preventing pregnancy with her doctor."( Pregnant pause.
Heidere, C, 1998
)
0.3
" The liquid formulation provides the same dosage as the capsules, but pharmacists may need to call the physician to authorize the change."( Ritonavir capsule manufacturing problems will require switch to liquid formulation.
James, JS, 1998
)
1.74
" The liquid form comes with a precise dosing cup and with instructions to help patients adhere to their dosage requirements."( Open your mouth and close your eyes....
, 1998
)
0.3
" Dosing information is included, along with a hotline number for patients or physicians with questions."( Warning: Norvir (Ritonavir) problems with capsule manufacturing.
,
)
0.47
" On recommendation from Federal guidelines, many treatment regimens approved for adults are being prescribed for children, but these have little information available about dosing and long-term effects."( Antivirals and children.
, 1998
)
0.3
" Protease inhibitors may require strict dosing schedules and food restrictions; most are associated with lipodystrophy (thinning of the arms, legs and face with fat accumulation in the breasts, stomach, and sometimes the upper back)."( What they say about protease inhibitors.
,
)
0.13
" Indinavir and ritonavir plasma concentrations remained above respective inhibitory and effective concentrations (IC95 and EC50) (uncorrected for protein binding) throughout the 24-hour dosing interval for 6 of 10 and 8 of 10 subjects, respectively."( A pilot trial of indinavir, ritonavir, didanosine, and lamivudine in a once-daily four-drug regimen for HIV infection.
Holodniy, M; Mole, L; Schmidgall, D, 2001
)
0.96
"Our pilot study demonstrates excellent virologic suppression despite low minimum protease inhibitor concentrations during a dosing interval in some patients and is supportive of further study."( A pilot trial of indinavir, ritonavir, didanosine, and lamivudine in a once-daily four-drug regimen for HIV infection.
Holodniy, M; Mole, L; Schmidgall, D, 2001
)
0.6
"To compare the steady state plasma pharmacokinetics of 1000 mg of saquinavir (SQV) in a soft-gel capsule (SGC) formulation in combination with 100 mg of ritonavir (RTV) (capsules) in a twice-daily dosing regimen in HIV-1-infected individuals with historical controls who used 400 mg of SQV in a hard-gel capsule (HGC) formulation in combination with 400 mg of RTV and to investigate the plasma pharmacokinetics of the 1000 mg/100 mg regimen after normal and high-fat breakfasts."( Steady-state pharmacokinetics of twice-daily dosing of saquinavir plus ritonavir in HIV-1-infected individuals.
Beijnen, JH; Hoetelmans, RM; Lange, JM; Meenhorst, PL; Mulder, JW; Schreij, G; van der Geest, S; van Heeswijk, RP; Veldkamp, AI, 2001
)
0.74
" The exposure to SQV in the dosing regimen of 1000 mg twice daily in combination with 100 mg of RTV twice daily was significantly higher than the exposure to SQV in a dosing regimen of 400 mg twice daily in combination with 400 mg of RTV twice daily."( Steady-state pharmacokinetics of twice-daily dosing of saquinavir plus ritonavir in HIV-1-infected individuals.
Beijnen, JH; Hoetelmans, RM; Lange, JM; Meenhorst, PL; Mulder, JW; Schreij, G; van der Geest, S; van Heeswijk, RP; Veldkamp, AI, 2001
)
0.54
"Our primary aim was to evaluate the plasma exposures and safety of rifabutin and its active 25-O-desacetyl metabolite during concomitant therapy of intermittent rifabutin dosing regimens with a combination of ritonavir and saquinavir."( A pharmacokinetic study of intermittent rifabutin dosing with a combination of ritonavir and saquinavir in patients infected with human immunodeficiency virus.
Cameron, DW; Carignan, G; Gallicano, K; Khaliq, Y; Tseng, A; Walmsley, S, 2001
)
0.73
" Blood samples were collected over the dosing intervals of the protease inhibitors at baseline (period 1) and of the 3 drugs after 4 weeks (period 2) and 8 weeks (period 3) for HPLC measurement of plasma concentrations of the 3 drugs and 25-O-desacetylrifabutin."( A pharmacokinetic study of intermittent rifabutin dosing with a combination of ritonavir and saquinavir in patients infected with human immunodeficiency virus.
Cameron, DW; Carignan, G; Gallicano, K; Khaliq, Y; Tseng, A; Walmsley, S, 2001
)
0.54
" Intermittent rifabutin dosing over 8 weeks provided a safe and manageable regimen for concurrent therapy with a combination of ritonavir and saquinavir."( A pharmacokinetic study of intermittent rifabutin dosing with a combination of ritonavir and saquinavir in patients infected with human immunodeficiency virus.
Cameron, DW; Carignan, G; Gallicano, K; Khaliq, Y; Tseng, A; Walmsley, S, 2001
)
0.74
" The concentrations at the end of the dosing interval were 10 to 25 times higher than that observed in the standard regimen of 800 mg of IDV q8h for IDV-RTV 800-100 and 800-200 mg regimens, respectively."( Pharmacokinetic profile and tolerability of indinavir-ritonavir combinations in healthy volunteers.
Deutsch, PJ; Nessly, ML; Rhodes, RR; Saah, AJ; Seniuk, MA; Winchell, GA, 2001
)
0.56
"To investigate the influence of combined ritonavir (RTV) and saquinavir (soft-gelatin capsule formulation; SQV) on systemic exposure to SQV with a view to optimizing the dosing regimen of combined RTV and SQV antiretroviral therapy."( Saquinavir and ritonavir pharmacokinetics following combined ritonavir and saquinavir (soft gelatin capsules) administration.
Bock, J; Buss, N; Hsu, A; Jorga, K; Snell, P, 2001
)
0.93
"The purpose of our study was to evaluate the efficacy of indinavir (IDV) in a twice daily dosing regimen with coadministration of 100 mg ritonavir (RTV) and to explore the influence of plasma drug levels in the rate of virologic response."( A clinical study of the combination of 100 mg ritonavir plus 800 mg indinavir as salvage therapy: influence of increased plasma drug levels in the rate of response.
Antela, A; Casado, JL; Dronda, F; Martí-Belda, P; Moreno, A; Moreno, S; Perez-Elías, MJ; Sabido, R,
)
0.59
"RTV/IDV 100/800 mg in a twice daily dosing regimen is associated with a significant virological response in patients with antiretroviral treatment failure."( A clinical study of the combination of 100 mg ritonavir plus 800 mg indinavir as salvage therapy: influence of increased plasma drug levels in the rate of response.
Antela, A; Casado, JL; Dronda, F; Martí-Belda, P; Moreno, A; Moreno, S; Perez-Elías, MJ; Sabido, R,
)
0.39
" Monte Carlo simulation was then used to predict amprenavir concentrations for various combinations of amprenavir and ritonavir in twice-daily and once-daily dosing regimens."( Pharmacokinetic modeling and simulations of interaction of amprenavir and ritonavir.
Sadler, BM; Sale, M; Stein, DS, 2002
)
0.75
" There was no statistically significant difference between the number of patients below an IDV plasma concentration of 150 ng/ml in the various dosage regimens."( Therapeutic drug monitoring of indinavir in HIV-infected patients undergoing HAART.
Desch, S; Klinker, H; Langmann, P; Väth, T; Weissbrich, B; Zilly, M, 2002
)
0.31
" Positive drug-drug interactions increase the exposure to the PIs, allowing administration of lower doses at reduced dosing frequencies with less dietary restrictions."( Combination of protease inhibitors for the treatment of HIV-1-infected patients: a review of pharmacokinetics and clinical experience.
Beijnen, JH; Hoetelmans, RM; Lange, JM; Meenhorst, PL; Mulder, JW; van Heeswijk, RP; Veldkamp, A, 2001
)
0.31
" Among patients with indinavir trough concentration >500 ng/mL, 46 of 49 had a dosage adjustment and 17 have had more than two dosage adjustments."( High indinavir Cmin is associated with higher toxicity in patients on indinavir-ritonavir 800/100 mg twice-daily regimen.
Basso, S; Durand, A; Gallais, H; Gastaut, JA; Lacarelle, B; Poizot-Martin, I; Ravaux, I; Solas, C, 2002
)
0.54
"The complexity of highly active antiretroviral therapy (HAART), with multiple medications, formulations, and dosing intervals, makes adherence challenging."( Reported adherence as a determinant of response to highly active antiretroviral therapy in children who have human immunodeficiency virus infection.
Johnson, GM; Krogstad, PA; Lee, S; Mohan, K; Morse, EV; Nachman, S; Stanley, K; Van Dyke, RB; Wiznia, A, 2002
)
0.31
" On the same day, the patient increased the risperidone dosage to 3 mg twice daily."( Extrapyramidal symptoms with ritonavir/indinavir plus risperidone.
Béïque, LC; Bowmer, MI; Kelly, DV, 2002
)
0.61
" Plasma samples for SQV assay were obtained from 97 healthy subjects following multiple dosing of a range of SQV (400-1800 mg) plus RTV (100-400 mg) dosages for 13-14 days."( The effect of ritonavir on saquinavir plasma concentration is independent of ritonavir dosage: combined analysis of pharmacokinetic data from 97 subjects.
Buss, N; Hill, A; Kilby, JM, 2002
)
0.68
" Higher SQV dosage correlates linearly with higher Cmax (P=0."( The effect of ritonavir on saquinavir plasma concentration is independent of ritonavir dosage: combined analysis of pharmacokinetic data from 97 subjects.
Buss, N; Hill, A; Kilby, JM, 2002
)
0.68
" Based on this concept of 'mini-dose' RTV, once-daily dosing of 1600 mg SQV/100 mg RTV and twice-daily 1000 mg SQV/100 mg RTV are currently being evaluated in clinical trials."( The effect of ritonavir on saquinavir plasma concentration is independent of ritonavir dosage: combined analysis of pharmacokinetic data from 97 subjects.
Buss, N; Hill, A; Kilby, JM, 2002
)
0.68
" The purpose of the meeting was to achieve a consensus among panel members on the following issues: (i) validity of data suggesting the utility of TDM in HAART; (ii) patient categories and clinical settings in which TDM may be of most benefit; (iii) target levels of antiretroviral agents; (iv) influence of covariables on target levels of drugs; (v) blood sampling and dosage adjustment strategies; and (vi) future research steps needed to elucidate issues regarding the applicability of TDM in clinical practice."( Therapeutic drug monitoring in HIV infection: current status and future directions.
Back, D; Fletcher, C; Garaffo, R; Gatti, G; Haubrich, R; Hoetelmans, R; Kurowski, M; Luber, A; Merry, C; Perno, CF, 2002
)
0.31
" In the pharmaceutical domain, the potential polymorphism of the active substances and the excipients could affect Since most of the pharmaceutical products consist of solid dosage forms administered by the oral route, it is not surprising that studies on polymorphism have become a mandatory chapter of any pharmaceutical dossier whether it concerns a new chemical entity or a generic."( [Crystallization and solid state properties of molecules of pharmaceutical interest].
Bauer, M, 2002
)
0.31
"In this randomized, open-label, one-sequence crossover study, nelfinavir 1250 mg twice a day was dosed for 17 days, followed by 14 days of nelfinavir 1250 mg twice a day plus low doses of ritonavir of either 100 mg or 200 mg orally."( Low-dose ritonavir moderately enhances nelfinavir exposure.
Kaeser, B; Kurowski, M; Mrozikiewicz, A; Popescu, M; Sawyer, A, 2002
)
0.92
" Paired plasma and intracellular samples were collected over a full dosing interval from patients (13 on SQV, 6 on RTV, 8 on IDV, 16 on SQV plus RTV, 7 on IDV plus RTV) with a plasma viral load of <400 copies/ml."( Intracellular accumulation of human immunodeficiency virus protease inhibitors.
Back, DJ; Beeching, N; Carey, P; Hoggard, PG; Jones, K; Khoo, SH; Lloyd, J; Meaden, ER; Newton, P; Peters, B; Smith, A; Tjia, JF; Wilkins, EG; Williams, I, 2002
)
0.31
" Discontinuation of atenolol, and nifedipine dosage reduction by 50% were effective in managing his orthostatic changes."( Symptomatic orthostasis with extended-release nifedipine and protease inhibitors.
Rathbun, RC; Rossi, DR; Slater, LN, 2002
)
0.31
" N given simultaneously with S decreases S hepatic intrinsic clearance 10-fold relative to that predicted for S given alone; and 3) R inhibits S hepatic intrinsic clearance even after R plasma levels have become undetectable (>48 h after dosing), implying that R, when used as a pharmacokinetic enhancer, can be dosed less frequently than might be predicted from the duration of detectable systemic concentrations."( Model-based analysis of the pharmacokinetic interactions between ritonavir, nelfinavir, and saquinavir after simultaneous and staggered oral administration.
Blaschke, TF; Flexner, C; Lu, JF; Rosenkranz, SL; Sheiner, LB, 2002
)
0.55
" The use of rit-boosted PI allows the reduction of pill burden, improves dosing schedules and enhances drug exposure, all factors that have been associated with a greater benefit of antiretroviral therapy."( Comparison of the efficacy, safety and predictive value of HIV genotyping using distinct ritonavir-boosted protease inhibitors.
Barreiro, P; Camino, N; de Mendoza, C; González-Lahoz, J; Martín-Carbonero, L; Núñez, M; Soriano, V; Valer, L, 2002
)
0.54
"The protease inhibitor (PI) ritonavir is used as a strong inhibitor of cytochrome P450 3A4, which boosts the activities of coadministered PIs, resulting in augmented plasma PI levels, simplification of the dosage regimen, and better efficacy against resistant viruses."( Steady-state pharmacokinetics of amprenavir coadministered with ritonavir in human immunodeficiency virus type 1-infected patients.
Bidault, R; Bollens, D; Choudet, N; Demarles, D; Gillotin, C; Goujard, C; Meynard, JL; Rousseau, C; Taburet, AM; Vincent, I, 2003
)
0.85
"The steady-state pharmacokinetics and pharmacodynamics of two oral doses of lopinavir-ritonavir (lopinavir/r; 400/100 and 533/133 mg) twice daily (BID) when dosed in combination with efavirenz, plus two nucleoside reverse transcriptase inhibitors, were assessed in a phase II, open-label, randomized, parallel arm study in 57 multiple protease inhibitor-experienced but non-nucleoside reverse transcriptase inhibitor-naive human immunodeficiency virus (HIV)-infected subjects."( Pharmacokinetic-pharmacodynamic analysis of lopinavir-ritonavir in combination with efavirenz and two nucleoside reverse transcriptase inhibitors in extensively pretreated human immunodeficiency virus-infected patients.
Bernstein, B; Bertz, R; Brun, S; Foit, C; Granneman, GR; Hsu, A; Isaacson, J; Kempf, DJ; King, M; Lam, W; Richards, B; Rode, R; Rynkiewicz, K; Sun, E, 2003
)
0.79
" Including GF120918 in a multiple (twice daily) dosing regimen, we found continued accumulation of saquinavir in brain over several days, resulting in 10-fold higher levels compared with vehicle-treated mice."( Assessing safety and efficacy of directed P-glycoprotein inhibition to improve the pharmacokinetic properties of saquinavir coadministered with ritonavir.
Beijnen, JH; Huisman, MT; Schinkel, AH; Smit, JW; Wiltshire, HR, 2003
)
0.52
"To evaluate the effect of acute dosing of garlic supplements on the single-dose pharmacokinetics of ritonavir."( Effect of short-term administration of garlic supplements on single-dose ritonavir pharmacokinetics in healthy volunteers.
Choudhri, S; Foster, B; Gallicano, K, 2003
)
0.77
"Acute dosing of the garlic capsules over 4 days did not significantly alter the single-dose pharmacokinetics of ritonavir in healthy volunteers."( Effect of short-term administration of garlic supplements on single-dose ritonavir pharmacokinetics in healthy volunteers.
Choudhri, S; Foster, B; Gallicano, K, 2003
)
0.76
" In 1998, a lower energy, more stable polymorph (form II) appeared, causing slowed dissolution of the marketed dosage form and compromising the oral bioavailability of the drug."( Elucidation of crystal form diversity of the HIV protease inhibitor ritonavir by high-throughput crystallization.
Almarsson, O; Cima, MJ; Levinson, D; Morissette, SL; Soukasene, S, 2003
)
0.55
" The pharmacokinetics of lopinavir did not appear to be dependent on age when dosing was based on body surface area but were decreased on coadministration with nevirapine."( Forty-eight-week evaluation of lopinavir/ritonavir, a new protease inhibitor, in human immunodeficiency virus-infected children.
Allen, U; Arpadi, S; Bernstein, B; Bertz, RJ; Cahn, P; Castrejón, MM; Chadwick, E; Deetz, CO; Gomez, P; Handelsman, E; Heuser, RS; Hsu, AF; Kempf, DJ; Pelton, S; Ramilo, O; Renz, CL; Rode, RA; Sáez-Llorens, X; Sun, E; Violari, A, 2003
)
0.58
" A number of clinically important drug interactions have been reported with lopinavir/ritonavir necessitating dosage adjustments of lopinavir/ritonavir and/or the interacting drugs, and several other drugs are contraindicated in patients receiving the coformulation."( Lopinavir/ritonavir: a review of its use in the management of HIV infection.
Cvetkovic, RS; Goa, KL, 2003
)
0.94
" However, a recent study in healthy volunteers suggests that adequate lopinavir concentrations may be achieved during rifampicin coadministration by increasing the twice-daily dosage of lopinavir/ritonavir in conjunction with therapeutic drug monitoring."( Lopinavir/ritonavir: a review of its use in the management of HIV infection.
Cvetkovic, RS; Goa, KL, 2003
)
0.91
"To evaluate the pharmacokinetic effect of adding delavirdine mesylate to the antiretroviral regimens of human immunodeficiency virus (HIV)-infected patients stabilized on a full dosage of ritonavir (600 mg every 12 h), 12 HIV-1-infected subjects had delavirdine mesylate (400 mg every 8 h) added to their current antiretroviral regimens for 21 days."( Pharmacokinetics of ritonavir and delavirdine in human immunodeficiency virus-infected patients.
Adams, J; Cox, S; Della-Coletta, A; Hewitt, RG; Morse, GD; Shelton, MJ, 2003
)
0.83
" The persistent effect of ritonavir suggests the possibility that lower doses and longer dosing intervals might be effective when ritonavir is used to boost concentrations of other protease inhibitors."( Effect of simultaneous versus staggered dosing on pharmacokinetic interactions of protease inhibitors.
Aberg, JA; Blaschke, TF; Flexner, C; Rosenkranz, SL; Segal, Y; Sheiner, LB; Washington, CB, 2003
)
0.62
" Crystallization processes were studied for indinavir sulfate, indinavir free base and a commercial indinavir capsule dosage form, respectively."( In-vitro crystallization of indinavir in the presence of ritonavir and as a function of pH.
Fleisher, D; Heimbach, T; Li, LY; Rodríguez-Hornedo, N, 2003
)
0.56
"This study examines the pharmacokinetic/pharmacodynamic interactions between (1) lopinavir-ritonavir (L/R), a fixed combination of protease inhibitors used for the treatment of HIV disease, and (2) ritonavir alone at the same dosage as that in the L/R formulation, with methadone, an opiate frequently used in substance abuse pharmacotherapy for opioid (heroin)-dependent injection drug users, many of whom are infected with HIV."( The protease inhibitor lopinavir-ritonavir may produce opiate withdrawal in methadone-maintained patients.
Friedland, G; Jatlow, P; McCance-Katz, EF; Rainey, PM, 2003
)
0.82
" Great caution is required in the management of tacrolimus dosage when Kaletra is introduced or withdrawn in HIV-positive patients after liver transplantation, particularly in the presence of hepatic dysfunction."( Effect of coadministered lopinavir and ritonavir (Kaletra) on tacrolimus blood concentration in liver transplantation patients.
Eghtesad, B; Fung, JJ; Jain, AB; Marcos, A; Rafail, AB; Ragni, M; Shapiro, R; Venkataramanan, R, 2003
)
0.59
" For efavirenz, the concentrations at 12 hours and 24 hours (Cmin) after dosing were assessed."( Pharmacokinetics of indinavir/ritonavir (800/100 mg) in combination with efavirenz (600 mg) in HIV-1-infected subjects.
Aarnoutse, RE; Boyd, MA; Burger, DM; Cooper, DA; Lange, JM; Phanuphak, P; Ruxrungtham, K; Stek, M; van Heeswijk, RP, 2003
)
0.61
" Maximal viral suppression (in vitro) was achieved when amprenavir free-drug concentrations remained greater than four times the 50% effective concentration (EC(50)) for 80% of the dosing interval."( In vitro-in vivo model for evaluating the antiviral activity of amprenavir in combination with ritonavir administered at 600 and 100 milligrams, respectively, every 12 hours.
Bilello, JA; Drusano, GL; Piliero, PJ; Preston, SL; Stein, DS; Symonds, WT, 2003
)
0.54
" In the latter study, drug-drug interactions, dosing errors, noncompliance and other important problems were identified and corrected."( Clinical use of a simultaneous HPLC assay for indinavir, saquinavir, ritonavir and nelfinavir in children and adults.
Brady, M; Cox, S; Crim, L; Gerber, N; Koranyi, K; Utkin, I; Walson, PD, 2003
)
0.55
" The aim of the study was to measure unbound plasma concentrations of lopinavir (LPV) and to relate them to the total plasma concentrations to establish the unbound percentage in vivo during a full dosage interval."( Lopinavir protein binding in vivo through the 12-hour dosing interval.
Back, DJ; Boffito, M; Bonora, S; Di Perri, G; Hoggard, PG; Khoo, SH; Lindup, WE; Sinicco, A, 2004
)
0.32
"The quality and performance of a solid oral dosage form depends on the choice of the solid phase, the formulation design, and the manufacturing process."( Phase transformation considerations during process development and manufacture of solid oral dosage forms.
Law, D; Qiu, Y; Schmitt, EA; Zhang, GG, 2004
)
0.32
" As this SQVOD-based regimen had considerable short-term virologic activity in treatment-experienced HIV-infected patients, it may be a reasonable option when non-nucleoside reverse transcriptase inhibitors cannot be administered and once-daily dosing is preferred by the patient."( Once-daily saquinavir and ritonavir in treatment-experienced HIV-1-infected individuals.
Cernuschi, M; Gianotti, N; Lazzarin, A; Soria, A, 2004
)
0.62
" For R-methadone (active isomer), C(max), AUC(0-24 h), and C(min) were unchanged, but percent unbound 4 hours after dosing was reduced by 12%."( The effects of once-daily saquinavir/minidose ritonavir on the pharmacokinetics of methadone.
Berenson, CS; Buggé, CJ; Cloen, D; de Caprariis, PJ; DiFrancesco, R; Esch, A; Espinosa, O; Hewitt, RG; Ljungqvist, A; Palic, B; Schur, JL; Shelton, MJ, 2004
)
0.58
" A retrospective study carried out of 72 probable SARS patients has shown that cases who received pulse methylprendisolone did not differ in cumulative steroid dosage or adverse reactions, although the former patients had less oxygen requirement, better radiographic outcome, and less likelihood of requiring rescue pulse steroid therapy than their counterparts."( SARS: pharmacotherapy.
Tsang, K; Zhong, NS, 2003
)
0.32
"When EFV is coadministered with the GW433908 700 mg + RTV 100 mg BID regimen, no dosage adjustment is recommended."( Pharmacokinetics and safety of GW433908 and ritonavir, with and without efavirenz, in healthy volunteers.
Ballow, C; Hendrix, CW; Lou, Y; Piliero, PJ; Preston, SL; Stein, DS; Wire, MB, 2004
)
0.58
" After 48 weeks, LPV/r was dosed open-label at 400/100 mg every 12 hours with stavudine and lamivudine."( Long-term safety and durable antiretroviral activity of lopinavir/ritonavir in treatment-naive patients: 4 year follow-up study.
Benson, C; Brun, SC; Eron, JJ; Gulick, RM; Hicks, C; Kessler, HA; King, KR; King, MS; Murphy, RL; White, AC, 2004
)
0.56
" The dosage of itraconazole was reduced when it was used in combination with lopinavir/ritonavir."( Drug-drug interaction between itraconazole and the antiretroviral drug lopinavir/ritonavir in an HIV-1-infected patient with disseminated histoplasmosis.
Beijnen, JH; Crommentuyn, KM; Huitema, AD; Mulder, JW; Schellens, JH; Sparidans, RW, 2004
)
0.77
" Each dosage was taken twice daily for 2 weeks before 12 h pharmacokinetics were obtained."( Comparison of two reduced-dose regimens of indinavir (600 mg vs 400 mg twice daily) and ritonavir (100 mg twice daily) in healthy volunteers (COREDIR).
Burger, DM; la Porte, CJ; Rockstroh, JK; Schneider, K; Wasmuth, JC, 2004
)
0.55
" Thus, no dosage adjustments are warranted when enfuvirtide is coadministered with low-dose ritonavir or saquinavir boosted with a low dose of ritonavir."( Lack of interaction between enfuvirtide and ritonavir or ritonavir-boosted saquinavir in HIV-1-infected patients.
Bellibas, SE; Boyd, M; Buss, N; Dorr, A; Kinchelow, T; Kolis, S; Patel, IH; Ruxrungtham, K; Zhang, X, 2004
)
0.8
" Reducing the pill burden with once-daily dosing may improve adherence."( Intracellular and plasma pharmacokinetics of saquinavir-ritonavir, administered at 1,600/100 milligrams once daily in human immunodeficiency virus-infected patients.
Back, D; Boffito, M; Ford, J; Gazzard, B; Hill, A; Khoo, S; Moyle, G; Nelson, M; Pozniak, A; Wildfire, A, 2004
)
0.57
" Subsequent PI dosage adjustments were based on real-time pharmacokinetic assessment."( The pharmacokinetics, safety, and initial virologic response of a triple-protease inhibitor salvage regimen containing amprenavir, saquinavir, and ritonavir.
Corbett, AH; Eron, JJ; Fiscus, SA; Kashuba, AD; Rezk, NL, 2004
)
0.52
" After studying 12-hour pharmacokinetic profiles in 3 HIV-positive patients after liver transplantation, we describe how dosing of cyclosporine A can be adjusted to maintain effective immunosuppressive drug levels on a daily dosing schedule when ritonavir-boosted indinavir or lopinavir-based antiretroviral therapy is given."( Management of drug-to-drug interactions between cyclosporine A and the protease-inhibitor lopinavir/ritonavir in liver-transplanted HIV-infected patients.
Michaelis, HC; Rockstroh, JK; Sauerbruch, T; Spengler, U; Sudhop, T; Türler, A; Vogel, M; Voigt, E; Wolff, M, 2004
)
0.72
" Management should be individualized to each patient; dosage or medication adjustments may be necessary."( Coadministration of lopinavir/ritonavir and phenytoin results in two-way drug interaction through cytochrome P-450 induction.
Bertz, RJ; Eron, JJ; Gaedigk, A; Kashuba, AD; Lim, ML; Min, SS; Robinson, M, 2004
)
0.61
"A pharmacokinetic comparison of three dosing regimens of saquinavir/ritonavir was carried out: 1600/100 mg once-daily with 1000/100 mg twice-daily, and 1600/100 mg once-daily with 2000/100 mg once-daily."( Pharmacokinetic study of saquinavir hard gel caps/ritonavir in HIV-1-infected patients: 1600/100 mg once-daily compared with 2000/100 mg once-daily and 1000/100 mg twice-daily.
Ananworanich, J; Apateerapong, W; Autar, RS; Burger, D; Cooper, D; Hill, A; Hirschel, B; Lange, J; Phanuphak, P; Ruxrungtham, K; Sankote, J, 2004
)
0.81
"Compared with saquinavir/ritonavir 1600/100 mg once-daily dosing, the saquinavir AUC and Cmin improved significantly when dosed as 1000/100 mg twice-daily (53% and 299%, respectively), and as 2000/100 mg once-daily (71% and 65%, respectively)."( Pharmacokinetic study of saquinavir hard gel caps/ritonavir in HIV-1-infected patients: 1600/100 mg once-daily compared with 2000/100 mg once-daily and 1000/100 mg twice-daily.
Ananworanich, J; Apateerapong, W; Autar, RS; Burger, D; Cooper, D; Hill, A; Hirschel, B; Lange, J; Phanuphak, P; Ruxrungtham, K; Sankote, J, 2004
)
0.88
"Saquinavir/ritonavir when dosed as 2000/100 mg once-daily or 1000/100 mg twice-daily achieves higher saquinavir plasma levels compared with saquinavir/ritonavir 1600/100 mg once-daily."( Pharmacokinetic study of saquinavir hard gel caps/ritonavir in HIV-1-infected patients: 1600/100 mg once-daily compared with 2000/100 mg once-daily and 1000/100 mg twice-daily.
Ananworanich, J; Apateerapong, W; Autar, RS; Burger, D; Cooper, D; Hill, A; Hirschel, B; Lange, J; Phanuphak, P; Ruxrungtham, K; Sankote, J, 2004
)
0.97
"This is the largest formal pharmacokinetic evaluation of 2 dosage combinations of IDV/RTV in HIV-infected individuals."( Comparison of two indinavir/ritonavir regimens in the treatment of HIV-infected individuals.
Acosta, EP; Eron, JJ; Ferguson, E; Fichtenbaum, CJ; Gerber, JG; Hammer, SM; Kuritzkes, DR; Neath, D; Pettinelli, C; Saah, AJ; Walawander, A; Wu, H; Yu, S, 2004
)
0.62
" The tissue distribution of radioactivity was examined in rats dosed with [14C]lopinavir in combination with ritonavir."( Metabolism and disposition of the HIV-1 protease inhibitor lopinavir (ABT-378) given in combination with ritonavir in rats, dogs, and humans.
Denissen, JF; Grabowski, BA; Jayanti, VK; Johnson, MK; Kempf, DJ; Kumar, GN; Lee, RD; Marsh, KC; Roberts, SA; Sham, HL; Sun, E; Thomas, S; Uchic, J, 2004
)
0.75
" On oral dosing to rats, ritonavir was found to increase the exposure of lopinavir-derived radioactivity 13-fold."( Metabolism and disposition of the HIV-1 protease inhibitor lopinavir (ABT-378) given in combination with ritonavir in rats, dogs, and humans.
Denissen, JF; Grabowski, BA; Jayanti, VK; Johnson, MK; Kempf, DJ; Kumar, GN; Lee, RD; Marsh, KC; Roberts, SA; Sham, HL; Sun, E; Thomas, S; Uchic, J, 2004
)
0.84
" Therefore, we studied the plasma and intracellular (cell-associated) steady-state pharmacokinetics of this PI combination in a dosage of 400/100 mg twice daily in a non-randomized cohort of HIV-1-infected individuals."( The plasma and intracellular steady-state pharmacokinetics of lopinavir/ritonavir in HIV-1-infected patients.
Beijnen, JH; Crommentuyn, KM; Huitema, AD; Mairuhu, AT; Meenhorst, PL; Mulder, JW; van Gorp, EC, 2004
)
0.56
" Blood samples were collected up to 24 h after dosing on days 10 (period 1) and 20 (period 2)."( Effect of efavirenz treatment on the pharmacokinetics of nelfinavir boosted by ritonavir in healthy volunteers.
Burger, DM; Colbers, EP; de Graaff-Teulen, MJ; Hekster, YA; Ibanez, SM; Koopmans, PP; la Porte, CJ; Voncken, DS, 2004
)
0.55
"In antiretroviral-experienced subjects with sustained viral suppression, dual therapy with NVP plus LPV/rtv at standard dosage was as potent and safe as standard-of-care HAART at 48 weeks of follow-up."( Lopinavir/ritonavir plus nevirapine as a nucleoside-sparing approach in antiretroviral-experienced patients (NEKA study).
Burger, D; Clotet, B; Côté, H; López, S; Martínez, E; Miró, O; Moltó, J; Montaner, J; Negredo, E; Puig, J; Rey-Joly, C; Ribalta, J; Ruiz, L; Salazar, J, 2005
)
0.73
" In this study, dosing didanosine enteric-coated 400 mg once daily + indinavir/ritonavir 1200/400 mg once daily with breakfast indicated no decrease in the amount of absorption for either didanosine and indinavir and that this regimen could be administered with food."( Pharmacokinetic interaction study of indinavir/ritonavir and the enteric-coated capsule formulation of didanosine in healthy volunteers.
Aarnoutse, R; Burger, D; Hekster, Y; Koopmans, P; la Porte, C; Reiss, P; Stek, M; van Ewijk, N; Verweij-van Wissen, C, 2005
)
0.81
" Switching from 800/100 to 400/100 mg dosage improved adverse events in 16 of 20 patients."( Indinavir/ritonavir-based therapy in HIV-1-infected antiretroviral therapy-naive patients: comparison of 800/100 mg and 400/100 mg twice daily.
Clumeck, N; Crommentuyn, K; De Wit, S; Huitema, A; Konopnicki, D; Poll, B, 2005
)
0.73
" Reduced ritonavir dosing may help to reduce ritonavir-related side effects and costs."( Boosted saquinavir hard gel formulation exposure in HIV-infected subjects: ritonavir 100 mg once daily versus twice daily.
Back, D; Boffito, M; Dickinson, L; Fletcher, C; Gazzard, B; Hill, A; Maitland, D; Moyle, G; Nelson, M; Pozniak, A, 2005
)
0.98
" This pharmacological effect, even at low doses (( Iatrogenic Cushing's syndrome with osteoporosis and secondary adrenal failure in human immunodeficiency virus-infected patients receiving inhaled corticosteroids and ritonavir-boosted protease inhibitors: six cases.
Cooper, DA; Gowers, A; McMurchie, M; Pett, S; Samaras, K, 2005
)
0.52
" The median (interquartile range) LPV AUC(0,24 h), maximum plasma concentration (C(max)) and concentration at the end of the dosing interval (C(24 h)) after am and pm dosing was, respectively, 143 (116-214) mg l(-1) h, 12."( Absence of circadian variation in the pharmacokinetics of lopinavir/ritonavir given as a once daily dosing regimen in HIV-1-infected patients.
Bourbeau, M; Cameron, DW; Garber, GE; Giguere, P; Seguin, I; van Heeswijk, RP, 2005
)
0.56
"No differences were observed in the pharmacokinetics of LPV/r after am or pm dosing with food, which suggests that this once daily combination, can be taken in the morning or evening."( Absence of circadian variation in the pharmacokinetics of lopinavir/ritonavir given as a once daily dosing regimen in HIV-1-infected patients.
Bourbeau, M; Cameron, DW; Garber, GE; Giguere, P; Seguin, I; van Heeswijk, RP, 2005
)
0.56
" Certain pharmacologic features of many PIs, such as their limited oral bioavailability, necessitate burdensome dosage schedules, creating a barrier to patient adherence."( Simplifying the treatment of HIV infection with ritonavir-boosted protease inhibitors in antiretroviral-experienced patients.
Scott, JD, 2005
)
0.58
" Indinavir dosing was then reduced to 400 mg (twice daily) and 1 week later an identical series of samples were drawn."( Low-doses of indinavir boosted with ritonavir in HIV-infected Thai patients: pharmacokinetics, efficacy and tolerability.
Cressey, TR; Jourdain, G; Kunkeaw, S; Lallemant, M; Leenasirimakul, P; Puttimit, C; Sukrakanchana, PO; Tod, M, 2005
)
0.6
"CTZ does not significantly affect the pharmacokinetic parameters of RTV, and the association does not, thus, require a modification of the dosage of the protease inhibitor."( Evaluation of pharmacokinetic interaction between cetirizine and ritonavir, an HIV-1 protease inhibitor, in healthy male volunteers.
Cremieux, AC; Delatour, F; Farinotti, R; Gautran, C; Melac, M; Moulaert, B; Otoul, C; Peytavin, G; Strolin-Benedetti, M, 2005
)
0.57
"As tolerance not only depends on protease inhibitors (PI) dosing but also on their trough concentrations, the enhanced exposure associated with PI boosting goes together with a reduced tolerance, especially at the biological level."( [Therapeutic strategies for HIV infection: tolerance to boost antiproteases].
Viard, JP, 2004
)
0.32
" However, many of the available agents in this class suffer shortcomings, including poor tolerability, difficult dosing regimens, and variable drug concentrations which may lead to generation of viral resistance."( Safety and antiviral activity of lopinavir/ritonavir-based therapy in human immunodeficiency virus type 1 (HIV-1) infection.
Hicks, CB; Kaplan, SS, 2005
)
0.59
" It binds strongly and selectively, has a favourable resistance profile, and is administered orally twice daily with a subtherapeutic dosage of ritonavir in a 'boosted' regimen (TPV/r) in order to increase its bioavailability."( Tipranavir: a ritonavir-boosted protease inhibitor.
Croom, KF; Keam, SJ, 2005
)
0.89
" However, many available agents suffer shortcomings that limit their clinical value, including adverse effects, difficult dosing requirements and rapid development of resistance."( Lopinavir/ritonavir in the treatment of human immunodeficiency virus infection.
Hicks, CB; Kaplan, SS, 2005
)
0.73
"Few data are available regarding clinical outcomes or dosing requirements for the protease inhibitor ritonavir in human immunodeficiency virus (HIV)-infected children younger than under 24 months of age."( Ritonavir-based highly active antiretroviral therapy in human immunodeficiency virus type 1-infected infants younger than 24 months of age.
Abrams, EJ; Borkowsky, W; Britto, P; Chadwick, EG; Flynn, PM; Hughes, M; Luzuriaga, K; Palumbo, P; Powell, C; Rodman, JH; Yogev, R, 2005
)
1.99
" Two sequential dosing cohorts were treated with 350 or 450 mg/m(2) ritonavir every 12 hours; this report includes results of pharmacokinetics, safety, tolerability and efficacy through 104 weeks of follow-up of all subjects."( Ritonavir-based highly active antiretroviral therapy in human immunodeficiency virus type 1-infected infants younger than 24 months of age.
Abrams, EJ; Borkowsky, W; Britto, P; Chadwick, EG; Flynn, PM; Hughes, M; Luzuriaga, K; Palumbo, P; Powell, C; Rodman, JH; Yogev, R, 2005
)
2.01
" When atazanavir 300 mg was coadministered with ritonavir 100 mg on a once-daily dosage regimen, atazanavir AUC from 0 to 24 hours and minimum plasma concentration were increased by 3- to 4-fold and approximately 10-fold, respectively, compared with atazanavir 300 mg alone."( Clinical pharmacokinetics and summary of efficacy and tolerability of atazanavir.
Barrail, A; Goujard, C; Le Tiec, C; Taburet, AM, 2005
)
0.58
"Saquinavir/ritonavir data, dosed as 1600/100 mg once daily, from three separate pharmacokinetic studies, in 45 patients from Thailand and the UK, were pooled."( Interindividual variability of once-daily ritonavir boosted saquinavir pharmacokinetics in Thai and UK patients.
Ananworanich, J; Autar, RS; Boffito, M; Burger, DM; Cooper, DA; Hassink, E; Lange, JM; Phanuphak, P; Pozniak, A; Ruxrungtham, K; Siangphoe, U; Wit, FW, 2005
)
0.98
" The dosage was 50 mg/kg twice daily (bid) for saquinavir and 230/57."( Pharmacokinetics and 24-week efficacy/safety of dual boosted saquinavir/lopinavir/ritonavir in nucleoside-pretreated children.
Ananworanich, J; Bergshoeff, A; Burger, D; Engchanil, C; Hill, A; Kosalaraksa, P; Pancharoen, C; Ruxrungtham, K; Siangphoe, U, 2005
)
0.55
"Ritonavir dosed at 100 mg bid significantly increased the concentration of total cholesterol, LDL cholesterol, total/HDL cholesterol ratio and triglycerides and reduced HDL cholesterol concentration."( The effect of low-dose ritonavir monotherapy on fasting serum lipid concentrations.
Mashinter, LD; Roberts, SE; Shafran, SD, 2005
)
2.08
"To improve the dosing frequency and pill burden of antiretroviral therapy, we compared two once-daily dosed regimens to a twice-daily dosed regimen."( Comparison of two once-daily regimens with a regimen consisting of nelfinavir, didanosine, and stavudine in antiretroviral therapy-naïve adults: 48-week results from the Antiretroviral Regimen Evaluation Study (ARES).
Borleffs, JC; Hassink, EA; Juttmann, JR; Koopmans, PP; Lange, JM; Lowe, SH; Richter, C; Schreij, G; ten Kate, RW; van der Tweel, I; Wensing, AM,
)
0.13
"HIV-1-infected, antiretroviral drug-naïve adults were randomized to either twice-daily nelfinavir and stavudine and once-daily didanosine (regimen A) or simplified once-daily dosed antiretroviral regimens consisting of nevirapine, didanosine, and lamivudine (regimen B) or saquinavir, ritonavir, didanosine, and lamivudine (regimen C)."( Comparison of two once-daily regimens with a regimen consisting of nelfinavir, didanosine, and stavudine in antiretroviral therapy-naïve adults: 48-week results from the Antiretroviral Regimen Evaluation Study (ARES).
Borleffs, JC; Hassink, EA; Juttmann, JR; Koopmans, PP; Lange, JM; Lowe, SH; Richter, C; Schreij, G; ten Kate, RW; van der Tweel, I; Wensing, AM,
)
0.31
"No statistically significant difference in efficacy was found between the two investigated once-daily dosed treatment regimens (B and C) and the reference (A)."( Comparison of two once-daily regimens with a regimen consisting of nelfinavir, didanosine, and stavudine in antiretroviral therapy-naïve adults: 48-week results from the Antiretroviral Regimen Evaluation Study (ARES).
Borleffs, JC; Hassink, EA; Juttmann, JR; Koopmans, PP; Lange, JM; Lowe, SH; Richter, C; Schreij, G; ten Kate, RW; van der Tweel, I; Wensing, AM,
)
0.13
" No significant relationship was established between the presence or the daily dosage of saquinavir in the treatment and lopinavir population pharmacokinetic parameters."( No significant influence of saquinavir hard-gel capsule administration on pharmacokinetics of lopinavir in combination with ritonavir: a population approach.
Allavena, C; Dailly, E; Gagnieu, MC; Jolliet, P; Raffi, F, 2005
)
0.54
"There is a need for new, clinically relevant interpretation algorithms for genotypic and phenotypic resistance for ritonavir-boosted saquinavir (SQV/r) at the current approved dosage [1000/100 mg twice a day (bid)]."( Analysis of genotypic and phenotypic clinical cut-off levels for ritonavir-boosted saquinavir.
Clotet, B; Hill, A; Molto, J; Walmsley, S, 2006
)
0.78
" For FPV 1,400 mg BID plus RTV 100 mg BID, the values for plasma amprenavir (APV) area under the concentration-time profile over the dosing interval (tau) at steady state [AUC(0-tau)], maximum concentration of drug in plasma (C(max)), and plasma concentration at the end of tau at steady state (C(tau)) were 54, 81, and 26% higher, respectively, and the values for plasma RTV AUC(0-tau), C(max), and C(tau) were 49% higher, 71% higher, and 11% lower, respectively, than those for FPV 700 mg BID plus RTV 100 mg BID."( Pharmacokinetic and safety evaluation of high-dose combinations of fosamprenavir and ritonavir.
Adamkiewicz, B; Lou, Y; Min, SS; Shelton, MJ; Wire, MB, 2006
)
0.56
" The Ccell/Ctot and Cu/Ctot ratio was unaffected by the addition of the second PI and remained stable throughout dosing interval."( Ritonavir-boosted atazanavir-lopinavir combination: a pharmacokinetic interaction study of total, unbound plasma and cellular exposures.
Biollaz, J; Buclin, T; Cavassini, M; Colombo, S; Decosterd, LA; Franc, C; Guignard, N; Khonkarly, M; Rochat, B; Tarr, PE; Telenti, A, 2006
)
1.78
" The clinical significance of this decrease is unknown and warrants further investigation to determine the need for tailoring LPV dosage in selected cases."( Ritonavir-boosted atazanavir-lopinavir combination: a pharmacokinetic interaction study of total, unbound plasma and cellular exposures.
Biollaz, J; Buclin, T; Cavassini, M; Colombo, S; Decosterd, LA; Franc, C; Guignard, N; Khonkarly, M; Rochat, B; Tarr, PE; Telenti, A, 2006
)
1.78
" Blood samples for tenofovir (TFV) and SQV/RTV PK were drawn over respective 24- and 12-h dosing intervals, and drug concentrations were measured by liquid chromatography-tandem mass spectrometry."( Pharmacokinetics of tenofovir disoproxil fumarate and ritonavir-boosted saquinavir mesylate administered alone or in combination at steady state.
Adda, N; Begley, JA; Blum, MR; Chittick, GE; Kearney, BP; Sorbel, JJ; Zong, J, 2006
)
0.58
" A Bayesian approach is proposed to fit this model to clinical data from ACTG A5055, a study of two dosage regimens of indinavir (IDV) with ritonavir (RTV) in subjects failing their first protease inhibitor treatment."( Pharmacodynamics of antiretroviral agents in HIV-1 infected patients: using viral dynamic models that incorporate drug susceptibility and adherence.
Acosta, EP; Eron, JJ; Gerber, JG; Huang, Y; Kuritzkes, DR; Park, JG; Perelson, AS; Rosenkranz, SL; Wu, H; Yu, S, 2006
)
0.54
"Lopinavir/ritonavir is approved for treatment of HIV-infected children at a dosage regimen of 230/57."( Lopinavir/ritonavir exposure in treatment-naive HIV-infected children following twice or once daily administration.
Bassetti, M; Dentone, C; Di Biagio, A; Gatti, G; Gattinara, GC; Giaquinto, C; Martino, AM; Merlo, M; Rampon, O; Rosso, R; Viganò, A; Viscoli, C, 2006
)
1.14
"Atazanavir (ATV) is recommended to be dosed at 400 mg once daily or 300 mg daily coadministered with 100 mg ritonavir (RTV)."( Atazanavir plasma concentrations vary significantly between patients and correlate with increased serum bilirubin concentrations.
Jeganathan, S; Ray, J; Smith, DE,
)
0.34
" These findings indicate that short-term ritonavir dosing has only minimal impact on the pharmacokinetic disposition of a single dose of bupropion in healthy volunteers."( Ritonavir has minimal impact on the pharmacokinetic disposition of a single dose of bupropion administered to human volunteers.
Court, MH; Greenblatt, DJ; Hesse, LM; von Moltke, LL, 2006
)
2.04
" The only effect on plasma ESO exposure was a 55% increase in area under the plasma concentration-time curve during a dosing interval, tau[AUC0-tau], after coadministration of ESO 20 mg qd with FPV 1400 mg bid."( Coadministration of esomeprazole with fosamprenavir has no impact on steady-state plasma amprenavir pharmacokinetics.
Borland, J; Ford, SL; Lou, Y; Min, SS; Shelton, MJ; Wire, MB; Xue, ZG; Yuen, G, 2006
)
0.33
" In both instances, the carbamazepine dosage was decreased by 33%, which resulted in resolution of symptoms."( Carbamazepine toxicity induced by lopinavir/ritonavir and nelfinavir.
Bates, DE; Herman, RJ, 2006
)
0.59
" Carbamazepine toxicity may be prevented by reducing the carbamazepine dosage by 25-50% when protease inhibitors are introduced."( Carbamazepine toxicity induced by lopinavir/ritonavir and nelfinavir.
Bates, DE; Herman, RJ, 2006
)
0.59
" The medications, including their pharmacokinetic properties, side effects, and dosing are reviewed."( Antiretroviral therapy 2006: pharmacology, applications, and special situations.
Bettiker, R; Samuel, R; Suh, B, 2006
)
0.33
" Once-daily dosing may offer an advantage to adherence."( Pharmacokinetics of a once-daily regimen of lopinavir/ritonavir in HIV-1-infected children.
Burger, D; de Groot, R; van der Lee, M; Verweel, G, 2006
)
0.58
" Further research, especially in young children, is necessary to determine whether a higher dosage of lopinavir/ritonavir once daily must be given to reach the target level for Cmin."( Pharmacokinetics of a once-daily regimen of lopinavir/ritonavir in HIV-1-infected children.
Burger, D; de Groot, R; van der Lee, M; Verweel, G, 2006
)
0.79
" Additional investigations are warranted to determine the optimal dosing of FPV with LPV/RTV."( Dose separation does not overcome the pharmacokinetic interaction between fosamprenavir and lopinavir/ritonavir.
Corbett, AH; Eron, JJ; Kalvass, LA; Kashuba, AD; Lim, ML; Ngo, LT; Patterson, KB; Tien, HC, 2006
)
0.55
"When given alone, the apparent oral clearance of voriconazole after single oral dosing was 26%+/-16% (P > ."( Potent cytochrome P450 2C19 genotype-related interaction between voriconazole and the cytochrome P450 3A4 inhibitor ritonavir.
Burhenne, J; Ding, R; Drzewinska, M; Haefeli, WE; Mikus, G; Rengelshausen, J; Riedel, KD; Schöwel, V; Thomsen, T; Weiss, J, 2006
)
0.54
" Since ATV plasma levels are highly variable and seem to be correlated with both viral response and toxicity, dosage individualization based on plasma concentration monitoring might be indicated."( Population pharmacokinetics of atazanavir in patients with human immunodeficiency virus infection.
Biollaz, J; Buclin, T; Cavassini, M; Colombo, S; Csajka, C; Décosterd, LA; Telenti, A, 2006
)
0.33
" The single daily dosing was expected to improve adherence to treatment."( Change to a once-daily combination including boosted atazanavir in HIV-1-infected children.
Blanche, S; Delaugerre, C; Jullien, V; Macassa, E; Rouzioux, C; Teglas, JP; Tréluyer, JM; Veber, F, 2006
)
0.33
" The pharmacokinetics, safety, and effectiveness of increased LPV/r dosing during the third trimester of pregnancy should be investigated."( Reduced lopinavir exposure during pregnancy.
Best, BM; Burchett, SK; Capparelli, E; Cotter, A; Elgie, C; Holland, DT; Hu, C; Mirochnick, M; Read, JS; Smith, E; Stek, AM; Tuomala, R, 2006
)
0.33
" Atazanavir (ATV) has clinical efficacy comparable to a standard of care regimen in naive patients and, when dosed with low-dose ritonavir (RTV), also in treatment-experienced patients."( Effect of rifampin on steady-state pharmacokinetics of atazanavir with ritonavir in healthy volunteers.
Agarwala, S; Been-Tiktak, A; Bertz, R; Burger, DM; Child, M; Wang, Y, 2006
)
0.77
"Tenofovir measurements with an area under the concentration-time curve over the dosing interval, maximum concentration, and concentration at the end of the dosing interval (Ctau) were 32%, 15%, and 51% higher, respectively, when TDF was coadministered with LPV/r (n = 24)."( Pharmacokinetics and safety of tenofovir disoproxil fumarate on coadministration with lopinavir/ritonavir.
Cheng, AK; Ebrahimi, R; Kearney, BP; Mathias, A; Mittan, A; Sayre, J, 2006
)
0.55
" Geometric least-square (GLS) mean ratios and the associated 90% confidence intervals (CIs) were estimated for plasma APV maximum plasma concentrations (Cmax), the area under the plasma concentration-time curve over the dosing period (AUC0-tau), and trough concentrations (Ctau) during each dosing period."( Plasma amprenavir pharmacokinetics and tolerability following administration of 1,400 milligrams of fosamprenavir once daily in combination with either 100 or 200 milligrams of ritonavir in healthy volunteers.
Lancaster, CT; Lou, Y; Luber, AD; Pappa, KA; Ruane, PJ; Shelton, MJ; Wire, MB, 2007
)
0.53
" After a regular oral dose of Kaletra (400 mg lopinavir, 100 mg ritonavir) analyte concentrations were detectable over a full dosing interval in plasma, ultrafiltrate, and PBMCs."( Monitoring of lopinavir and ritonavir in peripheral blood mononuclear cells, plasma, and ultrafiltrate using a selective and highly sensitive LC/MS/MS assay.
Burhenne, J; Ehrhardt, M; Haefeli, WE; Mikus, G; Möck, M, 2007
)
0.87
" Melt extrusion technology was used to produce a tablet formulation reducing the number of dosage units administered per day and simplifying storage requirements."( The tablet formulation of lopinavir/ritonavir provides similar bioavailability to the soft-gelatin capsule formulation with less pharmacokinetic variability and diminished food effect.
Awni, W; Breitenbach, J; Brun, SC; Chiu, YL; Doan, T; Hanna, GJ; Heuser, RS; Klein, CE; Morris, JB; Zhu, T, 2007
)
0.61
" SQV pharmacokinetics were significantly higher when dosed with RTV compared to ATV (P < ."( Pharmacokinetics of saquinavir with atazanavir or low-dose ritonavir administered once daily (ASPIRE I) or twice daily (ASPIRE II) in seronegative volunteers.
Acosta, EP; Becker, SL; Kakuda, TN; King, JR; Paul, S; Tse, MM, 2007
)
0.58
" Blood samples were collected before and 1, 2, 3, 4, 6, 8, 10 and 12 h after dosing for measurement of nelfinavir, the nelfinavir metabolite M8 and saquinavir using liquid chromatography tandem mass spectrometry (LC-MS/MS)."( Saquinavir, nelfinavir and M8 pharmacokinetics following combined saquinavir, ritonavir and nelfinavir administration.
Arastéh, K; Becker, M; Berger, M; Breske, A; Herzmann, C; Hill, A; Kruse, G; Kurowski, M; Schulbin, H; Steinmüller, J; Stocker, H, 2007
)
0.57
" Twice-daily dosing or higher saquinavir doses in once-daily administration should be tested to obtain more appropriate plasma levels."( Pharmacokinetic interaction between rifampicin and the once-daily combination of saquinavir and low-dose ritonavir in HIV-infected patients with tuberculosis.
Azuaje, C; Colomer, J; Crespo, M; Curran, A; Domingo, P; Falcó, V; Feijoo, M; Lopez, RM; Lopez-Colomes, JL; Ocaña, I; Pahissa, A; Pou, L; Ribera, E; Sambeat, MA; Sánchez, P, 2007
)
0.55
"Most antiretrovirals are metabolized in the liver, and lower dosing could be advisable in patients with severe liver insufficiency."( Influence of liver fibrosis stage on plasma levels of antiretroviral drugs in HIV-infected patients with chronic hepatitis C.
Barreiro, P; Gonzalez-Lahoz, J; Jiménez-Nácher, I; Labarga, P; Martín-Carbonero, L; Rodríguez-Novoa, S; Ruiz, A; Soriano, V, 2007
)
0.34
" Lack of PK alteration for FTC, tenofovir (TFV), and GS-9137 was defined as a 90% confidence interval (CI) for the estimated ratio of geometric least squares means (coadministration/alone) between 70% and 143% for the primary PK parameters: maximum observed plasma concentration (Cmax), area under the plasma concentration-time curve over dosing interval (AUCtau), and trough concentration (Ctau)."( Pharmacokinetics of emtricitabine, tenofovir, and GS-9137 following coadministration of emtricitabine/tenofovir disoproxil fumarate and ritonavir-boosted GS-9137.
Cheng, A; Kearney, BP; Ramanathan, S; Shen, G, 2007
)
0.54
" The ATV area under the concentration-time curve from dosing to 24 hours after the dose (AUC0-24; GM: 36."( Beneficial pharmacokinetic interaction between atazanavir and lopinavir/ritonavir.
Agarwala, S; Barditch-Crovo, P; Carson, K; Flexner, C; Fuchs, E; Parsons, T; Pham, PA; Vasist, L, 2007
)
0.57
" The dosage was increased to 1600/100 mg in the remaining 3 episodes to achieve the target levels."( Efficacy of low-dose boosted saquinavir once daily plus nucleoside reverse transcriptase inhibitors in pregnant HIV-1-infected women with a therapeutic drug monitoring strategy.
Camacho, A; García-Lazaro, M; Lopez-Cortes, LF; Marquez-Solero, M; Ocampo, A; Rivero, A; Rodriguez-Baños, J; Ruiz-Valderas, R; Santos, J; Viciana, P, 2007
)
0.34
" This coformulation was designed to overcome the problems of earlier agents of this class of drugs concerning unfavorable pharmacokinetics with a higher frequency of dosing and therapy failure."( Lopinavir/ritonavir: appraisal of its use in HIV therapy.
von Hentig, N, 2007
)
0.74
" In clinical practice, it became common for clinicians to prescribe it with a ritonavir pharmacokinetic 'boost' to remove the food restriction, reduce the pill burden and enable a more convenient twice-daily dosing schedule."( Indinavir: the forgotten HIV-protease inhibitor. Does it still have a role?
Boyd, M, 2007
)
0.57
" Comparisons between 2 ritonavir dosing schedules revealed significant improvement in phase 1/2 decay constants in favor of the higher dose."( Virologic response to potent antiretroviral therapy and modeling of HIV dynamics in early pediatric infection.
Chadwick, E; Luzuriaga, K; Palumbo, P; Rodman, J; Ruan, P; Wu, H; Yogev, R, 2007
)
0.65
" Similar low dosing levels in children have also proven successful, but data in pregnant women remains limited."( Indinavir/ritonavir remains an important component of HAART for the treatment of HIV/AIDS, particularly in resource-limited settings.
Chokephaibulkit, K; Cressey, TR; Fregonese, F; Plipat, N, 2007
)
0.74
" Upon co-dosing either VX-950 or SCH 503034 with ritonavir in rats, plasma exposure of the HCV protease inhibitors was increased by > 15-fold, and plasma concentrations 8 h after dosing were increased by > 50-fold."( Pharmacokinetic enhancement of the hepatitis C virus protease inhibitors VX-950 and SCH 503034 by co-dosing with ritonavir.
Chen, HJ; Chovan, LE; Dandliker, PJ; Guan, Z; Hernandez, L; Kempf, DJ; Klein, C; Klein, LL; Lau, YY; Marsh, KC; Randolph, JT; Turner, TM; Yeung, C, 2007
)
0.8
" Tipranavir requires pharmacokinetic boosting by ritonavir (200 mg) to achieve therapeutic levels with twice-daily dosing and must be administered with food for optimal absorption."( Tipranavir: a new option for the treatment of drug-resistant HIV infection.
Feinberg, J; Temesgen, Z, 2007
)
0.59
" The approved dosage of darunavir is 600 mg in combination with ritonavir 100mg twice daily."( Clinical pharmacokinetics of darunavir.
Arastéh, K; Rittweger, M, 2007
)
0.58
"0% (oral) of dosed radioactivity in males and females, respectively."( Biotransformation and mass balance of tipranavir, a nonpeptidic protease inhibitor, when co-administered with ritonavir in Sprague-Dawley rats.
Beers, W; Chen, L; Macha, S; Mao, Y; Norris, SH; Philip, E; Silverstein, H; Struble, C, 2007
)
0.55
" When efavirenz (four patients) or a nucleoside analogue combination (one patient) was added, very little change in tacrolimus dosing was required."( Effect of highly active antiretroviral therapy on tacrolimus pharmacokinetics in hepatitis C virus and HIV co-infected liver transplant recipients in the ANRS HC-08 study.
Abbara, C; Barrail, A; Boissonnas, A; Bonhomme-Faivre, L; Duclos-Vallée, JC; Samuel, D; Taburet, AM; Teicher, E; Vincent, I; Vittecoq, D, 2007
)
0.34
" Tacrolimus dosing must be optimised according to therapeutic drug monitoring and the antiretroviral drug combination."( Effect of highly active antiretroviral therapy on tacrolimus pharmacokinetics in hepatitis C virus and HIV co-infected liver transplant recipients in the ANRS HC-08 study.
Abbara, C; Barrail, A; Boissonnas, A; Bonhomme-Faivre, L; Duclos-Vallée, JC; Samuel, D; Taburet, AM; Teicher, E; Vincent, I; Vittecoq, D, 2007
)
0.34
" Over the whole dosing interval, therapeutic drug concentrations well above the wild-type HIV 90% inhibitory concentration are maintained."( Atazanavir plus low-dose ritonavir in pregnancy: pharmacokinetics and placental transfer.
Airoldi, M; Bertuletti, P; Cattaneo, D; Frigerio, L; Maggiolo, F; Ripamonti, D; Ruggeri, M; Suter, F, 2007
)
0.64
" Increased systemic exposure to RFB and its equipotent active metabolite, 25-O-desacetyl-RFB (dAc-RFB), has been reported during concomitant administration of CYP3A4 inhibitors, including ritonavir (RTV), lopinavir, and amprenavir (APV); therefore, a reduction in the RFB dosage is recommended when it is coadministered with these protease inhibitors."( Pharmacokinetic interaction between fosamprenavir-ritonavir and rifabutin in healthy subjects.
Borland, J; Chen, YC; Ford, SL; Lou, Y; Min, SS; Shelton, MJ; Yuen, GJ, 2008
)
0.79
"Pharmacokinetic parameters were determined for indinavir, ritonavir, and amprenavir: area under the concentration-time curve from time 0 to 12 hours after dosing (AUC(0-12)), maximum plasma concentration (C(max)), and 12-hour plasma concentration (C(12))."( Pharmacokinetics of an indinavir-ritonavir-fosamprenavir regimen in patients with human immunodeficiency virus.
Acosta, EP; Easley, KA; Lennox, JL; Ofotokun, I; Pan, Y, 2008
)
0.87
"We assessed the safety and efficacy and evaluated the adherence to lopinavir/ritonavir (LPV/r) dosed QD or BID in antiretroviral-naive, HIV-1-infected subjects through 96 weeks of treatment."( A lopinavir/ritonavir-based once-daily regimen results in better compliance and is non-inferior to a twice-daily regimen through 96 weeks.
Domingo, P; Hairrell, JM; Hanna, GJ; Johnson, MA; King, MS; Molina, JM; Myers, R; Podsadecki, TJ; Rode, RA; Wilkin, A, 2007
)
0.95
" Rosuvastatin and lopinavir/ritonavir should be used with caution until the safety, efficacy, and appropriate dosing of this combination have been demonstrated in larger populations."( Drug/Drug interaction between lopinavir/ritonavir and rosuvastatin in healthy volunteers.
Flynn, DM; Gerber, JG; Hoody, DW; Kiser, JJ; Predhomme, JA; Wolfe, P, 2008
)
0.91
" Ritonavir (100 mg every 12 h) was dosed from day 3 to day 18."( Effect of low-dose ritonavir on the pharmacokinetics of the CXCR4 antagonist AMD070 in healthy volunteers.
Becker, S; Cao, YJ; Conley, J; Dunaway, S; Flexner, CW; Hendrix, CW; Kashuba, AD; Klingman, K; MacFarland, R; Park, JG; Radebaugh, C; Wiggins, I, 2008
)
1.58
" From days 6-15, volunteers were randomized to receive lopinavir/ritonavir tablets dosed as either 600/150 or 800/200 mg twice daily, both in addition to 600 mg rifampicin once daily."( High incidence of adverse events in healthy volunteers receiving rifampicin and adjusted doses of lopinavir/ritonavir tablets.
Aarnoutse, RE; Boeree, MJ; Burger, DM; Koopmans, PP; L'homme, RF; Nijland, HM; Rongen, GA; van Crevel, R; van Uden, P, 2008
)
0.8
" In this study, we investigated the long-term efficacy and safety of RTV-boosted ATV in rats with a clinical relevant dosage of ATV and RTV, 7 mg/kg and 2 mg/kg, respectively, and drew a direct comparison with RTV-boosted ATV and the previously reported ATV pharmaceutical formulation based on a solid dispersion system (ATV-SLS SD+G)."( Long-term pharmacokinetic efficacy and safety of low-dose ritonavir as a booster and atazanavir pharmaceutical formulation based on solid dispersion system in rats.
Fukushima, K; Haraya, K; Ito, Y; Sugioka, N; Takada, K; Terasaka, S, 2008
)
0.59
"Fifty children at 2 sites in Thailand were treated with standard dosing of SQV and LPV/r."( Double boosted protease inhibitors, saquinavir, and lopinavir/ritonavir, in nucleoside pretreated children at 48 weeks.
Ananworanich, J; Boonrak, P; Bunupuradah, T; Burger, D; Engchanil, C; Intasan, J; Kosalaraksa, P; Lumbiganon, P; Ruxrungtham, K; Schutz, M, 2008
)
0.59
" The subjects had 11 plasma samples drawn over a dosing interval on days 1, 15, and 30."( Effects of minocycline and valproic acid coadministration on atazanavir plasma concentrations in human immunodeficiency virus-infected adults receiving atazanavir-ritonavir.
Cruttenden, K; DiCenzo, R; Gelbard, H; Hochreiter, J; Mariuz, P; Peterson, DR; Rezk, NL; Schifitto, G, 2008
)
0.54
" N-desmethyl sildenafil Cmax and AUC from the time of administration until the last time point with a measurable concentration after dosing (calculated by linear trapezoidal summation [AUClast]) values decreased by approximately 95% when sildenafil 25 mg was co-administered with DRV/r compared with sildenafil 100 mg alone."( Effect of repeated doses of darunavir plus low-dose ritonavir on the pharmacokinetics of sildenafil in healthy male subjects: phase I randomized, open-label, two-way crossover study.
De Marez, T; De Paepe, E; De Pauw, M; Hoetelmans, RM; Lefebvre, E; Sekar, V; Vangeneugden, T, 2008
)
0.6
"The objective of this study was to examine the potential of once-daily dosing with darunavir/ritonavir 800/100 mg in a HIV-infected, treatment-experienced patient population with no baseline darunavir resistance-associated mutations (RAMs)."( Efficacy of once-daily darunavir/ritonavir 800/100 mg in HIV-infected, treatment-experienced patients with no baseline resistance-associated mutations to darunavir.
de Béthune, MP; De Meyer, SM; Miralles, GD; Spinosa-Guzman, S; Vangeneugden, TJ, 2008
)
0.85
"The pharmacokinetics of EFV and LPV/RTV in hemodialysis suggests that no dosing adjustments are necessary in treatment-naive patients."( The pharmacokinetics and pharmacogenomics of efavirenz and lopinavir/ritonavir in HIV-infected persons requiring hemodialysis.
Amorosa, V; Cramer, YS; Gupta, SK; Hall, SD; Koletar, SL; Rosenkranz, SL; Szczech, LA, 2008
)
0.58
" This study systematically evaluated the ritonavir dose-response relationship on presystemic and systemic CYP3A metabolism using the human immunodeficiency virus integrase inhibitor elvitegravir and midazolam as probe substrates."( Dose-response of ritonavir on hepatic CYP3A activity and elvitegravir oral exposure.
Hui, J; Kearney, BP; Mathias, AA; West, S, 2009
)
0.96
" Further exploration of regimens and dosing of antiretrovirals for children in these settings is needed."( Two-year outcomes of children on non-nucleoside reverse transcriptase inhibitor and protease inhibitor regimens in a South African pediatric antiretroviral program.
Berrisford, AE; Boulle, AM; Jaspan, HB, 2008
)
0.35
"The aim of this study was to develop and validate a population pharmacokinetic model in order to describe ritonavir-boosted saquinavir concentrations dosed twice and once daily in human immunodeficiency virus (HIV)-infected patients from the UK, Uganda and Thailand and to identify factors that may influence saquinavir pharmacokinetics."( Population pharmacokinetics of ritonavir-boosted saquinavir regimens in HIV-infected individuals.
Aarons, LJ; Autar, RS; Back, DJ; Boffito, M; Burger, DM; Dickinson, L; Khoo, SH; Merry, C; Mugyenyi, P; Pozniak, AL, 2008
)
0.85
" The ritonavir area under the curve over the dosing interval was significantly associated with saquinavir CL/F and V/F."( Population pharmacokinetics of ritonavir-boosted saquinavir regimens in HIV-infected individuals.
Aarons, LJ; Autar, RS; Back, DJ; Boffito, M; Burger, DM; Dickinson, L; Khoo, SH; Merry, C; Mugyenyi, P; Pozniak, AL, 2008
)
1.15
" The model could be used for dosage adaptation following therapeutic drug monitoring and to assess patients' suitability for once-daily boosted saquinavir therapy."( Population pharmacokinetics of ritonavir-boosted saquinavir regimens in HIV-infected individuals.
Aarons, LJ; Autar, RS; Back, DJ; Boffito, M; Burger, DM; Dickinson, L; Khoo, SH; Merry, C; Mugyenyi, P; Pozniak, AL, 2008
)
0.63
"The pharmacokinetics and safety of BILR 355 following oral repeated dosing coadministered with low doses of ritonavir (RTV) were investigated in 12 cohorts of healthy male volunteers with a ratio of 6 to 2 for BILR 355 versus the placebo."( Pharmacokinetics of BILR 355 after multiple oral doses coadministered with a low dose of ritonavir.
Ballow, C; Castles, M; Drda, K; Huang, F; MacGregor, TR; Nguyen, T; Robinson, P; Rowland, L; Scherer, J, 2009
)
0.79
"Use of standard adult lopinavir/ritonavir (LPV/RTV) dosing (400/100 mg) during the third trimester of pregnancy results in reduced LPV exposure."( Lopinavir exposure with an increased dose during pregnancy.
Best, BM; Burchett, SK; Capparelli, E; Gaddipati, S; Holland, DT; Hu, C; Mirochnick, M; Read, JS; Smith, E; Stek, AM, 2008
)
0.63
" These data suggest that the higher LPV/RTV dose should be used in third trimester pregnant women; that it should be considered in second trimester pregnant women, especially those who are protease inhibitor experienced; and that postpartum LPV/RTV dosing can be reduced to standard dosing by 2 weeks after delivery."( Lopinavir exposure with an increased dose during pregnancy.
Best, BM; Burchett, SK; Capparelli, E; Gaddipati, S; Holland, DT; Hu, C; Mirochnick, M; Read, JS; Smith, E; Stek, AM, 2008
)
0.35
" The objectives of this study were to investigate the effect of ketoconazole on the pharmacokinetics of saquinavir/ritonavir and vice versa using the approved dosage regimens of saquinavir/ritonavir at 1,000/100 mg twice daily and ketoconazole at 200 mg once daily."( Drug-drug interaction study of ketoconazole and ritonavir-boosted saquinavir.
Bour, F; Kaeser, B; Schmitt, C; Zandt, H; Zhang, X; Zwanziger, E, 2009
)
0.82
" Three different dosing regimens of elvucitabine were administered with lopinavir-ritonavir to 24 subjects with moderate levels of HIV."( Multiple-dose pharmacokinetic behavior of elvucitabine, a nucleoside reverse transcriptase inhibitor, administered over 21 days with lopinavir-ritonavir in human immunodeficiency virus type 1-infected subjects.
Colucci, P; Ducharme, MP; Hoepelman, IM; Pottage, JC; Robison, H; Schürmann, D; Turgeon, J, 2009
)
0.78
" These values were lower compared with the half-life over the respective dosing intervals (7."( Pharmacokinetics of atazanavir/ritonavir once daily and lopinavir/ritonavir twice and once daily over 72 h following drug cessation.
Back, D; Boffito, M; Else, L; Gazzard, B; Khoo, S; Moyle, G; Pozniak, A; Sousa, M; Taylor, J, 2008
)
0.63
" The PK parameters of ATV/RTV at a dosage of 200/100 mg once daily, plus two nucleoside reverse transcriptase inhibitors, were significantly lower than those associated with a dosage of 300/100 mg once daily in the same patients."( A low dose of ritonavir-boosted atazanavir provides adequate pharmacokinetic parameters in HIV-1-infected Thai adults.
Avihingsanon, A; Burger, DM; Chanmano, S; Cooper, DA; Gorowara, M; Kerr, SJ; Lange, J; Ohata, P; Phanuphak, P; Ruxrungtham, K; van der Lugt, J, 2009
)
0.71
" No evidence is available to guide dosing of lopinavir/ritonavir in tablet formulation in this setting."( Lopinavir/ritonavir pharmacokinetics in a substitution of high-dose soft-gelatin capsule to tablet formulation.
Guillemi, S; Harrigan, PR; Harris, M; Hull, MW; Lima, V; Montaner, JS, 2009
)
1
" Group 1 (n = 20) followed a sequence of 10-day dosing of elvitegravir/r (150/100 mg once daily) and elvitegravir/r plus etravirine (200 mg twice daily) or the reverse (n = 10 per sequence)."( Pharmacokinetics of elvitegravir and etravirine following coadministration of ritonavir-boosted elvitegravir and etravirine.
Kakuda, TN; Kearney, BP; Mack, R; Ramanathan, S; West, S, 2008
)
0.57
" Elvitegravir pharmacokinetic GMR was 6-7% higher following elvitegravir/r plus etravirine dosing versus elvitegravir/r."( Pharmacokinetics of elvitegravir and etravirine following coadministration of ritonavir-boosted elvitegravir and etravirine.
Kakuda, TN; Kearney, BP; Mack, R; Ramanathan, S; West, S, 2008
)
0.57
" We have some theoretical and clinical data available that enables us to consider the possibility of administering DRV/r once a day in some patients with a few mutations in the protease and in those where this dosing regime is considered important."( [Darunavir as first-line therapy. The TITAN study].
Curran, A; Ribera Pascuet, E, 2008
)
0.35
" Consequently, the coadministration of elvitegravir with the protease inhibitor ritonavir (a substantial CYP3A4 inhibitor) results in significantly enhanced bioavailability and a longer half-life than with elvitegravir alone, allowing for the once-daily dosing of elvitegravir."( Elvitegravir, an oral HIV integrase inhibitor, for the potential treatment of HIV infection.
Klibanov, OM, 2009
)
0.58
"A total of 191 patients were randomized 2:2:2:1 to one of three APL dosing regimens or to lamivudine (3TC)/zidovudine (ZDV) twice daily (bid), each in combination with lopinavir/ritonavir (LPV/r) 400 mg/100 mg bid."( Antiviral activity and safety of aplaviroc, a CCR5 antagonist, in combination with lopinavir/ritonavir in HIV-infected, therapy-naïve patients: results of the EPIC study (CCR100136).
Adkison, KK; Berger, D; Bonny, T; Cimoch, P; Lamarca, A; Lazzarin, A; Madison, SJ; McCarty, D; Millard, J; Nichols, WG; Salvato, P; Smaill, FM; Teofilo, E; Yeni, P, 2009
)
0.77
" For the overall POWER trial population, with significant baseline resistance to PIs, the rates of HIV RNA suppression <50 copies/mL at Week 24 for darunavir/ritonavir 800/100 mg once-daily [DOSAGE ERROR CORRECTED] were lower than for the 600/100 mg twice-daily dosage (31% vs."( Pharmacokinetics, efficacy, and safety of darunavir/ritonavir 800/100 mg once-daily in treatment-naïve and -experienced patients.
Boffito, M; Hill, A; Miralles, D,
)
0.58
" Study M05-730 compared LPV/r tablets dosed once daily vs."( A once-daily lopinavir/ritonavir-based regimen is noninferior to twice-daily dosing and results in similar safety and tolerability in antiretroviral-naive subjects through 48 weeks.
Bernstein, B; Cohen, DE; da Silva, BA; Fredrick, L; Gathe, J; Gibbs, S; Loutfy, MR; Marsh, T; Naylor, C; Podzamczer, D; Rubio, R, 2009
)
0.66
" Safety and tolerability of once-daily and twice-daily dosing was also comparable."( A once-daily lopinavir/ritonavir-based regimen is noninferior to twice-daily dosing and results in similar safety and tolerability in antiretroviral-naive subjects through 48 weeks.
Bernstein, B; Cohen, DE; da Silva, BA; Fredrick, L; Gathe, J; Gibbs, S; Loutfy, MR; Marsh, T; Naylor, C; Podzamczer, D; Rubio, R, 2009
)
0.66
" We report the pharmacokinetics of standard LPV-ritonavir dosing (400/100 mg twice daily) in the immediate and early postpartum period when initiated during labor."( Early postpartum pharmacokinetics of lopinavir initiated intrapartum in Thai women.
Achalapong, J; Chotivanich, N; Cressey, TR; Jourdain, G; Maupin, R; Mirochnick, M; Prommas, S; Puthanakit, T; Roongpisuthipong, A; Shapiro, DE; Smith, E; Van Dyke, R; Yuthavisuthi, P, 2009
)
0.61
" Statistical analyses were performed on pharmacokinetic parameters: the area under the concentration-time curve from 0 to 4 h (AUC(0-4)), the maximum concentration of the drug (C(max)), and the residual concentration of the drug at the end of the dosing interval (C(trough)) for plasma and the AUC(0-4) and C(trough) for intracellular data."( Pilot pharmacokinetic study of human immunodeficiency virus-infected patients receiving tenofovir disoproxil fumarate (TDF): investigation of systemic and intracellular interactions between TDF and abacavir, lamivudine, or lopinavir-ritonavir.
Ayen, R; Clotet, B; Grassi, J; Levi, M; Negredo, E; Pruvost, A; Puig, J; Théodoro, F, 2009
)
0.54
" The availability of antiretrovirals that are coformulated and dosed once daily have reduced pill burden and have simplified dosing schedules, but have not lowered drug exposure or cost."( Simplification strategies to reduce antiretroviral drug exposure: progress and prospects.
McKinnon, JE; Mellors, JW; Swindells, S, 2009
)
0.35
" The model was used to investigate other, particularly lower, ritonavir-boosted atazanavir dosing strategies."( Population pharmacokinetics of ritonavir-boosted atazanavir in HIV-infected patients and healthy volunteers.
Aarons, L; Back, D; Boffito, M; Davies, G; Dickinson, L; Else, L; Khoo, S; Pozniak, A; Waters, L, 2009
)
0.88
" However, oxymethylphosphate (OMP) and oxyethylphosphate (OEP) prodrugs provided improved rates of cleavage, high levels of aqueous solubility, and high plasma levels of the parent drugs when dosed orally in rats and dogs."( Water-soluble prodrugs of the human immunodeficiency virus protease inhibitors lopinavir and ritonavir.
DeGoey, DA; Flosi, WJ; Grampovnik, DJ; Kati, WM; Kempf, DJ; Klein, LL; Liu, Y; Long, MA; Marsh, KC; McDaniel, KF; Molla, A; Wang, XC, 2009
)
0.57
" Although a few interactions can be managed with adequate drug dosing others preclude to use these medications in combination."( Drug interactions of tipranavir, a new HIV protease inhibitor.
Jimenez-Nacher, I; Morello, J; Rodriguez-Novoa, S; Soriano, V, 2007
)
0.34
" Pharmacokinetic studies of higher dosages of rifampin are urgently needed in children to assist in placing the dosage of rifampin used in childhood on a more scientific foundation."( Rifampin pharmacokinetics in children, with and without human immunodeficiency virus infection, hospitalized for the management of severe forms of tuberculosis.
Cilliers, K; Donald, PR; Hussey, GD; Labadarios, D; Maritz, JS; McIlleron, H; Schaaf, HS; Smith, P; Willemse, M, 2009
)
0.35
" A few days later, typical secondary adrenal failure developed and was confirmed by dosage of cortisol and ACTH, both low."( [Iatrogenic Cushing's syndrome, diabetes mellitus and secondary adrenal failure in a human immunodeficiency virus patient treated with ritonavir boosted atazanavir and fluticasone].
Beressi, J-; Collet-Gaudillat, C; Desforges-Bullet, V; Doll, J; Petit-Aubert, G; Roussin-Bretagne, S, 2009
)
0.56
"In total, 50 children from two sites in Thailand were treated with standard dosing of SQV and LPV/r."( Safety and efficacy of a double-boosted protease inhibitor combination, saquinavir and lopinavir/ritonavir, in pretreated children at 96 weeks.
Ananworanich, J; Boonrak, P; Bunupuradah, T; Burger, D; Engchanil, C; Kosalaraksa, P; Lumbiganon, P; Mahanontharit, A; Mengthaisong, T; Puthanakit, T; Ruxrungtham, K; Tompkins, E; van der Lugt, J, 2009
)
0.57
"This study suggests that the pharmacokinetics of lopinavir after twice-daily and once-daily dosing are similar, with no observable difference in tolerability, in this group of patients between 5 and 15 years old."( Pharmacokinetics and tolerability of once- versus twice-daily lopinavir/ritonavir treatment in HIV-1-infected children.
la Porte, C; Mitchell, CD; Parker, J; Rongkavilit, C; van Heeswijk, R; Zhang, G, 2009
)
0.59
"HIV-1-infected adults receiving a regimen containing ritonavir-boosted atazanavir or fosamprenavir were recruited into a prospective study of adherence and dosage timing of both agents."( Occurrence of selective ritonavir nonadherence and dose-staggering in recipients of boosted HIV-1 protease inhibitor therapy.
Rode, RA; Sarlo, JA; Shuter, J; Zingman, BS,
)
0.69
"Atazanavir 400 mg/day plus fosamprenavir 1400 mg/day significantly decreased concentrations of atazanavir compared with standard dosing regimens of each drug alone."( Pharmacokinetics of concurrent administration of fosamprenavir and atazanavir without ritonavir in human immunodeficiency virus-negative subjects.
Anderson, PL; Clay, PG; Glaros, AG; McRae, M, 2009
)
0.58
" Ritonavir is therefore able to enhance the effectiveness of PI treatment by reducing the pill burden, simplifying dosing regimens and improving therapy adherence."( Pharmacokinetic enhancers for HIV drugs.
Desai, MC; Xu, L, 2009
)
1.26
" For subjects with mild hepatic impairment, the studied regimen of fosamprenavir 700 mg twice daily plus ritonavir 100 mg once daily delivered 17% higher values for the maximum plasma amprenavir concentration at the steady state (C(max)), 22% higher values for the area under the plasma concentration versus time curve over the dosing interval at the steady state [AUC(0-tau)], similar values for the concentration at the end of the dosing interval (C(tau)), and 114% higher unbound C(tau) values."( Pharmacokinetics of fosamprenavir plus ritonavir in human immunodeficiency virus type 1-infected adult subjects with hepatic impairment.
Clotet, B; Felizarta, F; Gutiérrez, F; Hernández-Quero, J; Lou, Y; Morellon, ML; Nichols, G; Ortega, E; Pérez-Elías, MJ; Pineda, JA; Rodríguez-Torres, M; Wire, MB, 2009
)
0.84
"A total of 136 antiretroviral-naïve patients, with a CD4 cell count above 100 cells/microL and a plasma HIV RNA below 100,000 HIV-1 RNA copies/mL, were randomized and dosed with either lopinavir/ritonavir monotherapy (n = 83) or lopinavir/ritonavir + zidovudine/lamivudine (n = 53)."( Long-term (96-week) follow-up of antiretroviral-naïve HIV-infected patients treated with first-line lopinavir/ritonavir monotherapy in the MONARK trial.
Chaix, ML; Cohen-Codar, I; Delfraissy, JF; Dellamonica, P; Flandre, P; Ghosn, J; Girard, PM; Ngovan, P; Norton, M; Raffi, F, 2010
)
0.76
" After dosing with TPV/r for 10 days, EFV 600 mg once a day was added to the regimen."( Lack of effect of efavirenz on the pharmacokinetics of tipranavir-ritonavir in healthy volunteers.
Béïque, L; Cameron, DW; la Porte, CJ; Sabo, JP, 2009
)
0.59
" Pharmacokinetics, safety and efficacy were assessed following 2-week dosing (part I), which determined dosing for part II (evaluated 48-week safety and efficacy)."( Pharmacokinetics, safety and efficacy of darunavir/ritonavir in treatment-experienced children and adolescents.
Blanche, S; Bologna, R; Cahn, P; Dierynck, I; Flynn, P; Fortuny, C; Rugina, S; Sekar, V; Spinosa-Guzman, S; van Baelen, B; Vis, P, 2009
)
0.6
" No dosage modification of BUP/NLX is required when co-administered with TPV/r."( Pharmacokinetic interactions between buprenorphine/naloxone and tipranavir/ritonavir in HIV-negative subjects chronically receiving buprenorphine/naloxone.
Altice, FL; Andrews, L; Bruce, RD; Conner, C; Fang, WB; Friedland, GH; Lin, SN; Moody, DE; Piliero, PJ; Sabo, JP; Wruck, JM, 2009
)
0.58
"Once-daily treatment with ritonavir-boosted saquinavir was well tolerated and resulted in similar saquinavir drug exposure despite much lower ritonavir concentrations when compared with a twice-daily dosing schedule."( Once-daily treatment with saquinavir mesylate (2000 mg) and ritonavir (100 mg) together with a fixed-dose combination of abacavir/lamivudine (600/300 mg) or tenofovir/emtricitabine (245/200 mg) in HIV-1-infected patients.
Bickel, M; Bodtländer, A; Gute, P; Klauke, S; Knecht, GK; Kurowski, M; Lutz, T; Stephan, C; von Hentig, N, 2009
)
0.9
" No differences were noted in adverse events among dosing periods."( Steady-state amprenavir and tenofovir pharmacokinetics after coadministration of unboosted or ritonavir-boosted fosamprenavir with tenofovir disoproxil fumarate in healthy volunteers.
Andrews, M; Condoluci, DV; Luber, AD; Olson, K; Pakes, GE; Pappa, KA; Peloquin, CA; Slowinski, PD, 2010
)
0.58
"Virologically controlled patients treated with lopinavir/ritonavir were included in a 48 week pilot trial during which lopinavir/ritonavir dosage was reduced if lopinavir concentration was >5000 ng/mL at inclusion."( Impact of reduced dosing of lopinavir/ritonavir in virologically controlled HIV-infected patients: the Kaledose trial.
Girard, PM; Lacombe, K; Meynard, JL; Morand-Joubert, L; Poirier, JM; Slama, L; Valantin, MA, 2010
)
0.88
"This pilot study evaluating the biochemical and virological impact of a reduced dosing of lopinavir/ritonavir suggests that lower exposure to lopinavir/ritonavir could be associated with a significant decrease in triglycerides during treatment, without occurrence of resistance mutations that might impact the virological response to treatment."( Impact of reduced dosing of lopinavir/ritonavir in virologically controlled HIV-infected patients: the Kaledose trial.
Girard, PM; Lacombe, K; Meynard, JL; Morand-Joubert, L; Poirier, JM; Slama, L; Valantin, MA, 2010
)
0.85
" Each subject had 8 plasma lopinavir concentrations determined over a 12-hour dosing interval and 1 CSF lopinavir C(trough) value determined at the end of the study."( Lopinavir cerebrospinal fluid steady-state trough concentrations in HIV-infected adults.
Cruttenden, K; DiCenzo, R; DiFrancesco, R; Donnelly, J; Schifitto, G, 2009
)
0.35
"In this case, dosing recommendations of itraconazole 200 mg daily with lopinavir/ritonavir were appropriate."( Drug-drug interaction between itraconazole and the protease inhibitor lopinavir/ritonavir.
Hills-Nieminen, C; Houston, S; Hughes, CA; Shafran, SD, 2009
)
0.81
"The dose of itraconazole was reduced to 200 mg daily as recommended by current guidelines, and therapeutic drug monitoring of both itraconazole and lopinavir concentrations confirmed that no further dosage adjustments were necessary."( Drug-drug interaction between itraconazole and the protease inhibitor lopinavir/ritonavir.
Hills-Nieminen, C; Houston, S; Hughes, CA; Shafran, SD, 2009
)
0.58
"Atazanavir (ATV) is a protease inhibitor (PI) in which its main qualities, compared to other PI are dosing convenience, good tolerability and excellent metabolic profile."( [Clinical utility of atazanavir].
Curran, A; Ribera Pascuet, E, 2008
)
0.35
"LPV/r dosed QD resulted in increased treatment adherence and was as efficacious as BID LPV/r while providing similar safety, tolerability, and limited resistance evolution."( Similar safety and efficacy of once- and twice-daily lopinavir/ritonavir tablets in treatment-experienced HIV-1-infected subjects at 48 weeks.
Andrade-Villanueva, J; Badal-Faesen, S; Bernstein, BM; Fredrick, LM; Gathe, J; Gaultier, IA; Mingrone, H; Podsadecki, TJ; Woodward, WC; Zajdenverg, R, 2010
)
0.6
" There was no relationship between elvitegravir dosage and adverse events."( Activity of elvitegravir, a once-daily integrase inhibitor, against resistant HIV Type 1: results of a phase 2, randomized, controlled, dose-ranging clinical trial.
Berger, DS; Cheng, AK; Chuck, SL; Enejosa, JV; Kearney, BP; Lampiris, H; Zhong, L; Zolopa, AR, 2010
)
0.36
" Multiple dosing produced weak induction of CYP1A2, moderate induction of CYP2C19, potent induction of intestinal P-gp, and potent inhibition of CYP2D6 and CYP3A, with no significant effects on CYP2C9 and hepatic P-gp."( A phenotype-genotype approach to predicting CYP450 and P-glycoprotein drug interactions with the mixed inhibitor/inducer tipranavir/ritonavir.
Castles, MA; Choi, SO; Drulak, M; Dumond, JB; Jennings, SH; Kashuba, AD; La-Beck, NM; Li, J; Macgregor, TR; Patterson, KB; Rezk, NL; Sabo, JP; Tien, HC; Vourvahis, M; Wagner, MJ; White, N, 2010
)
0.56
" To compare efficacy of once-daily (QD) versus twice-daily (BID) antiretroviral therapy, we randomized human immunodeficiency virus (HIV)-positive, treatment-naive patients to lopinavir-ritonavir (LPV/r) administered at a dosage of 400 mg of lopinavir and 100 mg of ritonavir BID (n = 160) or 800 mg of lopinavir and 200 mg of ritonavir QD (n = 161), plus either emtricitabine 200 mg QD and extended-release stavudine at a dosage of 100 mg QD or tenofovir at a dosage of 300 mg QD."( Comparison of once-daily versus twice-daily combination antiretroviral therapy in treatment-naive patients: results of AIDS clinical trials group (ACTG) A5073, a 48-week randomized controlled trial.
Aberg, J; Andrade, A; Dehlinger, M; Eshleman, SH; Ferguson, E; Flexner, C; Gross, R; Kashuba, A; Kmack, A; Lalama, C; Mildvan, D; Parsons, T; Rosenkranz, SL; Sanne, I; Tierney, C, 2010
)
0.55
" A total of 82% of subjects completed the study, and 71% continued to receive the initially assigned dosage schedule."( Comparison of once-daily versus twice-daily combination antiretroviral therapy in treatment-naive patients: results of AIDS clinical trials group (ACTG) A5073, a 48-week randomized controlled trial.
Aberg, J; Andrade, A; Dehlinger, M; Eshleman, SH; Ferguson, E; Flexner, C; Gross, R; Kashuba, A; Kmack, A; Lalama, C; Mildvan, D; Parsons, T; Rosenkranz, SL; Sanne, I; Tierney, C, 2010
)
0.36
" A total of 10 patients receiving TPV/ritonavir (TPV/RTV) 500/200 for longer than 6 months were randomized to stay on the same dosing schedule or switch to TPV/RTV 500/100."( The benefit of simplification from tipranavir/ritonavir 500/200 bid to 500/100 bid guided by therapeutic drug monitoring.
Barreiro, P; Blanco, F; González-Lahoz, J; Gonzalez-Pardo, G; Jiménez-Nácher, I; Morello, J; Rodríguez-Novoa, S; Soriano, V; Vispo, E, 2010
)
0.89
"Downward dosage adjustment of quinine appears necessary when concurrently administered with ritonavir."( Pharmacokinetic interactions between ritonavir and quinine in healthy volunteers following concurrent administration.
Cook, JM; Omoruyi, SI; Onyeji, CO; Owolabi, AR; Sarma, PV; Soyinka, JO, 2010
)
0.85
"5, the dosage was switched to 600/100 mg twice daily."( Treatment simplification to once daily darunavir/ritonavir guided by the darunavir inhibitory quotient in heavily pretreated HIV-infected patients.
Barbanoj, MJ; Cedeño, S; Clotet, B; Miranda, C; Moltó, J; Mothe, B; Negredo, E; Santos, JR; Valle, M; Videla, S; Yritia, M, 2010
)
0.62
" These data suggest that 2 tablets of lopinavir-ritonavir BD may be inadequate when dosed with NNRTIs in Ugandan adults, and the dosage should be increased by the addition of an additional adult tablet or a half-dose tablet (100/25 mg), where available."( Pharmacokinetics of lopinavir-ritonavir with and without nonnucleoside reverse transcriptase inhibitors in Ugandan HIV-infected adults.
Dickinson, L; Gibb, DM; Gilks, CF; Kayiwa, J; Khoo, S; Kityo, C; Lutwama, F; Munderi, P; Nalumenya, R; Reid, A; Ssali, F; Tumukunde, D; Walker, AS, 2010
)
0.9
" Dosage adjustment of bupropion may be needed when administered with ritonavir."( Dose-related reduction in bupropion plasma concentrations by ritonavir.
Ascher, J; Brittain, C; Iavarone, L; McConn, DJ; Muir, KT; Park, J; Sutherland, SM; Vousden, M, 2010
)
0.84
" No dosage adjustment for S/GSK1349572 is required when used with lopinavir/ritonavir or darunavir/ritonavir."( The effect of lopinavir/ritonavir and darunavir/ritonavir on the HIV integrase inhibitor S/GSK1349572 in healthy participants.
Borland, J; Chen, S; Ishibashi, T; Lou, Y; Min, SS; Patel, P; Piscitelli, SC; Song, I, 2011
)
0.91
" Lopinavir CL/F increased linearly during the dosing interval."( Population pharmacokinetics of lopinavir in combination with rifampicin-based antitubercular treatment in HIV-infected South African children.
Elsherbiny, D; Maartens, G; McIlleron, H; Ren, Y; Simonsson, US, 2010
)
0.36
" The time-dependent lopinavir CL/F might be due to a time-dependent recovery from ritonavir inhibition of lopinavir metabolism during the dosing interval."( Population pharmacokinetics of lopinavir in combination with rifampicin-based antitubercular treatment in HIV-infected South African children.
Elsherbiny, D; Maartens, G; McIlleron, H; Ren, Y; Simonsson, US, 2010
)
0.59
" During a 12 h dosing interval plasma and peripheral blood mononuclear cells were collected."( Intracellular and plasma steady-state pharmacokinetics of raltegravir, darunavir, etravirine and ritonavir in heavily pre-treated HIV-infected patients.
Beijnen, JH; Huitema, AD; Mulder, JW; Ter Heine, R; van Gorp, EC; Wagenaar, JF, 2010
)
0.58
" This strategy results in prolonged dosage intervals and reduced tablet intake due to a reduced overall dose that is needed."( [Cushing syndrome after steroid-infiltration in two HIV-patients with antiretroviral therapy].
Günthard, HF; Herold, MA, 2010
)
0.36
" The exposure (AUC within the dosage interval and at steady state [AUC(τ)]) to desRFB was considerably increased (by 881%) following treatment with DRV/r/RFB."( Pharmacokinetics of darunavir/ritonavir and rifabutin coadministered in HIV-negative healthy volunteers.
Berckmans, C; De Pauw, M; Hoetelmans, R; Lavreys, L; Sekar, V; Spinosa-Guzman, S; Van de Casteele, T; Vangeneugden, T, 2010
)
0.65
" Moreover, an inverse correlation between dosage of ritonavir and mean nadir neutrophil count was found."( Detrimental clinical interaction between ritonavir-boosted protease inhibitors and vinblastine in HIV-infected patients with Hodgkin's lymphoma.
Cingolani, A; de Gaetano Donati, K; Larocca, LM; Murri, R; Pinnetti, C; Tacconelli, E; Teofili, L; Torti, L, 2010
)
0.88
" Buprenorphine/naloxone and LPV/r can be safely coadministered without need for dosage modification."( Pharmacokinetic interactions between buprenorphine/naloxone and once-daily lopinavir/ritonavir.
Altice, FL; Andrews, L; Bruce, RD; Fang, WB; Friedland, GH; Lin, SN; Ma, Q; Moody, DE; Morse, GD, 2010
)
0.58
" Emergence of postbaseline resistance mutations occurred at similar low rates in each dosing group."( Short communication: Comparable safety and efficacy with once-daily versus twice-daily dosing of lopinavir/ritonavir tablets with emtricitabine + tenofovir DF in antiretroviral-naïve, HIV type 1-infected subjects: 96 week final results of the randomized t
Bernstein, B; Cohen, D; da Silva, B; Fredrick, L; González-García, J; Johnson, M; Naylor, C; Sloan, L, 2010
)
0.57
" Coadministration with LPV/r resulted in a 51% decrease in steady-state area under plasma concentration-time curve from 0 to 12 hours (AUC(0-12,ss)) and steady-state maximum measured plasma concentration over a dosing interval (C(max,ss)) and a 50% decrease in steady-state plasma concentration 12 hours post last dosing (C(12,ss)) for BILR 355."( Coadministration with lopinavir and ritonavir decreases exposure to BILR 355, a nonnucleoside reverse transcriptase inhibitor, in healthy volunteers.
Berger, F; Castles, MA; Huang, DB; Huang, F; MacGregor, TR; Robinson, P; Scholl, P; Vinisko, R, 2011
)
0.64
"Addition of darunavir/ritonavir to etravirine, all dosed once daily, did not have a clinically significant effect on the pharmacokinetics of etravirine."( Pharmacokinetics of once-daily etravirine without and with once-daily darunavir/ritonavir in antiretroviral-naive HIV type-1-infected adults.
DeJesus, E; Kakuda, TN; Lalezari, JP; Osiyemi, OO; Ruane, PJ; Ryan, R; Witek, J, 2010
)
0.9
" Conclusions The above target concentrations achieved in the majority of women and similarities in the fu% suggest standard dosing of the LPV/r tablet is appropriate during pregnancy."( Therapeutic drug monitoring of lopinavir/ritonavir in pregnancy.
Back, DJ; Boyle, N; Breiden, J; Brennan, M; Connor, EO; Coulter-Smith, S; Dickinson, L; Else, LJ; Fleming, C; Gibbons, S; Jackson, V; Khoo, SH; Lambert, JS, 2011
)
0.64
"Ritonavir-related adverse events have been reported in patients taking tipranavir/ritonavir at the licensed dosage of 500/200 mg twice daily (bid)."( Efficacy and safety of ritonavir dose reduction based on the tipranavir inhibitory quotient in HIV-infected patients on salvage antiretroviral therapy with tipranavir/ritonavir.
Cedeño, S; Clotet, B; Miranda, C; Moltó, J; Negredo, E; Santos, JR; Valle, M; Videla, S; Yritia, M, 2010
)
2.11
" The geometric mean raltegravir maximum concentration, the concentration at the end of the dosing interval, and the area under the concentration-time curve during the dose interval in plasma versus PBMCs were 2,640 ng/ml (range, 887 to 10,605 ng/ml) versus 199 ng/ml (range, 82 to 857 ng/ml) (geometric mean ratio [GMR], 13."( Plasma and intracellular (peripheral blood mononuclear cells) pharmacokinetics of once-daily raltegravir (800 milligrams) in HIV-infected patients.
Back, D; Barbanoj, MJ; Cedeño, S; Clotet, B; Egan, D; Liptrott, N; Miranda, C; Moltó, J; Valle, M; Watson, V, 2011
)
0.37
"Administration of the HIV PI ritonavir to wild type mice increased plasma triacylglycerols and cholesterol levels and this effect was exacerbated by dosing ritonavir to mice harbouring a disrupted FXR."( The bile acid sensor FXR protects against dyslipidemia and aortic plaques development induced by the HIV protease inhibitor ritonavir in mice.
Baldelli, F; Cipriani, S; Distrutti, E; Fiorucci, S; Francisci, D; Mencarelli, A; Palladino, G; Renga, B, 2010
)
0.86
" A month after the aripiprazole dosage was increased to 50 mg daily, the patient developed confusion and loss of coordination."( Increased aripiprazole concentrations in an HIV-positive male concurrently taking duloxetine, darunavir, and ritonavir.
Aung, GL; Kawamoto, LS; O'Brien, JG; Tien, PG, 2010
)
0.57
" However, ENF treatment is associated with injection site reactions and dosing fatigue."( A 48-week pilot study switching suppressed patients to darunavir/ritonavir and etravirine from enfuvirtide, protease inhibitor(s), and non-nucleoside reverse transcriptase inhibitor(s).
Alas, B; Perniciaro, A; Ruane, P; Ryan, R; Witek, J, 2010
)
0.6
" In turn, ART commonly requires complex dosing schedules and leads to the emergence of viral resistance and treatment failures."( Analyses of nanoformulated antiretroviral drug charge, size, shape and content for uptake, drug release and antiviral activities in human monocyte-derived macrophages.
Balkundi, S; Bronich, T; Gendelman, HE; Kabanov, AV; Kanmogne, G; Martinez-Skinner, A; McMillan, J; Mosley, RL; Nowacek, AS; Roy, U, 2011
)
0.37
" Rifabutin reduced the area under the plasma drug concentration-time curve from 0 to 12 h postdose (AUC(0-12)), maximum observed concentration of drug in plasma (C(max)), and minimum observed concentration of drug in plasma at the end of the dosing interval (C(min)) for saquinavir by 13%, 15%, and 9%, respectively, for subjects receiving rifabutin (150 mg) every 3 days with saquinavir-ritonavir BID."( Pharmacokinetic interaction study of ritonavir-boosted saquinavir in combination with rifabutin in healthy subjects.
Fettner, S; Rowell, L; Salgo, M; Zhang, X; Zwanziger, E, 2011
)
0.81
" Healthy volunteer studies have demonstrated a decrease in glucose disposal associated with dosing with specific antiretrovirals."( Effect of boosted fosamprenavir or lopinavir-based combinations on whole-body insulin sensitivity and lipids in treatment-naive HIV-type-1-positive men.
Back, DJ; Boffito, M; Jackson, AG; Mandalia, S; Moyle, GJ; Randell, PA; Taylor, J; Tjia, JF, 2010
)
0.36
" Seventeen healthy male volunteers received sequential dosing of the studied product: TAD (day 1) alone in a single dose for 7 days followed by TAD (day 8) in a single dose with TPV/r (500/200 mg twice daily, days 8-18)."( Effect of tipranavir/ritonavir combination on the pharmacokinetics of tadalafil in healthy volunteers.
Dellamonica, P; Durant, J; Ferrando, S; Garraffo, R; Lavrut, T; MacGregor, TR; Rouyrre, N; Sabo, JP, 2011
)
0.69
"Indinavir boosted with ritonavir (IDV/r) dosing with 400/100 mg, twice daily, is preferred in Thai adults, but this dose can lead to concentrations close to the boundaries of its therapeutic window."( Influence of body weight on achieving indinavir concentrations within its therapeutic window in HIV-infected Thai patients receiving indinavir boosted with ritonavir.
Bowonwatanuwong, C; Cressey, TR; Halue, G; Hirt, D; Jourdain, G; Lallemant, M; Leenasirimakul, P; Techapornroong, M; Treluyer, JM; Urien, S, 2011
)
0.88
" Co-administration with ATV/RTV resulted in increased plasma S/GSK1349572 area under the concentration-time curve during a dosing interval (AUC(0,τ)), observed maximal concentration (C(max) ), and concentration at the end of dosing interval at steady state (C(τ) ) by 62%, 34% and 121%, respectively."( Effect of atazanavir and atazanavir/ritonavir on the pharmacokinetics of the next-generation HIV integrase inhibitor, S/GSK1349572.
Borland, J; Chen, S; Lou, Y; Min, S; Peppercorn, A; Piscitelli, SC; Song, I; Wajima, T, 2011
)
0.64
"Nonlinear mixed-effects modeling was applied to explore the relationship between lopinavir and ritonavir concentrations over 72 h following drug cessation and also to assess other lopinavir and ritonavir dosing strategies compared to the standard 400-mg-100-mg twice-daily dose."( Sequential population pharmacokinetic modeling of lopinavir and ritonavir in healthy volunteers and assessment of different dosing strategies.
Aarons, L; Back, D; Boffito, M; Davies, G; Dickinson, L; Else, L; Khoo, S; Moyle, G; Pozniak, A; von Hentig, N, 2011
)
0.83
"ATV/RTV with dose-normalized EE/NGM resulted in geometric mean reductions of 16% in EE peak plasma concentration (C(max)), 19% in EE area under the concentration-time curve for a dosing interval (AUC([τ])) and 37% in EE lowest plasma concentration (C(min)), compared with EE 35 μg with NGM in the absence of ATV/RTV."( The effect of atazanavir/ritonavir on the pharmacokinetics of an oral contraceptive containing ethinyl estradiol and norgestimate in healthy women.
Bertz, R; Chung, E; Eley, T; Mahnke, L; Persson, A; Xu, X; Yones, C; Zhang, J, 2011
)
0.67
"Adequate plasma trough concentrations (Ctrough) of protease inhibitors are required to maintain antiviral activity throughout the dosing interval."( Therapeutic monitoring and variability of atazanavir in HIV-infected patients, with and without HCV coinfection, receiving boosted or unboosted regimens.
Barassi, A; Brandolini, M; Cusato, M; D'Arminio Monforte, A; Gulminetti, R; Maserati, R; Melzi D'Eril, GV; Regazzi, M; Sighinolfi, L; Tinelli, C; Villani, P, 2011
)
0.37
" We aimed at determining the optimal dosing regimen in neonates and infants weighing 1 to 10."( Lopinavir/ritonavir population pharmacokinetics in neonates and infants.
Anderson, ST; Blanche, S; Faye, A; Firtion, G; Giraud, C; Hirt, D; Khoo, S; Leprevost, M; Lyall, H; Peytavin, G; Solas, C; Thuret, I; Tréluyer, JM; Urien, S, 2011
)
0.77
" Based upon trough concentration limitations, suggested LPV/r dosing regimens were 40 mg 12 h(-1), 80 mg 12 h(-1) and 120 mg 12 h(-1) in the 1-2 kg, 2-6 kg and 6-10 kg group, respectively."( Lopinavir/ritonavir population pharmacokinetics in neonates and infants.
Anderson, ST; Blanche, S; Faye, A; Firtion, G; Giraud, C; Hirt, D; Khoo, S; Leprevost, M; Lyall, H; Peytavin, G; Solas, C; Thuret, I; Tréluyer, JM; Urien, S, 2011
)
0.77
" The purpose of this study was to assess the safety, efficacy and appropriate dosing regimen for ritonavir (RTV)-boosted atazanavir in HIV-1-infected pregnant women."( Safety and exposure of once-daily ritonavir-boosted atazanavir in HIV-infected pregnant women.
Bertz, R; Botes, M; Child, M; Conradie, F; Eley, T; Hu, W; Josipovic, D; McGrath, D; Osiyemi, O; Vandeloise, E; Wirtz, V; Zorrilla, C, 2011
)
0.87
"In this nonrandomized, open-label study, HIV-infected pregnant women were dosed with either 300/100 mg (n=20) or 400/100 mg (n=21) atazanavir/RTV once-daily (qd) in combination with zidovudine (300 mg) and lamivudine (150 mg) twice daily in the third trimester."( Safety and exposure of once-daily ritonavir-boosted atazanavir in HIV-infected pregnant women.
Bertz, R; Botes, M; Child, M; Conradie, F; Eley, T; Hu, W; Josipovic, D; McGrath, D; Osiyemi, O; Vandeloise, E; Wirtz, V; Zorrilla, C, 2011
)
0.65
" We proposed a systematic classification scheme using FDA-approved drug labeling to assess the DILI potential of drugs, which yielded a benchmark dataset with 287 drugs representing a wide range of therapeutic categories and daily dosage amounts."( FDA-approved drug labeling for the study of drug-induced liver injury.
Chen, M; Fang, H; Liu, Z; Shi, Q; Tong, W; Vijay, V, 2011
)
0.37
"To describe laboratory abnormalities among HIV-infected women and their infants with standard and increased lopinavir/ritonavir (LPV/r) dosing during the third trimester of pregnancy."( Lopinavir/ritonavir dosing during pregnancy in Brazil and maternal/infant laboratory abnormalities.
Ceriotto, M; João, EC; Kreitchmann, R; Melo, VH; Mussi-Pinhata, MM; Peixoto, MF; Pilotto, JH; Read, J; Souza, Rda S; Stoszek, SK,
)
0.74
"164 women received LPV/r standard dosing [(798/198 or 800/200 mg/day) (Group 1)] and 70 increased dosing [(> 800/200 mg/day) (Group 2)]."( Lopinavir/ritonavir dosing during pregnancy in Brazil and maternal/infant laboratory abnormalities.
Ceriotto, M; João, EC; Kreitchmann, R; Melo, VH; Mussi-Pinhata, MM; Peixoto, MF; Pilotto, JH; Read, J; Souza, Rda S; Stoszek, SK,
)
0.53
" Increased LPV/r dosing during the third trimester of pregnancy appears to be safe for HIV-infected women and their infants."( Lopinavir/ritonavir dosing during pregnancy in Brazil and maternal/infant laboratory abnormalities.
Ceriotto, M; João, EC; Kreitchmann, R; Melo, VH; Mussi-Pinhata, MM; Peixoto, MF; Pilotto, JH; Read, J; Souza, Rda S; Stoszek, SK,
)
0.53
"The object of this study was to investigate the pharmacokinetics of darunavir-ritonavir and atazanavir-ritonavir once-daily dosing over 72 h (h) following drug intake cessation."( Pharmacokinetics of once-daily darunavir-ritonavir and atazanavir-ritonavir over 72 hours following drug cessation.
Amara, A; Back, D; Boffito, M; Gazzard, B; Higgs, C; Jackson, A; Khoo, S; Moyle, G; Seymour, N, 2011
)
0.86
" Therapeutic drug monitoring (TDM) is occasionally used to guide chronic dosing to achieve target trough concentrations, but its clinical success assumes minimal intrasubject variability."( Within-patient atazanavir trough concentration monitoring in HIV-1-infected patients.
Crutchley, RD; Hochreitter, J; Ma, Q; Morse, GD; Sulaiman, A, 2011
)
0.37
" This study evaluates an intermittent dosing regimen for rifabutin when it is co-administered with ritonavir-boosted atazanavir."( Determination of rifabutin dosing regimen when administered in combination with ritonavir-boosted atazanavir.
Arikan, D; Bertz, R; Coumbis, J; Farajallah, A; Stonier, M; Wu, Y; Xu, X; Zhang, J; Zhu, L, 2011
)
0.81
" Lopinavir was active at levels well below those achieved with standard dosing of coformulated lopinavir-ritonavir."( In vitro activity of antiretroviral drugs against Plasmodium falciparum.
Nsanzabana, C; Rosenthal, PJ, 2011
)
0.58
"For use in chronic oral chemotherapeutic regimens, the potent anticancer drug docetaxel needs a solid oral dosage form."( Pharmaceutical development and preliminary clinical testing of an oral solid dispersion formulation of docetaxel (ModraDoc001).
Beijnen, JH; Huitema, AD; Koolen, SL; Moes, JJ; Nuijen, B; Schellens, JH, 2011
)
0.37
" Dosing was separated by a wash-out period of 14 days."( Bioequivalence study of two oral tablet formulations containing saquinavir mesylate boosted with ritonavir in healthy male subjects.
Feleder, EC; Halabe, EK; Yerino, GA; Zini, E, 2011
)
0.59
" Notably, this interaction was independent of the dosage of darunavir and not due to effects of raltegravir on the pharmacokinetics of ritonavir."( Co-administration of raltegravir reduces daily darunavir exposure in HIV-1 infected patients.
Baldelli, S; Boreggio, G; Cattaneo, D; Clementi, E; Cozzi, V; Fucile, S; Gervasoni, C; Landonio, S; Meraviglia, P; Rizzardini, G, 2012
)
0.58
" Blood samples were collected up to 24 hours after dosing on prespecified days."( Effect of vicriviroc with or without ritonavir on oral contraceptive pharmacokinetics: a randomized, open-label, parallel-group, fixed-sequence crossover trial in healthy women.
Kasserra, C; Li, J; March, B; O'Mara, E, 2011
)
0.64
"Atazanavir geometric mean (CV%) C(max), C(min) and AUC over the dosing interval were 2897 (46) ng/mL, 526 (57) ng/mL and 28 605 (46) ng · h/mL, respectively, and for lopinavir they were 10 655 (51) ng/mL, 5944 (68) ng/mL and 90 946 (59) ng · h/mL, respectively."( Pharmacokinetics and inhibitory quotient of atazanavir/ritonavir versus lopinavir/ritonavir in HIV-infected, treatment-naive patients who participated in the CASTLE Study.
Bertz, R; Child, M; Eley, T; Farajallah, A; Krystal, M; Liao, S; McGrath, D; Molina, JM; Persson, A; Sevinsky, H; Xu, X; Zhang, J; Zhu, L, 2012
)
0.63
"To describe the pharmacokinetics of maraviroc when dosed at 150 or 300 mg once daily with 800/100 mg of darunavir/ritonavir."( Once daily maraviroc 300 mg or 150 mg in combination with ritonavir-boosted darunavir 800/100 mg.
Ainsworth, J; Cook, R; Dufty, N; Gandhi, K; Hickinbottom, G; Khonyongwa, K; Okoli, C; Owen, A; Siccardi, M; Taylor, S; Thomas-William, S; Watson, J, 2012
)
0.83
" A population PK analysis was performed using NONMEM to characterize changes in lopinavir (LPV) PK relating to maturational changes in infants, and to assess dosing requirements in this population."( Assessment of lopinavir pharmacokinetics with respect to developmental changes in infants and the impact on weight band-based dosing.
Alvero, C; Capparelli, EV; Chadwick, EG; Hazra, R; Heckman, BE; Hughes, ML; Nikanjam, M; Palumbo, P; Pinto, J; Robbins, B; Yogev, R, 2012
)
0.38
"In this study, a slight but not significant decrease in the plasma lopinavir C(trough) was found during the third trimester of pregnancy, suggesting that standard dosing of the tablet formulation is also appropriate during the later stages of pregnancy."( Lopinavir/ritonavir trough concentrations with the tablet formulation in HIV-1-infected women during the third trimester of pregnancy.
Borderi, M; Calza, L; Colangeli, V; Grossi, G; Manfredi, R; Motta, R; Salvadori, C; Trapani, F; Viale, P, 2012
)
0.78
" However, some patients may have received atazanavir at a suboptimal dosage or had suboptimal susceptibility to BT agents."( Atazanavir: in pediatric patients with HIV-1 infection.
Deeks, ED, 2012
)
0.38
" Owing to the very large variability, trough drug levels might be very low under the standard dosing regimen, raising the question of a potential relevance of therapeutic drug monitoring of RAL in some situations."( Population pharmacokinetic analysis and pharmacogenetics of raltegravir in HIV-positive and healthy individuals.
Aouri, M; Arab-Alameddine, M; Boffito, M; Buclin, T; Cavassini, M; Csajka, C; Decosterd, LA; di Iulio, J; Fayet-Mello, A; Günthard, HF; Lubomirov, R; Neely, M; Owen, A; Rentsch, K; Rotger, M; Telenti, A, 2012
)
0.38
" However, although the levels of BILR 355 increased upon concomitant administration of RTV, a metabolite of BILR 355, BILR 516, which was not detected previously in humans dosed with BILR 355 alone, became a disproportionate human metabolite with levels exceeding the parent levels at steady state."( Metabolic switching of BILR 355 in the presence of ritonavir. II. Uncovering novel contributions by gut bacteria and aldehyde oxidase.
Lai, WG; Li, Y; Tweedie, DJ; Whitcher-Johnstone, A; Xu, J, 2012
)
0.63
" We also collected data on dosing of atazanavir, and on demographic (age, gender, and ethnicity), physiological (weight and body mass index), and clinical parameters (CD4+ cell count, HIV-RNA viremia, co-medication, and hepatitis C co-infection)."( Effects of hepatitis C virus infection on the pharmacokinetics of ritonavir-boosted atazanavir in HIV-1-infected patients.
Cenderello, G; Di Biagio, A; Loregian, A; Pagni, S; Palù, G; Rosso, R; Sormani, MP; Viscoli, C, 2012
)
0.62
"19) for the concentration at the end of the dosing interval."( Effect of milk thistle on the pharmacokinetics of darunavir-ritonavir in HIV-infected patients.
Cedeño, S; Clotet, B; Miranda, C; Moltó, J; Negredo, E; Valle, M, 2012
)
0.62
" Intensive pharmacokinetic sampling was performed 7 days apart after LPV/r dosing under moderate fat, high fat, and fasted meal conditions."( Steady-state pharmacokinetics of lopinavir plus ritonavir when administered under different meal conditions in HIV-infected Ugandan adults.
Back, D; Boffito, M; Byakika-Kibwika, P; Khoo, S; Lamorde, M; Mayito, J; Merry, C; Nabukeera, L; Ogwal-Okeng, J; Ryan, M; Tjia, J, 2012
)
0.63
" Observed ART dosing took place for at least 2 weeks."( Untreated HIV infection is associated with higher blood alcohol levels.
Beatty, G; Gruber, VA; Lum, PJ; McCance-Katz, EF; Peters, M; Rainey, PM, 2012
)
0.38
" Repeated dosing of LB42908 in rats did not significantly affect its own metabolism, indicating that long-term administration of LB42908 would not alter its pharmacokinetic profiles."( Preclinical metabolism of LB42908, a novel farnesyl transferase inhibitor, and its effects on the cytochrome P450 isozyme activities.
Aeri, K; Chang, M; Kim, HJ; Koh, JS; Lee, SH, 2012
)
0.38
"This is the first study to compare darunavir CSF concentrations in patients taking the once-daily or the twice-daily dosage: our data show that darunavir and ritonavir dosing significantly affects not only CSF concentrations but also the extent of drug penetration into the CSF."( Determinants of darunavir cerebrospinal fluid concentrations: impact of once-daily dosing and pharmacogenetics.
Bertucci, R; Bonora, S; Calcagno, A; Cusato, J; D'Avolio, A; Di Perri, G; Marinaro, L; Siccardi, M; Simiele, M; Yilmaz, A, 2012
)
0.58
"003) from their corresponding baseline value during the 100 mg dosing period; there were no significant changes on 50 mg."( Ritonavir boosting dose reduction from 100 to 50 mg does not change the atazanavir steady-state exposure in healthy volunteers.
Antonijoan, RM; Barbanoj, MJ; Cedeño, S; Clotet, B; Domingo, P; Estévez, JA; Mangues, MA; Moltó, J; Puntes, M; Tuneu, L; Valle, M, 2012
)
1.82
" Clinical studies in HCV patients have shown a potential need for a high danoprevir daily dose and/or dosing frequency."( Impact of low-dose ritonavir on danoprevir pharmacokinetics: results of computer-based simulations and a clinical drug-drug interaction study.
Blotner, S; Chen, Y; Fretland, J; Haznedar, JO; Reddy, MB; Smith, P; Tran, JQ, 2012
)
0.71
" Following results from this drug-drug interaction (DDI) model, a crossover study was performed in healthy volunteers to investigate the effects of acute and repeat dosing of low-dose ritonavir on danoprevir single-dose pharmacokinetics."( Impact of low-dose ritonavir on danoprevir pharmacokinetics: results of computer-based simulations and a clinical drug-drug interaction study.
Blotner, S; Chen, Y; Fretland, J; Haznedar, JO; Reddy, MB; Smith, P; Tran, JQ, 2012
)
0.9
" Prospective pharmacokinetic studies to devise a rational dosing strategy for vinblastine in patients receiving ritonavir/lopinavir are warranted."( Incidence, predictors and significance of severe toxicity in patients with human immunodeficiency virus-associated Hodgkin lymphoma.
Boro, J; Cheung, MC; Ezzat, HM; Harris, M; Hicks, LK; Leitch, HA; Lima, VD; Montaner, JS, 2012
)
0.59
" Factors that may have contributed to the suboptimal results with nevirapine include elevated viral load at baseline, selection for nevirapine resistance, background regimen of nucleoside reverse-transcriptase inhibitors, and the standard ramp-up dosing strategy."( Nevirapine versus ritonavir-boosted lopinavir for HIV-infected children.
Abrams, EJ; Barlow-Mosha, L; Bobat, R; Chi, BH; Cotton, MF; Eshleman, SH; Hughes, MD; Jean-Philippe, P; Kamthunzi, P; Khadse, S; Lindsey, JC; Millar, L; Mofenson, LM; Moultrie, H; Mujuru, HA; Palumbo, P; Petzold, E; Purdue, L; Schimana, W; Violari, A, 2012
)
0.71
" To identify risk factors for nonoptimal anticoagulation and to determine if warfarin dosing is differentially affected by specific antiretroviral agents."( Warfarin therapy in the HIV medical home model: low rates of therapeutic anticoagulation despite adherence and differences in dosing based on specific antiretrovirals.
Anderson, AM; Chane, T; Chen, S; Easley, KA; Patel, M; Xue, W, 2012
)
0.38
" However, there are challenges to initiate ART in early life, including the possibility of drug resistance in the context of prevention of mother-to-child transmission (PMTCT) programs, a paucity of drug choices , uncertain dosing for some medications and long-term toxicities."( Effectiveness of antiretroviral therapy in HIV-infected children under 2 years of age.
Cotton, M; Gibb, D; Penazzato, M; Prendergast, A; Tierney, J, 2012
)
0.38
" Lopinavir AUC(0-24) following twice-daily and once-daily dosing was 169."( Once- versus twice-daily lopinavir/ritonavir tablets in virologically suppressed, HIV-infected, treatment-experienced children: comparative pharmacokinetics and virological outcome after switching to once-daily lopinavir/ritonavir.
Chokephaibulkit, K; Cressey, TR; Lapphra, K; Nuntarukchaikul, M; Phongsamart, W; Vanprapar, N; Wittawatmongkol, O, 2012
)
0.66
" Further efficacy studies of lopinavir/ritonavir once daily in children are necessary before routinely recommending this dosing strategy."( Once- versus twice-daily lopinavir/ritonavir tablets in virologically suppressed, HIV-infected, treatment-experienced children: comparative pharmacokinetics and virological outcome after switching to once-daily lopinavir/ritonavir.
Chokephaibulkit, K; Cressey, TR; Lapphra, K; Nuntarukchaikul, M; Phongsamart, W; Vanprapar, N; Wittawatmongkol, O, 2012
)
0.93
"1 for the concentration prior to the next dose (C(12)) with no differences between dosing groups."( Pharmacokinetics of darunavir/ritonavir in Asian HIV-1-infected children aged ≥7 years.
Ananworanich, J; Burger, D; Chokephaibulkit, K; Gorowara, M; Kerr, SJ; Pasomsap, C; Phongsamart, W; Prasitsuebsai, W; Puthanakit, T; Sekar, V; Sophonphan, J; Suwanlerk, T; Vanprapar, N; Wittawatmongkol, O, 2012
)
0.67
"Thai children aged ≥7 years who were on standard darunavir dosing with 100 mg ritonavir boosting had adequate and comparable darunavir AUC(0-12), C(max) and C(12) to non-Asian children who mainly used lower doses of ritonavir boosting."( Pharmacokinetics of darunavir/ritonavir in Asian HIV-1-infected children aged ≥7 years.
Ananworanich, J; Burger, D; Chokephaibulkit, K; Gorowara, M; Kerr, SJ; Pasomsap, C; Phongsamart, W; Prasitsuebsai, W; Puthanakit, T; Sekar, V; Sophonphan, J; Suwanlerk, T; Vanprapar, N; Wittawatmongkol, O, 2012
)
0.9
" This suggests that 300/50 mg and 200/100 mg dosing are preferred candidate regimens in future clinical studies."( Simultaneous population pharmacokinetic modelling of atazanavir and ritonavir in HIV-infected adults and assessment of different dose reduction strategies.
Austin, R; Back, D; Boffito, M; Davies, G; Dickinson, L; Khoo, S; Owen, A; Schipani, A, 2013
)
0.63
"In HIV-infected patients receiving rifampicin-based treatment for tuberculosis (TB), the dosage of lopinavir/ritonavir (LPV/r) is adjusted to prevent sub-therapeutic lopinavir concentrations."( Coadministration of lopinavir/ritonavir and rifampicin in HIV and tuberculosis co-infected adults in South Africa.
Gandhi, RT; Kuritzkes, DR; Marconi, VC; Murphy, RA; Sunpath, H, 2012
)
0.88
" The durability of coadministered treatment was significantly shorter in patients who received super-boosted lopinavir/ritonavir with TB treatment compared to patients who received standard lopinavir/ritonavir dosing (log rank, P = 0."( Coadministration of lopinavir/ritonavir and rifampicin in HIV and tuberculosis co-infected adults in South Africa.
Gandhi, RT; Kuritzkes, DR; Marconi, VC; Murphy, RA; Sunpath, H, 2012
)
0.88
" Use of exact dosing data halved unexplained variability in ATV clearance."( Effect of adherence as measured by MEMS, ritonavir boosting, and CYP3A5 genotype on atazanavir pharmacokinetics in treatment-naive HIV-infected patients.
Barrail-Tran, A; Descamps, D; Duval, X; Goujard, C; Mentré, F; Nembot, G; Panhard, X; Savic, RM; Taburet, AM; Verstuyft, C; Vrijens, B, 2012
)
0.64
"Uncertainty surrounds the correct dosing of lopinavir/r (LPV/r) in HIV-infected children not receiving non-nucleoside reverse transcriptase inhibitors."( Lopinavir dosing in HIV-infected children in the United Kingdom and Ireland.
Butler, K; Doerholt, K; Donegan, K; Gibb, DM; Judd, A; Lyall, H; Menson, E; Riordan, A; Shingadia, D; Tookey, P; Tudor-Williams, G; Walker, AS, 2013
)
0.39
" Results highlight the importance of optimizing dosing in HIV-infected children of all ages."( Lopinavir dosing in HIV-infected children in the United Kingdom and Ireland.
Butler, K; Doerholt, K; Donegan, K; Gibb, DM; Judd, A; Lyall, H; Menson, E; Riordan, A; Shingadia, D; Tookey, P; Tudor-Williams, G; Walker, AS, 2013
)
0.39
" This therapeutic principle called 'boosting' helped to overcome the obstacles of this class of drugs concerning unfavorable pharmacokinetics, resulting in either a high frequency of dosing or subtherapeutic plasma concentrations."( Safety of pharmacoenhancers for HIV therapy.
Haberl, A; von Hentig, N, 2012
)
0.38
" Coadministration with boceprevir decreased RTV AUC during a dosing interval τ (AUC(τ)) by 22%-36%."( Pharmacokinetic interactions between the hepatitis C virus protease inhibitor boceprevir and ritonavir-boosted HIV-1 protease inhibitors atazanavir, darunavir, and lopinavir.
Butterton, JR; Feng, HP; Hughes, EA; Hulskotte, EG; O'Mara, E; Treitel, MA; van Zutven, MG; Wagner, JA; Xuan, F; Youngberg, SP, 2013
)
0.61
" Blood sampling at the end of the dosing interval (Ctrough) was performed."( Intracellular accumulation of ritonavir combined with different protease inhibitors and correlations between concentrations in plasma and peripheral blood mononuclear cells.
Agati, S; Baietto, L; Bonora, S; Calcagno, A; Cusato, J; D'Avolio, A; Di Perri, G; Larovere, G; Sciandra, M; Siccardi, M; Simiele, M; Tettoni, M; Trentini, L, 2013
)
0.68
"A pretransplantation pharmacokinetic model of tacrolimus in these patients was developed, and a posttransplantation dosing advice was established for each individual patient."( Pretransplantation pharmacokinetic curves of tacrolimus in HIV-infected patients on ritonavir-containing cART: a pilot study.
Crommelin, HA; Mudrikova, T; van den Broek, MP; van Maarseveen, EM; van Zuilen, AD, 2013
)
0.61
" Pharmacokinetic data from drug-development studies in rats often determine the dosage used in human clinical trials."( Three-dimensional quantitative structure-activity relationship analysis of inhibitors of human and rat cytochrome P4503A enzymes.
Gouda, H; Handa, K; Hirono, S; Nakagome, I; Yamaotsu, N, 2013
)
0.39
" Micro/small dosing is useful for examining the mechanism of drug interactions without safety concern."( Mechanisms of pharmacokinetic enhancement between ritonavir and saquinavir; micro/small dosing tests using midazolam (CYP3A4), fexofenadine (p-glycoprotein), and pravastatin (OATP1B1) as probe drugs.
Ando, Y; Deguchi, M; Hirota, T; Ieiri, I; Irie, S; Izumi, N; Kanda, E; Kimura, M; Kotani, N; Kusuhara, H; Maeda, K; Matsuguma, K; Matsuki, S; Morishita, M; Okuzono, T; Sugiyama, Y; Tsunemitsu, S; Yamane, N, 2013
)
0.64
" Blood plasma (for TFV, FTC, EFV, ATV and RTV concentrations) and peripheral blood mononuclear cells [PBMCs; for tenofovir diphosphate (TFV-DP) and emtricitabine triphosphate (FTC-TP) concentrations] were collected at 11 time-points over a 24-hour dosing interval."( Pharmacokinetics of two common antiretroviral regimens in older HIV-infected patients: a pilot study.
Adams, JL; Corbett, AH; Dumond, JB; Forrest, A; Jennings, SH; Kashuba, AD; Kendrick, RL; Malone, S; Patterson, KB; Prince, HM; Sykes, C; Wang, R; White, N, 2013
)
0.39
" Patients received ABT-450/r 150/100 mg once daily and ABT-072 400 mg once daily with weight-based ribavirin 1000-1200 mg/day dosed twice daily for 12 weeks."( A phase 2a trial of 12-week interferon-free therapy with two direct-acting antivirals (ABT-450/r, ABT-072) and ribavirin in IL28B C/C patients with chronic hepatitis C genotype 1.
Bernstein, B; Cohen, DE; Koev, G; Kowdley, KV; Larsen, L; Lawitz, E; Menon, R; Pilot-Matias, T; Podsadecki, T; Poordad, F; Siggelkow, S; Tripathi, R, 2013
)
0.39
" The MEMS-defined adherence for correct dosing (-0."( Adherence profiles and therapeutic responses of treatment-naive HIV-infected patients starting boosted atazanavir-based therapy in the ANRS 134-COPHAR 3 trial.
Barrail-Tran, A; Descamps, D; Duval, X; Goujard, C; Mentré, F; Nembot, G; Panhard, X; Parienti, JJ; Taburet, AM; Vigan, M; Vrijens, B, 2013
)
0.39
" Lopinavir plasma exposure as estimated by the area under the concentration-time curve over the 12-h dosing interval (AUC(0-12h), AUCτ) was determined on day 14 and again on day 21."( Co-administration of a commonly used Zimbabwean herbal treatment (African potato) does not alter the pharmacokinetics of lopinavir/ritonavir.
Aweeka, F; Greenblatt, R; Guglielmo, BJ; Gwaza, L; Huang, L; Lizak, P, 2013
)
0.59
" We assessed the changes in total, conjugated, and unconjugated bilirubin and the effect on ATV pharmacokinetics (PK) after single and 14-day dosing of ZnSO(4)."( Coadministration of atazanavir-ritonavir and zinc sulfate: impact on hyperbilirubinemia and pharmacokinetics.
Back, D; Boffito, M; Else, L; Gazzard, B; Jackson, A; Karolia, Z; Moyle, G; Ringner-Nackter, L; Seymour, N; Yapa, MH, 2013
)
0.68
"Results from intensive pharmacokinetic sampling of subjects enrolled in single dose visits was used to determine individualized dosing for the first 6-10 subjects in each of 2 cohorts (4 weeks to <6 months, 6 months to <2 years); steady state pharmacokinetic data were then used to select the dosage regimen for the remaining subjects recruited to the cohort."( Pharmacokinetics, safety and antiviral activity of fosamprenavir/ritonavir-containing regimens in HIV-infected children aged 4 weeks to 2 years-48-week study data.
Cassim, H; Cheng, K; Cotton, M; Ford, SL; Garges, HP; Givens, N; Lou, Y; Pavía-Ruz, N; Perger, T; Ross, LL; Sievers, J; Wire, MB, 2014
)
0.64
"Final FPV/RTV dosing regimens achieved plasma amprenavir exposures comparable with those from regimens approved in adults, with the exception of trough exposures in the <6-month-old infants."( Pharmacokinetics, safety and antiviral activity of fosamprenavir/ritonavir-containing regimens in HIV-infected children aged 4 weeks to 2 years-48-week study data.
Cassim, H; Cheng, K; Cotton, M; Ford, SL; Garges, HP; Givens, N; Lou, Y; Pavía-Ruz, N; Perger, T; Ross, LL; Sievers, J; Wire, MB, 2014
)
0.64
"A total of 54 patients infected with hepatitis C virus genotype 1 received an oral drug cocktail (2 mg midazolam, 10 mg warfarin and 10 mg vitamin K) before and after 14 days of dosing with either danoprevir/r or placebo plus low-dose ritonavir (placebo/r)."( A randomised study of the effect of danoprevir/ritonavir or ritonavir on substrates of cytochrome P450 (CYP) 3A and 2C9 in chronic hepatitis C patients using a drug cocktail.
Brennan, BJ; Chang, L; Giraudon, M; Kulkarni, R; Morcos, PN; Shulman, N; Smith, PF; Tran, JQ, 2013
)
0.83
" Blood samples were withdrawn from retro-orbital plexus on first day in single dose PK study (SDS) and on 15th day in multiple dosing PK study (MDS)."( Enhanced oral bioavailability of felodipine by naringenin in Wistar rats and inhibition of P-glycoprotein in everted rat gut sacs in vitro.
Naveen Babu, K; Puneeth, Y; Ravindra Babu, P; Sridhar, V; Surya Sandeep, M, 2014
)
0.4
" No dosage adjustment is needed for MMT when coadministered with DNVr."( Effect of ritonavir-boosted danoprevir, a potent hepatitis C virus protease inhibitor, on the pharmacokinetics of methadone in healthy subjects undergoing methadone maintenance therapy.
Bech, N; Brennan, BJ; Morcos, PN; Moreira, SA; Navarro, MT; Smith, PF, 2014
)
0.8
" No clinically relevant interactions between daclatasvir and tenofovir disoproxil fumarate were observed for either drug, and no dosing adjustments were indicated."( Assessment of pharmacokinetic interactions of the HCV NS5A replication complex inhibitor daclatasvir with antiretroviral agents: ritonavir-boosted atazanavir, efavirenz and tenofovir.
Bertz, R; Bifano, M; Grasela, D; Hartstra, J; Hwang, C; Kandoussi, H; Oosterhuis, B; Sevinsky, H; Tiessen, R; Velinova-Donga, M, 2013
)
0.59
" Pharmacokinetic parameters, assessed on day 5 of period 1 and on days 10 and 20 of period 2, included the maximum plasma concentration (Cmax), the concentration at end of dosing interval (Cτ), and the area under the curve over dosing interval (AUCτ)."( Pharmacokinetic interaction between maraviroc and fosamprenavir-ritonavir: an open-label, fixed-sequence study in healthy subjects.
Fang, A; Heera, J; Mendes da Costa, L; Plotka, A; Vourvahis, M, 2013
)
0.63
"In this study, treatment-naive HIV-1 patients (aged 18-60 years) received 1 of the 4 dosing regimens of TMC310911: 150 mg twice-daily (bid) (n = 8), 300 mg bid (n = 8), 75 mg bid (n = 9), or 300 mg once-daily (qd) (n = 8), for 14 days, all coadministered with 100 mg of ritonavir, as only antiretroviral therapy."( Antiviral activity, pharmacokinetics, and safety of the HIV-1 protease inhibitor TMC310911, coadministered with ritonavir, in treatment-naive HIV-1-infected patients.
Arastéh, K; Dierynck, I; Hoetelmans, RM; Jacquemyn, B; Mariën, K; Meyvisch, P; Schürmann, D; Simmen, K; Smyej, I; Stellbrink, HJ; Stephan, C; Truyers, C; Verloes, R, 2014
)
0.79
" Pharmacokinetic studies suggest increasing the dosage of lopinavir/ritonavir (LPV/r) in pregnancy."( A randomized controlled trial to assess safety, tolerability, and antepartum viral load with increased lopinavir/ritonavir dosage in pregnancy.
Bonafe, SM; Castelo, A; Costa, DA; Machado, RH; Pott-Junior, H; Senise, JF; Vaz, MJ, 2013
)
0.84
"06 μg/ml), suggesting that the RBT dosage may be inadequate."( Pharmacokinetics of rifabutin during atazanavir/ritonavir co-administration in HIV-infected TB patients in India.
Bhavani, PK; Chandrasekaran, C; Hemanth Kumar, AK; Raja, K; Ramachandran, G; Srinivasan, R; Sudha, V; Swaminathan, S; Venkatesh, S, 2013
)
0.65
" The molecular method of response determination by 3H-TDR incorporation and ritonavir dose-energy model are reliable to avoid chemo-resistance by identifying the optimal dosing regimens and schedules prior therapy allowing the use of much lower dose of ritonavir and thus decreases the drug side effects and risks of relapse."( Identifying the optimal dose of ritonavir in the treatment of malignancies.
Moawad, EY, 2014
)
0.92
"No major changes were identified in the group treated with the lowest dosing compared with the control."( Effects of combined zidovudine/lopinavir/ritonavir therapy during rat pregnancy: morphological aspects.
de Carvalho, LP; Kulay, L; Nakamura, MU; Oliveira-Filho, RM; Simões, RS; Tavella, JS; Wagner, A, 2013
)
0.66
" This comprehensive modelling and simulation approach could be used as a surrogate assessment of antiretroviral (ARV) activity where adequate early-phase dose-ranging studies are lacking in order to define target trough concentrations and possibly refine dosing recommendations."( Integrated population pharmacokinetic/viral dynamic modelling of lopinavir/ritonavir in HIV-1 treatment-naïve patients.
Acosta, EP; D'Argenio, DZ; Delille, C; Kerstner-Wood, C; Lennox, JL; Ofotokun, I; Sheth, AN; Wang, K, 2014
)
0.63
"Our model predicts that even with maximal clinical dosing regimens of epidural fentanyl over 24 h, ritonavir-induced cytochrome P450 3A4 inhibition is unlikely to produce plasma fentanyl concentrations associated with a decrease in minute ventilation."( Effect of ritonavir-induced cytochrome P450 3A4 inhibition on plasma fentanyl concentrations during patient-controlled epidural labor analgesia: a pharmacokinetic simulation.
Avram, MJ; Cambic, CR; Gupta, DK; Wong, CA, 2014
)
1.02
" Dosing is based on body weight bands or body surface area under FDA approval and only body surface area by the EMA."( Pharmacokinetics of pediatric lopinavir/ritonavir tablets in children when administered twice daily according to FDA weight bands.
Bastiaans, DE; Burger, DM; Chalermpantmetagul, S; Colbers, AP; Compagnucci, A; Cressey, TR; Forcat, S; Giaquinto, C; Hansudewechakul, R; Harper, LM; Inshaw, JR; Kanjanavanit, S; Königs, C; Lyall, H; Noguera-Julian, A; Saïdi, Y, 2014
)
0.67
"FDA weight band-based dosing recommendations provide adequate exposure to LPV when using LPV/r pediatric tablets."( Pharmacokinetics of pediatric lopinavir/ritonavir tablets in children when administered twice daily according to FDA weight bands.
Bastiaans, DE; Burger, DM; Chalermpantmetagul, S; Colbers, AP; Compagnucci, A; Cressey, TR; Forcat, S; Giaquinto, C; Hansudewechakul, R; Harper, LM; Inshaw, JR; Kanjanavanit, S; Königs, C; Lyall, H; Noguera-Julian, A; Saïdi, Y, 2014
)
0.67
"The use of rivaroxaban in fixed dosing regimens without need for routine coagulation monitoring may lead to the misconception that there is a minimal risk of drug-drug interactions."( Gastrointestinal bleeding associated with rivaroxaban administration in a treated patient infected with human immunodeficiency virus.
Battegay, M; Elzi, L; Lakatos, B; Marzolini, C; Stoeckle, M, 2014
)
0.4
" With both dosing regimens, 2 - 5 fold increases of the 25-O-desacetyl- rifabutin metabolite were observed when rifabutin was given with lopinavir/ritonavir compared with rifabutin alone."( Randomised pharmacokinetic trial of rifabutin with lopinavir/ritonavir-antiretroviral therapy in patients with HIV-associated tuberculosis in Vietnam.
Barrail-Tran, A; Borand, L; Connolly, C; Duc, NH; Dung, NH; Harries, AD; Lagarde, D; Lan, NN; Lan, NT; Laureillard, D; Lien, TT; Lienhardt, C; Pym, A; Quillet, C; Taburet, AM; Thu, NT, 2014
)
0.84
" Patients in treatment arm 1 (non-ritonavir HAART) received standard sunitinib dosing (50 mg/day)."( A phase 1/pharmacokinetic study of sunitinib in combination with highly active antiretroviral therapy in human immunodeficiency virus-positive patients with cancer: AIDS Malignancy Consortium trial AMC 061.
Aboulafia, D; Cianfrocca, ME; Deeken, JF; Dezube, BJ; Dowlati, A; Gerecitano, J; Haigentz, M; Henry, DH; Ivy, SP; Little, RF; Mitsuyasu, RT; Moore, PC; Ratner, L; Rudek, MA; Sullivan, R, 2014
)
0.68
"Patients receiving non-ritonavir-based HAART regimens tolerated standard dosing of sunitinib."( A phase 1/pharmacokinetic study of sunitinib in combination with highly active antiretroviral therapy in human immunodeficiency virus-positive patients with cancer: AIDS Malignancy Consortium trial AMC 061.
Aboulafia, D; Cianfrocca, ME; Deeken, JF; Dezube, BJ; Dowlati, A; Gerecitano, J; Haigentz, M; Henry, DH; Ivy, SP; Little, RF; Mitsuyasu, RT; Moore, PC; Ratner, L; Rudek, MA; Sullivan, R, 2014
)
0.71
" LPV concentrations were also monitored on a routine basis, after 2 weeks of treatment initiation, between 1 and 24 hours after dosing in all children."( Concentration-response model of lopinavir/ritonavir in HIV-1-infected pediatric patients.
Benaboud, S; Blanche, S; Bouazza, N; Foissac, F; Frange, P; Hirt, D; Tréluyer, JM; Urien, S, 2014
)
0.67
"The CLPV90 derived from this model supports current dosing guidelines."( Concentration-response model of lopinavir/ritonavir in HIV-1-infected pediatric patients.
Benaboud, S; Blanche, S; Bouazza, N; Foissac, F; Frange, P; Hirt, D; Tréluyer, JM; Urien, S, 2014
)
0.67
" As a stand-alone agent, elvitegravir will require twice-daily dosing to achieve effective viral reductions."( Elvitegravir for the treatment of HIV infection.
Reviriego, C, 2014
)
0.4
" It is prescribed at standard dosage regimens of 600/100 mg twice daily in treatment-experienced patients and 800/100 mg once daily in naive patients."( Population pharmacokinetic modelling and evaluation of different dosage regimens for darunavir and ritonavir in HIV-infected individuals.
Aouri, M; Arab-Alameddine, M; Buclin, T; Cavassini, M; Csajka, C; Décosterd, LA; Fayet-Mello, A; Gatri, M; Guidi, M; Ledergerber, B; Lubomirov, R; Panchaud, A; Rentsch, K; Rotger, M; Telenti, A; Widmer, N, 2014
)
0.62
" Finally, model-based simulations were performed to compare trough concentrations (Cmin) between the recommended dosage regimen and alternative combinations of darunavir and ritonavir."( Population pharmacokinetic modelling and evaluation of different dosage regimens for darunavir and ritonavir in HIV-infected individuals.
Aouri, M; Arab-Alameddine, M; Buclin, T; Cavassini, M; Csajka, C; Décosterd, LA; Fayet-Mello, A; Gatri, M; Guidi, M; Ledergerber, B; Lubomirov, R; Panchaud, A; Rentsch, K; Rotger, M; Telenti, A; Widmer, N, 2014
)
0.81
" The simulations predicted darunavir Cmin much higher than the IC50 thresholds for wild-type and protease inhibitor-resistant HIV-1 strains (55 and 550 ng/mL, respectively) under standard dosing in >98% of experienced and naive patients."( Population pharmacokinetic modelling and evaluation of different dosage regimens for darunavir and ritonavir in HIV-infected individuals.
Aouri, M; Arab-Alameddine, M; Buclin, T; Cavassini, M; Csajka, C; Décosterd, LA; Fayet-Mello, A; Gatri, M; Guidi, M; Ledergerber, B; Lubomirov, R; Panchaud, A; Rentsch, K; Rotger, M; Telenti, A; Widmer, N, 2014
)
0.62
" the area under the plasma concentration-time curve over a dosing interval at steady state (AUC(0-τ))] and for peak exposure [i."( Steady-state pharmacokinetics of darunavir/ritonavir and pitavastatin when co-administered to healthy adult volunteers.
Campbell, SE; Medlock, MM; Morgan, RE; Small, DS; Sponseller, CA; Yu, CY, 2014
)
0.67
" Twelve-hour intensive pharmacokinetic sampling after observed LPV/r intake (plus 2 nucleoside reverse transcriptase inhibitors) following World Health Organization 2010 dosing with food was performed 4 weeks after enrollment."( The pharmacokinetics and acceptability of lopinavir/ritonavir minitab sprinkles, tablets, and syrups in african HIV-infected children.
Burger, D; Fillekes, Q; Gibb, DM; Keishanyu, R; Kekitiinwa, A; Kendall, L; Lallemant, M; Musiime, V; Namuddu, R; Opilo, W; Walker, AS; Young, N, 2014
)
0.65
" However, there are challenges to initiation of ART in early life, including the possibility of drug resistance in the context of prevention of mother-to-child transmission (PMTCT) programs, a paucity of drug choices , uncertain dosing for some medications and long-term toxicities."( Optimisation of antiretroviral therapy in HIV-infected children under 3 years of age.
Abrams, E; Muhe, LM; Penazzato, M; Prendergast, AJ; Tindyebwa, D, 2014
)
0.4
" In South Africa, clinicians are advised to use 'double-dose' LPV/r dosed at 800 mg/200 mg twice daily during anti-tuberculosis treatment."( Double-dose lopinavir-ritonavir in combination with rifampicin-based anti-tuberculosis treatment in South Africa.
Chelin, N; Cohen, S; Gandhi, RT; Murphy, RA; Sunpath, H; Tennant, I; Winternheimer, P, 2014
)
0.72
"Patients with HIV on antiretroviral therapy might benefit from regimen simplification to reduce pill burden and dosing frequency."( Simplification to coformulated elvitegravir, cobicistat, emtricitabine, and tenofovir versus continuation of ritonavir-boosted protease inhibitor with emtricitabine and tenofovir in adults with virologically suppressed HIV (STRATEGY-PI): 48 week results o
Arribas, JR; Di Perri, G; Ebrahimi, R; Gathe, J; Nguyen, T; Pialoux, G; Piontkowsky, D; Reynes, J; Tebas, P; White, K, 2014
)
0.61
" We therefore propose a 50% dosage increase of olanzapine when combining with a ritonavir-boosted protease inhibitor."( Effect of fosamprenavir/ritonavir on the pharmacokinetics of single-dose olanzapine in healthy volunteers.
Burger, DM; Colbers, AP; Jacobs, BS; Schouwenberg, BJ; Velthoven-Graafland, K, 2014
)
0.94
" Pharmacokinetic sampling occurred at the end of each dosing period."( Phase I safety, pharmacokinetics, and pharmacogenetics study of the antituberculosis drug PA-824 with concomitant lopinavir-ritonavir, efavirenz, or rifampin.
Allen, R; Aweeka, F; Bao, J; Cramer, Y; Dooley, KE; Haas, DW; Koletar, SL; Luetkemeyer, AF; Marzan, F; Murray, S; Park, JG; Savic, R; Sutherland, D, 2014
)
0.61
"Atazanavir/ritonavir 200/100 mg dosing provided adequate ATV AUC 0-24 when used with TDF in HIV-infected Thai children weighing between 25 and 50 kg."( Pharmacokinetics of atazanavir/ritonavir among HIV-infected Thai children concomitantly taking tenofovir disoproxil fumarate.
Ananworanich, J; Bunupuradah, T; Keadpudsa, S; Prasitsuebsai, W; Puthanakit, T; Sahakijpicharn, T; Srimuan, A; Techasaensiri, C; Thammajaruk, N, 2014
)
1.08
" Blood sampling was performed at the end of the dosing interval (Ctrough)."( Intracellular accumulation of atazanavir/ritonavir according to plasma concentrations and OATP1B1, ABCB1 and PXR genetic polymorphisms.
Allegra, S; Bonora, S; Calcagno, A; Carcieri, C; Cusato, J; D'Avolio, A; Di Perri, G; Sciandra, M; Simiele, M; Trentini, L, 2014
)
0.67
" Efficacious, safe bedaquiline dosing for MDR-TB patients receiving antiretrovirals is important."( Impact of lopinavir-ritonavir or nevirapine on bedaquiline exposures and potential implications for patients with tuberculosis-HIV coinfection.
Dooley, KE; Karlsson, MO; Svensson, EM, 2014
)
0.73
" Fosamprenavir-ritonavir decreased the DTG area under the concentration-time curve, maximum concentration in plasma, and concentration in plasma at the end of the dosing interval by 35%, 24%, and 49%, respectively."( Effect of fosamprenavir-ritonavir on the pharmacokinetics of dolutegravir in healthy subjects.
Borland, J; Chen, S; Peppercorn, A; Piscitelli, SC; Song, I; Wajima, T, 2014
)
1.06
" However, because the PIs are metabolized by cytochrome P450 (CYP) 3A enzymes, intentional inhibition of these enzymes leads to higher drug exposure, lower pill burden, and therefore simplified dosing schedules with this class of drug."( Pharmacokinetic enhancers in HIV therapeutics.
Acosta, EP; Delille, C; Larson, KB; Otofokun, I; Wang, K, 2014
)
0.4
" Most patients (70%) received a 400 mg ATV dosing per day, once (26%) or twice daily (44%)."( Efficacy and safety of switching to raltegravir plus atazanavir dual therapy in pretreated HIV-1-infected patients over 144 weeks: a cohort study.
Batard, ML; Bernard-Henry, C; Cheneau, C; De Mautort, E; Fafi-Kremer, S; Gantner, P; Koeppel, C; Muret, P; Partisani, M; Priester, M; Rey, D; Sueur, C, 2014
)
0.4
"Standard dosing provides adequate lopinavir trough concentrations for the majority of pregnant women but increased doses may be preferable for women weighing >100 kg and with a history of lopinavir/ritonavir use and/or adherence issues."( Impact of body weight and missed doses on lopinavir concentrations with standard and increased lopinavir/ritonavir doses during late pregnancy.
Best, BM; Buranabanjasatean, S; Capparelli, EV; Cressey, TR; Jourdain, G; Lallemant, M; Limtrakul, A; Mirochnick, M; Rawangban, B; Sabsanong, P; Stek, A; Treluyer, JM; Urien, S, 2015
)
0.82
" A pediatric population pharmacokinetic model for APV was developed and simulation was used to identify dosing regimens for pediatric patients receiving FPV in combination with ritonavir (RTV) which resulted in concentrations similar to those in adults receiving FPV/RTV 700/100 mg BID."( Population pharmacokinetic modeling and simulation of amprenavir following fosamprenavir/ritonavir administration for dose optimization in HIV infected pediatric patients.
Barbour, AM; Gibiansky, L; Wire, MB, 2014
)
0.82
" This work aimed to develop appropriate drug ratios and dosing for each FDC."( Lopinavir/ritonavir plus lamivudine and abacavir or zidovudine dose ratios for paediatric fixed-dose combinations.
Bouazza, N; Burger, D; Capparelli, EV; Fauchet, F; Foissac, F; Kiechel, JR; Lallemant, M; Treluyer, JM; Urien, S, 2015
)
0.82
" Monte-Carlo simulations of WHO and FDA dosing recommendations were performed to assess their ability to provide optimal exposure in children weighing 4 to 25 kg based on consensus plasma targets."( Lopinavir/ritonavir plus lamivudine and abacavir or zidovudine dose ratios for paediatric fixed-dose combinations.
Bouazza, N; Burger, D; Capparelli, EV; Fauchet, F; Foissac, F; Kiechel, JR; Lallemant, M; Treluyer, JM; Urien, S, 2015
)
0.82
" Given the recommended drug ratios, the dosage for the 4-5."( Lopinavir/ritonavir plus lamivudine and abacavir or zidovudine dose ratios for paediatric fixed-dose combinations.
Bouazza, N; Burger, D; Capparelli, EV; Fauchet, F; Foissac, F; Kiechel, JR; Lallemant, M; Treluyer, JM; Urien, S, 2015
)
0.82
"We conducted an open-label study of rifabutin dosed at 5 mg/kg three times a week in HIV-infected children≤5 years of age receiving lopinavir/ritonavir."( Pharmacokinetics and safety of rifabutin in young HIV-infected children receiving rifabutin and lopinavir/ritonavir.
Gous, H; Kellermann, T; Kindra, G; McIlleron, H; Moultrie, H; Sawry, S; Van Rie, A; Wiesner, L, 2015
)
0.83
"Rifabutin dosed at 5 mg/kg three times per week resulted in lower AUC0-48, AUC0-24 and Cmax values for rifabutin and 25-O-desacetyl rifabutin compared with adults receiving 150 mg of rifabutin daily, the current recommended dose."( Pharmacokinetics and safety of rifabutin in young HIV-infected children receiving rifabutin and lopinavir/ritonavir.
Gous, H; Kellermann, T; Kindra, G; McIlleron, H; Moultrie, H; Sawry, S; Van Rie, A; Wiesner, L, 2015
)
0.63
" As these anti-HIV lipid nanoparticles also prolonged plasma drug exposure, they hold promise as a long-acting dosage form for HIV patients in addressing residual virus in cells and tissue."( Anti-HIV drug-combination nanoparticles enhance plasma drug exposure duration as well as triple-drug combination levels in cells within lymph nodes and blood in primates.
Freeling, JP; Ho, RJ; Koehn, J; Shu, C; Sun, J, 2015
)
0.42
" The development of new antiretroviral drugs with simpler dosing regimens and lower toxicity has led to evaluation of innovative strategies such as dual therapy for initial ART in treatment-naive, with the aim of preventing long-term toxicity and increasing treatment adherence."( [Lopinavir/ritonavir in new initial antiretroviral treatment strategies].
Cahn, P; Figueroa, MI; Rolón, MJ; Sued, O, 2014
)
0.79
" In patients with renal failure, LPV/r does not require dosage adjustment because it is metabolized in the liver."( [Lopinavir/ritonavir in patients with human immunodeficiency virus infection in special situations].
Aldeguer, JL; Tasias, M, 2014
)
0.79
" Although combinations of ritonavir and lopinavir have shown higher plasma concentration level of LPV in clinical settings, dosage adjustment is still required to maintain an adequate therapeutic efficacy and reduce side effects."( A pharmacokinetic model of lopinavir in combination with ritonavir in human.
Duangchaemkarn, K; Lohitnavy, M; Reisfeld, B, 2014
)
0.95
" Plasma progesterone concentrations were measured every 2 weeks after DMPA dosing through week 12."( Depot medroxyprogesterone acetate in combination with a twice-daily lopinavir-ritonavir-based regimen in HIV-infected women showed effective contraception and a lack of clinically significant interactions, with good safety and tolerability: results of the
Aweeka, F; Cohn, SE; Cramer, Y; Klingman, KL; Livingston, E; Luque, AE; Park, JG; Watts, DH; Weinberg, A, 2015
)
0.65
" This phase 2, randomized, open-label study evaluated an IFN- and RBV-free regimen of once-daily ombitasvir (ABT-267), an NS5A inhibitor, plus paritaprevir (ABT-450), an NS3/4A protease inhibitor dosed with ritonavir (paritaprevir/ritonavir), in pegylated IFN/RBV treatment-experienced Japanese patients with hepatitis C virus subtype 1b or genotype 2 infection."( Randomized trial of interferon- and ribavirin-free ombitasvir/paritaprevir/ritonavir in treatment-experienced hepatitis C virus-infected patients.
Badri, P; Chayama, K; Kumada, H; Kurosaki, M; Notsumata, K; Pilot-Matias, T; Rodrigues, L; Sato, K; Setze, C; Vilchez, RA, 2015
)
0.83
" Thirteen studies were conducted to characterize drug-drug interactions for the 3D regimen of OBV, PTV/r, and DSV and various medications in healthy volunteers to inform dosing recommendations in HCV-infected patients."( Drug-drug interaction profile of the all-oral anti-hepatitis C virus regimen of paritaprevir/ritonavir, ombitasvir, and dasabuvir.
Awni, WM; Badri, PS; Coakley, EP; Dutta, S; Hu, B; Khatri, A; Menon, RM; Podsadecki, TJ; Polepally, AR; Wang, H; Wang, T; Zha, J, 2015
)
0.64
" We examined the efficacy and safety of an all-oral interferon-free regimen of ombitasvir, an NS5A inhibitor, and paritaprevir (ABT-450), an NS3/4A protease inhibitor dosed with ritonavir (ombitasvir plus paritaprevir plus ritonavir), given with or without ribavirin."( Ombitasvir plus paritaprevir plus ritonavir with or without ribavirin in treatment-naive and treatment-experienced patients with genotype 4 chronic hepatitis C virus infection (PEARL-I): a randomised, open-label trial.
Asselah, T; Berenguer, M; Fleischer-Stepniewska, K; Hall, C; Hassanein, T; Hézode, C; Marcellin, P; Mobashery, N; Pilot-Matias, T; Pol, S; Reddy, KR; Redman, R; Schnell, G; Vilchez, RA, 2015
)
0.89
" Compared with the results obtained by administration of BMS-663068 alone, coadministration of BMS-663068 with ATV/r increased the BMS-626529 maximum concentration in plasma (Cmax) and the area under the concentration-time curve in one dosing interval (AUCtau) by 68% and 54%, respectively."( Pharmacokinetic interactions between BMS-626529, the active moiety of the HIV-1 attachment inhibitor prodrug BMS-663068, and ritonavir or ritonavir-boosted atazanavir in healthy subjects.
Bertz, R; Furlong, M; Hanna, GJ; Hruska, M; Hwang, C; Landry, IS; Shah, V; Zhu, L, 2015
)
0.62
"We investigated the distribution of ATV plasma trough concentrations according to drug dosage and the potential relationship between ATV plasma trough concentrations and drug-related adverse events in a consecutive series of 240 HIV-infected patients treated with ATV/r 300/100 mg (68%) or ATV 400 mg (32%)."( Metabolic and kidney disorders correlate with high atazanavir concentrations in HIV-infected patients: is it time to revise atazanavir dosages?
Cattaneo, D; Charbe, N; Clementi, E; Cozzi, V; Ferraris, L; Galli, M; Gervasoni, C; Landonio, S; Meraviglia, P; Minisci, D; Molinari, L; Riva, A; Rizzardini, G, 2015
)
0.42
"A significant proportion of patients treated with the conventional dosage of ATV (300/100) had plasma concentrations exceeding the upper therapeutic threshold."( Metabolic and kidney disorders correlate with high atazanavir concentrations in HIV-infected patients: is it time to revise atazanavir dosages?
Cattaneo, D; Charbe, N; Clementi, E; Cozzi, V; Ferraris, L; Galli, M; Gervasoni, C; Landonio, S; Meraviglia, P; Minisci, D; Molinari, L; Riva, A; Rizzardini, G, 2015
)
0.42
" RAUC values were not significantly related to inhibitor dosage or to duration of inhibitor pre-exposure prior to administration of MDZ."( Ritonavir is the best alternative to ketoconazole as an index inhibitor of cytochrome P450-3A in drug-drug interaction studies.
Greenblatt, DJ; Harmatz, JS, 2015
)
1.86
"To describe darunavir (DRV) pharmacokinetics with once-and twice-daily dosing during pregnancy and postpartum in HIV-infected women."( Pharmacokinetics of Once Versus Twice Daily Darunavir in Pregnant HIV-Infected Women.
Best, BM; Burchett, SK; Capparelli, EV; Cressey, TR; Kreitchmann, R; Mirochnick, M; Mofenson, LM; Rungruengthanakit, K; Shapiro, D; Smith, E; Stek, A; Wang, J, 2015
)
0.42
"Women were enrolled in International Maternal Pediatric Adolescent AIDS Clinical Trials Network Protocol P1026s, a prospective nonblinded study of antiretroviral pharmacokinetics in HIV-infected pregnant women that included separate cohorts receiving DRV/ritonavir dosed at either 800 mg/100 mg once daily or 600 mg/100 mg twice daily."( Pharmacokinetics of Once Versus Twice Daily Darunavir in Pregnant HIV-Infected Women.
Best, BM; Burchett, SK; Capparelli, EV; Cressey, TR; Kreitchmann, R; Mirochnick, M; Mofenson, LM; Rungruengthanakit, K; Shapiro, D; Smith, E; Stek, A; Wang, J, 2015
)
0.6
" Median DRV area under the concentration-time curve (AUC) and maximum concentration were significantly reduced during pregnancy with both dosing regimens compared with postpartum, whereas the last measurable concentration (Clast) was also reduced during pregnancy with once daily DRV."( Pharmacokinetics of Once Versus Twice Daily Darunavir in Pregnant HIV-Infected Women.
Best, BM; Burchett, SK; Capparelli, EV; Cressey, TR; Kreitchmann, R; Mirochnick, M; Mofenson, LM; Rungruengthanakit, K; Shapiro, D; Smith, E; Stek, A; Wang, J, 2015
)
0.42
"Atazanavir/ritonavir (ATV/r) recently became the preferred protease inhibitor (PI) for use in Nigeria since it is dosed once daily, which may improve treatment adherence and has fewer side effects than lopinavir/ritonavir (LPV/r)--the most widely available PI in resource-limited settings."( Immunological and Virological Outcomes of Patients Switched from LPV/r to ATV/r-Containing Second- Line Regimens.
Adeyemo, T; Akanmu, AS; Awolola, A; Bello, FO; Kanki, PJ; Lesi, F; Ogunsola, FT; Okonkwo, P; Okwuegbuna, K; Oloko, K, 2015
)
0.81
" The dosing regimen improved treatment outcomes more than two fold from untargeted nanoATV/r."( Pharmacodynamics of folic acid receptor targeted antiretroviral nanotherapy in HIV-1-infected humanized mice.
Araínga, M; Dash, P; Gendelman, HE; Gorantla, S; McMillan, J; Mosley, RL; Palandri, D; Poluektova, L; Puligujja, P, 2015
)
0.42
" The dosing of ATV powder, boosted with 80 mg RTV liquid, was based on three baseline weight bands (5 to <10 kg=150 mg, 10 to <15 kg=200 mg and 15 to <25 kg=250 mg)."( PRINCE-1: safety and efficacy of atazanavir powder and ritonavir liquid in HIV-1-infected antiretroviral-naïve and -experienced infants and children aged ≥3 months to <6 years.
Arce, PM; Biguenet, S; Cambilargiu, D; Correll, T; Donati, AP; Hardy, H; Lissens, J; Strehlau, R; Yang, R, 2015
)
0.66
" A total of 321 patients without cirrhosis were randomized and dosed with double-blind study drug (106 received double-blind placebo and later received open-label OBV/PTV/r), and 42 patients with cirrhosis were enrolled and dosed with open-label OBV/PTV/r."( Randomized phase 3 trial of ombitasvir/paritaprevir/ritonavir for hepatitis C virus genotype 1b-infected Japanese patients with or without cirrhosis.
Burroughs, M; Chayama, K; Ikeda, K; Karino, Y; Kioka, K; Kumada, H; Matsuzaki, Y; Patwardhan, M; Pilot-Matias, T; Redman, R; Rodrigues, L; Sato, K; Setze, C; Suzuki, F; Toyoda, H; Vilchez, RA, 2015
)
0.67
" The impact of HAART on imatinib may depend on whether it is being initiated or has already been dosed chronically in patients."( Human hepatocyte assessment of imatinib drug-drug interactions - complexities in clinical translation.
Beumer, JH; Christner, SM; Kiesel, BF; Parise, RA; Pillai, VC; Rudek, MA; Venkataramanan, R, 2015
)
0.42
"Major protease inhibitor resistance mutations were common in this study of HIV-1-infected children, with the timing of tuberculosis treatment and subsequent protease inhibitor dosing strategy proving to be important associated factors."( Factors Associated with the Development of Drug Resistance Mutations in HIV-1 Infected Children Failing Protease Inhibitor-Based Antiretroviral Therapy in South Africa.
Avenant, T; du Plessis, NM; Feucht, UD; Melikian, G; Rossouw, TM; Thomas, W; van Dyk, G, 2015
)
0.42
" This study confirmed that patients with kidney impairment and the concurrent use of atazanavir/ritonavir will require the dosage of tenofovir to be adjusted to ensure efficacy and prevent unwanted toxicities."( Population pharmacokinetics of tenofovir in HIV/HBV co-infected patients.
Avihingsanon, A; Burger, D; Lewin, SR; Matthews, G; Punyawudho, B; Ruxrungtham, K; Thammajaruk, N; Thongpeang, P, 2015
)
0.64
" Where interaction is possible, risk can be mitigated by paying careful attention to concomitant medications, adjusting drug dosage as needed, and monitoring patient response and/or clinical parameters."( Dosing Recommendations for Concomitant Medications During 3D Anti-HCV Therapy.
Badri, PS; Dutta, S; King, JR; McGovern, BH; Menon, RM; Polepally, AR, 2016
)
0.43
"The histological alterations occurred in both the maternal livers and the maternal kidneys, particularly in group E3, which received the greatest therapeutic dosage (nine times)."( Chronic action of lamivudine and ritonavir on maternal and fetal liver and kidney of albino pregnant rats (Rattus norvegicus albinus, Rodentia, Mammalia): morphological and biochemical aspects.
Amed, AM; Araujo Júnior, E; Júnior, LK; Nakamura, MU; Simões, J; Vangelotti, AM, 2015
)
0.7
" Drug-drug interactions were evaluated in healthy volunteers to develop dosing recommendations for HCV-infected subjects."( Drug Interactions with the Direct-Acting Antiviral Combination of Ombitasvir and Paritaprevir-Ritonavir.
Awni, WM; Badri, PS; Chiu, YL; Dutta, S; Khatri, A; Menon, RM; Podsadecki, TJ; Polepally, AR; Wang, H; Zha, J, 2016
)
0.65
" A drug-drug interaction (DDI) study was conducted to guide dosing recommendations for UDCA and GCR when coadministered with the 2D regimen."( Drug Interactions Between Hepatoprotective Agents Ursodeoxycholic Acid or Glycyrrhizin and Ombitasvir/Paritaprevir/Ritonavir in Healthy Japanese Subjects.
Alves, K; Badri, PS; Ding, B; Dutta, S; Menon, RM; Redman, R; Rodrigues, L; Uchiyama, N; Zha, J, 2015
)
0.63
" In addition, linear mixed-effect analysis was used to compare the area under the concentration-time curve from 0 to 12 h (AUC0-12) and concentration prior to dosing (Cpredose) in pregnant women and nonpregnant subjects."( No Need for Lopinavir Dose Adjustment during Pregnancy: a Population Pharmacokinetic and Exposure-Response Analysis in Pregnant and Nonpregnant HIV-Infected Subjects.
Jones, AK; Klein, CE; Nilius, AM; Patterson, KB; Salem, AH; Santini-Oliveira, M; Taylor, GP, 2016
)
0.43
") dosing in children."( Once vs. twice-daily lopinavir/ritonavir in HIV-1-infected children.
, 2015
)
0.7
" dosing remains an option in selected, adherent children, with close viral load monitoring."( Once vs. twice-daily lopinavir/ritonavir in HIV-1-infected children.
, 2015
)
0.7
" To characterize the pharmacokinetics, safety, and tolerability of paritaprevir and determine an optimal dosing regimen for subsequent evaluations, clinical studies were conducted with paritaprevir alone or with ritonavir, a cytochrome P450 3A4 inhibitor anticipated to increase paritaprevir exposure."( Pharmacokinetics and tolerability of paritaprevir, a direct acting antiviral agent for hepatitis C virus treatment, with and without ritonavir in healthy volunteers.
Awni, WM; Chiu, YL; Dutta, S; Klein, CE; Menon, RM; Podsadecki, TJ, 2016
)
0.82
"Twenty-nine subjects completed both dosing cohorts."( Efavirenz but Not Atazanavir/Ritonavir Significantly Reduces Atovaquone Concentrations in HIV-Infected Subjects.
Alfaro, RM; Calderón, MM; Kovacs, JA; Kumar, P; McManus, M; Pau, AK; Penzak, SR, 2016
)
0.73
"The aim of this study was to develop a nanotechnology to formulate a fixed-dose combination of poorly water-soluble drugs in a children-friendly, flexible solid dosage form."( Development and in vivo evaluation of child-friendly lopinavir/ritonavir pediatric granules utilizing novel in situ self-assembly nanoparticles.
Dong, X; Guo, S; Li, D; Penzak, S; Pham, K, 2016
)
0.67
" Thus, co-medications should be systematically reviewed when switching the pharmacokinetic enhancer to anticipate potential dosage adjustments."( Cobicistat versus ritonavir boosting and differences in the drug-drug interaction profiles with co-medications.
Back, D; Gibbons, S; Khoo, S; Marzolini, C, 2016
)
0.77
"57-fold on repeated dosing compared with the first dose."( Dose- and Formulation-Dependent Non-Linear Pharmacokinetic Model of Paritaprevir, a Protease Inhibitor for the Treatment of Hepatitis C Virus Infection: Combined Analysis from 12 Phase I Studies.
Awni, WM; Beck, D; Dutta, S; Khatri, A; Liu, W; Menon, RM; Mensing, S; Polepally, AR, 2016
)
0.43
"The complex pharmacokinetics of paritaprevir were well described by the model, which can be used as a basis for clinical trial dosing and further evaluations in patients with HCV."( Dose- and Formulation-Dependent Non-Linear Pharmacokinetic Model of Paritaprevir, a Protease Inhibitor for the Treatment of Hepatitis C Virus Infection: Combined Analysis from 12 Phase I Studies.
Awni, WM; Beck, D; Dutta, S; Khatri, A; Liu, W; Menon, RM; Mensing, S; Polepally, AR, 2016
)
0.43
" This study sheds light on the complex dissolution and solution phase behavior of multicomponent amorphous dosage forms, in particular those containing poorly water soluble drugs, which may undergo supersaturation in vivo."( Compromised in vitro dissolution and membrane transport of multidrug amorphous formulations.
Alhalaweh, A; Bergström, CAS; Taylor, LS, 2016
)
0.43
"Among 2,053 patients enrolled and dosed with study drug, 410 (20%) were receiving concomitant ARAs; of these, 308 (15%) were taking concomitant PPIs."( Safety and Efficacy of Ombitasvir/Paritaprevir/Ritonavir Plus Dasabuvir With or Without Ribavirin in HCV-Infected Patients Taking Concomitant Acid-Reducing Agents.
Asselah, T; Bennett, M; Forns, X; Liu, L; Moller, J; Pedrosa, M; Planas Vila, R; Reau, N; Rustgi, V; Shiffman, ML, 2016
)
0.69
" Here we present a population pharmacokinetic model to describe the longitudinal change of unbound lopinavir/ritonavir (LPV/RTV) PK parameters with gestational age, and to predict unbound LPV concentrations under different dosing regimens."( Model-Based Analysis of Unbound Lopinavir Pharmacokinetics in HIV-Infected Pregnant Women Supports Standard Dosing in the Third Trimester.
Chen, J; Dumond, JB; Malone, S; Patterson, KB; Prince, HM, 2016
)
0.65
" The novel ISNP nanotechnology is a platform to manufacture oral solid dosage forms for poorly water-soluble drugs, especially for pediatric formulation development."( Novel in situ self-assembly nanoparticles for formulating a poorly water-soluble drug in oral solid granules, improving stability, palatability, and bioavailability.
Dong, X; Guo, S; Li, D; Penzak, SR; Pham, K, 2016
)
0.43
"RPV PK in this adolescent population was similar to adults when dosed without DRV/r."( Rilpivirine Pharmacokinetics Without and With Darunavir/Ritonavir Once Daily in Adolescents and Young Adults.
Acosta, EP; Britto, P; Carey, VJ; Cressey, TR; Foca, M; Graham, B; Hazra, R; Jean-Philippe, P; King, J; Wiznia, A; Yogev, R, 2016
)
0.68
"Cobicistat and ritonavir are structurally distinct compounds that both potently inhibit cytochrome P450 (CYP) 3A, the metabolizing enzyme primarily responsible for the elimination of several antiretroviral medications, and, as such, are pharmacokinetic boosters for antiretroviral agents that require longer dosing intervals."( Drug Interactions with Cobicistat- or Ritonavir-Boosted Elvitegravir.
Custodio, JM; McNicholl, I; Nguyen, T; Piontkowsky, D; Szwarcberg, J,
)
0.76
" The success of these dosing recommendations was evaluated by analyzing pharmacokinetic data from liver transplant recipients in the CORAL-I study."( Pharmacokinetics of Tacrolimus and Cyclosporine in Liver Transplant Recipients Receiving 3 Direct-Acting Antivirals as Treatment for Hepatitis C Infection.
Awni, WM; Badri, PS; Coakley, EP; Ding, B; Dutta, S; Menon, RM; Parikh, A, 2016
)
0.43
"A population pharmacokinetic model was developed using tacrolimus dosing and Ctrough data before and during 3D treatment (n = 29)."( Pharmacokinetics of Tacrolimus and Cyclosporine in Liver Transplant Recipients Receiving 3 Direct-Acting Antivirals as Treatment for Hepatitis C Infection.
Awni, WM; Badri, PS; Coakley, EP; Ding, B; Dutta, S; Menon, RM; Parikh, A, 2016
)
0.43
"Observed data for tacrolimus and CSA in liver transplant recipients confirm that the recommended dosing strategies are valid and therapeutic levels of immunosuppression can be maintained during 3D treatment."( Pharmacokinetics of Tacrolimus and Cyclosporine in Liver Transplant Recipients Receiving 3 Direct-Acting Antivirals as Treatment for Hepatitis C Infection.
Awni, WM; Badri, PS; Coakley, EP; Ding, B; Dutta, S; Menon, RM; Parikh, A, 2016
)
0.43
" C57BL/6J mice infected with the Daniel's strain of Theiler's murine encephalomyelitis virus were treated with the FDA-approved drug ritonavir using a dosing regimen that resulted in plasma concentrations within the therapeutic range for calpain inhibition."( Neuroprotection mediated by inhibition of calpain during acute viral encephalitis.
Buenz, EJ; Howe, CL; LaFrance-Corey, RG; McGovern, RM; Mirchia, K; Reid, JM; Sauer, BM, 2016
)
0.64
"Overall, 24 subjects, six in each of four renal function groups (normal, mild, moderate, and severe), received a single dose of the 3D and 2D regimens in separate dosing periods."( Pharmacokinetics and Tolerability of Anti-Hepatitis C Virus Treatment with Ombitasvir, Paritaprevir, Ritonavir, with or Without Dasabuvir, in Subjects with Renal Impairment.
Awni, WM; Dutta, S; Khatri, A; Marbury, TC; Menon, RM; Preston, RA; Rodrigues, L; Wang, H, 2017
)
0.67
" One hundred patients with pulmonary tuberculosis (65% coinfected with HIV-1) were intensively sampled to determine rifampin, isoniazid, and pyrazinamide plasma concentrations after 7 to 8 weeks of a daily quadruple-therapy regimen dosed according to World Health Organization (WHO) weight bands."( HIV-1 Coinfection Does Not Reduce Exposure to Rifampin, Isoniazid, and Pyrazinamide in South African Tuberculosis Outpatients.
Chirehwa, M; Denti, P; McIlleron, H; Meintjes, G; Rockwood, N; Wiesner, L; Wilkinson, RJ, 2016
)
0.43
"There were higher rates of clinical TB cure in individuals on a boosted protease inhibitor-based ART regimen with daily RBT compared to intermittently dosed RBT."( Improved tuberculosis outcomes with daily vs. intermittent rifabutin in HIV-TB coinfected patients in India.
Ambrose, P; Benson, CA; Devaraj, C; Ezhilarasi, C; Jenks, JD; Kumarasamy, N; Poongulali, S; Yepthomi, T, 2016
)
0.43
" A hyperinsulinaemic euglycaemic clamp was performed prior to and following each 2-week dosing phase."( An open-label, randomized study of the impact on insulin sensitivity, lipid profile and vascular inflammation by treatment with lopinavir/ritonavir or raltegravir in HIV-negative male volunteers.
Boffito, M; Jackson, A; Milinkovic, A; Moyle, G; Randell, P, 2017
)
0.66
" When combined with the CYP3A4 substrate daclatasvir, the daclatasvir dosage should be reduced from 60 to 30 mg once daily."( Daclatasvir 30 mg/day is the correct dose for patients taking atazanavir/cobicistat.
Burger, DM; Colbers, EP; de Kanter, CT; Drenth, JP; Smolders, EJ; Velthoven-Graafland, K, 2017
)
0.46
" For treatment, the WHO dosing guidelines should be suitable to maintain values above the therapeutic pharmacokinetic targets in most infants."( Are Prophylactic and Therapeutic Target Concentrations Different?: the Case of Lopinavir-Ritonavir or Lamivudine Administered to Infants for Prevention of Mother-to-Child HIV-1 Transmission during Breastfeeding.
Blanche, S; Blume, J; Bouazza, N; Foissac, F; Harper, K; Hirt, D; Illamola, SM; Kankasa, C; Meda, N; Nagot, N; Singata-Madliki, M; Tréluyer, JM; Tumwine, JK; Tylleskär, T; Van de Perre, P, 2017
)
0.68
" To normalise bedaquiline exposure in patients with concomitant LPV/r therapy, an adjusted bedaquiline dosing regimen is proposed for further study."( Confirming model-predicted pharmacokinetic interactions between bedaquiline and lopinavir/ritonavir or nevirapine in patients with HIV and drug-resistant tuberculosis.
Brill, MJ; Karlsson, MO; Maartens, G; Pandie, M; Svensson, EM, 2017
)
0.68
"The dosage of darunavir/ritonavir is 800/100 mg once daily for treatment-naive patients or treatment-experienced patients with no prior darunavir resistance associated mutations, and 600/100 mg twice daily for treatment-experienced patients with one or more darunavir resistance associated mutations."( Pharmacokinetics and Safety of Darunavir/Ritonavir in HIV-Infected Pregnant Women.
Brown, K; Burger, D; Hadacek, MB; Hill, A; Khoo, S; La Porte, C; Moecklinghoff, C; Peytavin, G,
)
0.7
" In the phase II/III clinical studies, ALT and bilirubin increases were reversible with continued dosing or after treatment cessation."( Exposure-Safety Response Relationship for Ombitasvir, Paritaprevir/Ritonavir, Dasabuvir, and Ribavirin in Patients with Chronic Hepatitis C Virus Genotype 1 Infection: Analysis of Data from Five Phase II and Six Phase III Studies.
Awni, W; DaSilva-Tillmann, B; Dutta, S; Lin, CW; Liu, W; Menon, R; Podsadecki, T; Shulman, N, 2017
)
0.69
"89 μIU/mL) despite gradual escalation of the levothyroxine dosage to 175 μg daily."( Probable interaction between levothyroxine and ritonavir: Case report and literature review.
Ahmed, RA; Foisy, MM; Hughes, CA; Sahajpal, R, 2017
)
0.71
" Patients were randomly assigned (1:1) to receive 25 mg ombitasvir, 150 mg paritaprevir, and 100 mg ritonavir once daily, with weight-based ribavirin dosed twice daily for either 12 weeks or 16 weeks."( Ombitasvir, paritaprevir, and ritonavir plus ribavirin in adults with hepatitis C virus genotype 4 infection and cirrhosis (AGATE-I): a multicentre, phase 3, randomised open-label trial.
Asselah, T; ElKhashab, M; Feld, JJ; Ferenci, P; Hassanein, T; Hézode, C; Mobashery, N; Moreno, C; Papatheodoridis, G; Pilot-Matias, T; Qaqish, RB; Redman, R; Yu, Y; Zeuzem, S, 2016
)
0.94
" We assessed the efficacy and safety of the two direct-acting antiviral drugs ombitasvir, an NS5A inhibitor, and paritaprevir, an NS3/4A protease inhibitor dosed with ritonavir, plus ribavirin in treatment of chronic HCV infection in Egypt."( Ombitasvir, paritaprevir, and ritonavir plus ribavirin for chronic hepatitis C virus genotype 4 infection in Egyptian patients with or without compensated cirrhosis (AGATE-II): a multicentre, phase 3, partly randomised open-label trial.
Allam, N; Asselah, T; Doss, W; Esmat, G; Hall, C; Hassany, M; Mobashery, N; Mohey, MA; Qaqish, RB; Redman, R; Shiha, G; Soliman, R; Waked, I; Yosry, A; Zayed, N, 2016
)
0.92
" Also, the exposure to LPV on the initial once-daily dosing regimen was determined."( Sustained Viral Suppression in HIV-infected Children on Once-daily Lopinavir/Ritonavir in Clinical Practice.
Bastiaans, DET; Burger, DM; de Groot, R; Driessen, GJA; Fraaij, PLA; Gondrie, IPE; Hartwig, NG; van der Knaap, LC; van Jaarsveld, P; van Rossum, AMC; Visser, EG, 2017
)
0.68
" Geometric mean LPV area under the plasma concentration-time curve (linear up-log down method) over a dosing interval from time 0 to 24 hours after dosing was 169."( Sustained Viral Suppression in HIV-infected Children on Once-daily Lopinavir/Ritonavir in Clinical Practice.
Bastiaans, DET; Burger, DM; de Groot, R; Driessen, GJA; Fraaij, PLA; Gondrie, IPE; Hartwig, NG; van der Knaap, LC; van Jaarsveld, P; van Rossum, AMC; Visser, EG, 2017
)
0.68
"Current presentations of the anti-HIV drugs lopinavir and ritonavir make appropriate dosing for children difficult."( Developing a Flexible Pediatric Dosage Form for Antiretroviral Therapy: A Fast-Dissolving Tablet.
Estrada, M; Lai, M; Lal, M; Zhu, C, 2017
)
0.7
" Darunavir concentrations at the end of the dosing interval were quantified by LC-MS/MS."( Darunavir concentrations in CSF of HIV-infected individuals when boosted with cobicistat versus ritonavir.
Bartels, H; Battegay, M; Decosterd, L; Marzolini, C, 2017
)
0.67
" Similar values were found at all 3 time points, thus indicating that there is probably no need to adapt MPA dosage to 3D."( Managing Drug-Drug Interaction Between Ombitasvir, Paritaprevir/Ritonavir, Dasabuvir, and Mycophenolate Mofetil.
Bellissant, E; Ben Ali, Z; Boglione-Kerrien, C; Guyader, D; Jezequel, C; Lemaitre, F; Tron, C; Verdier, MC, 2017
)
0.69
" Next, population fetal pharmacokinetic profiles were simulated for different maternal darunavir/ritonavir dosing regimens."( Prediction of Fetal Darunavir Exposure by Integrating Human Ex-Vivo Placental Transfer and Physiologically Based Pharmacokinetic Modeling.
Buaben, AO; Burger, DM; Colbers, AP; Freriksen, JJM; Greupink, R; Russel, FGM; Schalkwijk, S, 2018
)
0.7
"The results indicate that the population fetal exposure after oral maternal darunavir dosing is therapeutic and this may provide benefits to the prevention of mother-to-child transmission of human immunodeficiency virus."( Prediction of Fetal Darunavir Exposure by Integrating Human Ex-Vivo Placental Transfer and Physiologically Based Pharmacokinetic Modeling.
Buaben, AO; Burger, DM; Colbers, AP; Freriksen, JJM; Greupink, R; Russel, FGM; Schalkwijk, S, 2018
)
0.48
" Therefore, it is likely safe to coadminister ritonavir with DE, while there is a potential need for reduced dosing and prudent clinical monitoring with the coadministration of cobicistat due to the greater net inhibition of intestinal P-gp transport and increased bioavailability."( Differential Influence of the Antiretroviral Pharmacokinetic Enhancers Ritonavir and Cobicistat on Intestinal P-Glycoprotein Transport and the Pharmacokinetic/Pharmacodynamic Disposition of Dabigatran.
Alfaro, RM; Brooks, KM; George, JM; Gordon, LA; Hadigan, C; Kellogg, A; Kumar, P; Lozier, J; McManus, M; Nghiem, K; Penzak, SR, 2017
)
0.95
" It is desirable for multidrug combinations to be coformulated into single dosage forms where possible, to promote patient convenience and adherence to dosage regimens, for which amorphous solid dispersion (ASD) is particularly well-suited."( Multidrug, Anti-HIV Amorphous Solid Dispersions: Nature and Mechanisms of Impacts of Drugs on Each Other's Solution Concentrations.
Arca, HÇ; Dahal, D; Edgar, KJ; Mosquera-Giraldo, LI; Taylor, LS, 2017
)
0.46
" The biphasic test revealed significantly lower degrees of supersaturation of RTV in the aqueous media from these dosage forms as compared to results of the two-stage dissolution test."( In Vitro Characterization of Ritonavir Drug Products and Correlation to Human in Vivo Performance.
Gao, P; Marroum, P; Shi, Y; Vela, S; Xu, H, 2017
)
0.75
"This is the first demonstration that intracellular atazanavir exposure remains unchanged during pregnancy supporting the standard 300/100 mg atazanavir/ritonavir dosing throughout pregnancy."( Atazanavir intracellular concentrations remain stable during pregnancy in HIV-infected patients.
Bonito, A; Bonora, S; Calcagno, A; Casari, S; Castelli, F; D'Avolio, A; Di Perri, G; Domenighini, E; Focà, E; Quiros Roldan, E; Simiele, M; Trentini, L, 2017
)
0.65
" These factors should be taken into consideration to guide the individual tenofovir disoproxil fumarate dosage regimen in Thai HIV-infected patients."( Pharmacogenetics-based population pharmacokinetic analysis of tenofovir in Thai HIV-infected patients.
Avihingsanon, A; Burger, DM; Mitruk, S; Punyawudho, B; Rungtivasuwan, K; Ruxrungtham, K; Sukasem, C; Thammajaruk, N, 2017
)
0.46
"The World Health Organization (WHO) recommends weight band dosing of antiretrovirals for children."( A Phase II/III Trial of Lopinavir/Ritonavir Dosed According to the WHO Pediatric Weight Band Dosing Guidelines.
Capparelli, EV; Cressey, TR; Mirochnick, M; Pinto, JA; Qin, M; Siberry, GK; Smith, B; Spector, SA; Warshaw, M; Zimmer, B, 2018
)
0.76
"International Maternal Pediatric Adolescent AIDS Clinical Trials (IMPAACT) P1083 was a phase II/III trial assessing the pharmacokinetics (PK) and short-term safety, tolerance and efficacy of lopinavir/ritonavir in human immunodeficiency virus-infected children 3-25 kg dosed according to WHO weight bands, with liquid solution or meltrex extrusion tablets."( A Phase II/III Trial of Lopinavir/Ritonavir Dosed According to the WHO Pediatric Weight Band Dosing Guidelines.
Capparelli, EV; Cressey, TR; Mirochnick, M; Pinto, JA; Qin, M; Siberry, GK; Smith, B; Spector, SA; Warshaw, M; Zimmer, B, 2018
)
0.95
" The median (Q1, Q3) difference between individual observed PK parameters and those expected if Food and Drug Administration dosing guidelines were followed was 30."( A Phase II/III Trial of Lopinavir/Ritonavir Dosed According to the WHO Pediatric Weight Band Dosing Guidelines.
Capparelli, EV; Cressey, TR; Mirochnick, M; Pinto, JA; Qin, M; Siberry, GK; Smith, B; Spector, SA; Warshaw, M; Zimmer, B, 2018
)
0.76
"WHO weight band dosing guidelines in children achieved adequate LPV plasma exposure but was higher than that expected with Food and Drug Administration dosing guidelines."( A Phase II/III Trial of Lopinavir/Ritonavir Dosed According to the WHO Pediatric Weight Band Dosing Guidelines.
Capparelli, EV; Cressey, TR; Mirochnick, M; Pinto, JA; Qin, M; Siberry, GK; Smith, B; Spector, SA; Warshaw, M; Zimmer, B, 2018
)
0.76
" Fifteen patients (five groups; 3 per group) completed dosing as follows: (1) LNF 200 mg twice-daily (BID; 12 weeks); (2) LNF 300 mg BID (12 weeks); (3) LNF 100 mg thrice-daily (5 weeks); (4) LNF 100 mg BID + pegylated interferon alfa (PEG-IFNα) 180 μg once-weekly (QW; 8 weeks); and (5) LNF 100 mg BID + ritonavir (RTV) 100 mg once-daily (QD; 8 weeks)."( Optimizing lonafarnib treatment for the management of chronic delta hepatitis: The LOWR HDV-1 study.
Bozdayi, AM; Çalişkan, A; Glenn, JS; Heller, T; Idilman, R; Kalkan, Ç; Karakaya, F; Karatayli, E; Karatayli, S; Keskin, O; Koh, C; Yurdaydin, C, 2018
)
0.66
" The phase 2b trial AI438011 investigated the safety, efficacy, and dose-response of fostemsavir vs ritonavir-boosted atazanavir (ATV/r) in treatment-experienced, HIV-1-infected subjects."( Viral Drug Resistance Through 48 Weeks, in a Phase 2b, Randomized, Controlled Trial of the HIV-1 Attachment Inhibitor Prodrug, Fostemsavir.
Hanna, GJ; Joshi, SR; Krystal, M; Lataillade, M; Lee, S; Stock, DA; Zhou, N, 2018
)
0.7
"Intensive 24-hour pharmacokinetic profiles at steady state compared ATV exposures (area under the concentration-time curve in one dosing interval) in 5 ATV + RTV baseline weight-band dosing categories, with historic data in adults receiving ATV + RTV 300/100 mg capsules."( Pharmacokinetics and Pharmacodynamics of Atazanavir in HIV-1-Infected Children Treated With Atazanavir Powder and Ritonavir: Combined Analysis of the PRINCE-1 and -2 Studies.
Correll, TA; Eley, T; Pikora, C; Sevinsky, H; Wang, R; Xu, X; Zaru, L, 2018
)
0.69
"Weight-band dosing of ATV + RTV plus optimized dual nucleos(t)ide reverse transcriptase inhibitors in young HIV-1-infected children achieved similar ATV exposure to that in adults; no unexpected safety findings occurred, and with the exception of lower virologic suppression in the lowest ATV CCQ, there was no apparent trend in virologic suppression across ATV CCQs."( Pharmacokinetics and Pharmacodynamics of Atazanavir in HIV-1-Infected Children Treated With Atazanavir Powder and Ritonavir: Combined Analysis of the PRINCE-1 and -2 Studies.
Correll, TA; Eley, T; Pikora, C; Sevinsky, H; Wang, R; Xu, X; Zaru, L, 2018
)
0.69
" RBV was dosed by physician discretion between 200 mg weekly and 200 mg daily."( High Efficacy of ombitasvir/paritaprevir/ritonavir plus dasabuvir in hepatitis C genotypes 4 and 1-infected patients with severe chronic kidney disease.
Afghani, AA; Alghamdi, AS; Alghamdi, MN; AlMousa, A; Alswat, K; AlZanbagi, A; Aseeri, M; Assiri, AM; Babatin, MA; Sanai, FM, 2018
)
0.75
" The developed stability indicating method is capable for determination of impurities of Atazanavir and Ritonavir in combined tablet dosage form as well as individual dosage forms also."( Simultaneous Determination of Impurities of Atazanavir and Ritonavir in Tablet Dosage Form by Using Reversed-Phase Ultra Performance Liquid Chromatographic Method.
Mantena, BPV; Mantripragada, MKVVN; Nutulapati, VVS; Rao, SV, 2018
)
0.94
" The study objective was to determine the effect of different dosage regimens of ritonavir, a strong CYP3A inhibitor, on the pharmacokinetics of venetoclax in 20 healthy subjects."( Impact of ritonavir dose and schedule on CYP3A inhibition and venetoclax clinical pharmacokinetics.
Freise, KJ; Hu, B; Salem, AH, 2018
)
1.11
" We describe the clinical management of ribavirin dosing in hepatitis C virus-infected patients receiving ombitasvir/paritaprevir/ritonavir and dasabuvir with ribavirin."( Ribavirin dose management in HCV patients receiving ombitasvir/paritaprevir/ritonavir and dasabuvir with ribavirin.
Bernstein, DE; Feld, JJ; Ferenci, P; Larsen, L; Tatsch, F; Vlierberghe, HV; Younes, Z, 2018
)
0.92
"In this multicenter pharmacokinetic study in HIV-infected children (6-12 years of age), we validated the approved once-daily darunavir/ritonavir dosing recommendations."( Pharmacokinetics, Short-term Safety and Efficacy of the Approved Once-daily Darunavir/Ritonavir Dosing Regimen in HIV-infected Children.
Bastiaans, DET; Burger, DM; Colbers, APH; Geelen, SPM; Roukens, M; van der Flier, M; van Rossum, AMC; Vermont, CL; Visser, EG, 2018
)
0.91
" In 4 macaques dosed subcutaneously, drug levels over 5 weeks in plasma, lymph node mononuclear cells (LNMCs), and peripheral blood mononuclear cells (PBMCs) were analyzed by liquid chromatography-tandem mass spectrometry."( Long-Acting Profile of 4 Drugs in 1 Anti-HIV Nanosuspension in Nonhuman Primates for 5 Weeks After a Single Subcutaneous Injection.
Collier, AC; Ho, RJY; Kinman, LM; Koehn, J; Kraft, JC; Lane, S; Lee, W; McConnachie, LA, 2018
)
0.48
"5%) patients (calcium channel blockers, ACE inhibitors, statins, diuretics, tacrolimus); four patients required further adjustment of antihypertensive drugs or tacrolimus dosage on-treatment."( Paritaprevir/Ritonavir/Ombitasvir Plus Dasabuvir Regimen in the Treatment of Genotype 1 Chronic Hepatitis C Infection in Patients with Severe Renal Impairment and End-Stage Renal Disease: a Real-Life Cohort.
Chmelova, K; Frankova, S; Kreidlova, M; Merta, D; Senkerikova, R; Sperl, J, 2018
)
0.85
" To establish a method of dosage adjustment for darunavir (DRV) based on pharmacokinetic theory, we analyzed the correlation between DRV levels in peripheral blood mononuclear cells (PBMCs) and plasma."( Darunavir concentration in PBMCs may be a better indicator of drug exposure in HIV patients.
Araki, T; Gohda, F; Handa, H; Nagano, D; Nakamura, T; Ogawa, Y; Uchiumi, H; Yamamoto, K; Yanagisawa, K, 2018
)
0.48
"To evaluate total and unbound darunavir exposures following standard darunavir/ritonavir dosing and to explore the value of potential optimized darunavir/ritonavir dosing regimens for HIV-positive pregnant women."( Evaluating darunavir/ritonavir dosing regimens for HIV-positive pregnant women using semi-mechanistic pharmacokinetic modelling.
Best, BM; Burger, DM; Capparelli, E; Colbers, A; Cressey, TR; Greupink, R; Karlsson, MO; Mirochnick, M; Moltó, J; Russel, FGM; Schalkwijk, S; Ter Heine, R, 2019
)
1.06
" The probability of therapeutic exposure (unbound) during pregnancy was higher for standard q12h dosing (99%) than for q24h dosing (94%)."( Evaluating darunavir/ritonavir dosing regimens for HIV-positive pregnant women using semi-mechanistic pharmacokinetic modelling.
Best, BM; Burger, DM; Capparelli, E; Colbers, A; Cressey, TR; Greupink, R; Karlsson, MO; Mirochnick, M; Moltó, J; Russel, FGM; Schalkwijk, S; Ter Heine, R, 2019
)
0.83
" The value of alternative dosing regimens seems limited."( Evaluating darunavir/ritonavir dosing regimens for HIV-positive pregnant women using semi-mechanistic pharmacokinetic modelling.
Best, BM; Burger, DM; Capparelli, E; Colbers, A; Cressey, TR; Greupink, R; Karlsson, MO; Mirochnick, M; Moltó, J; Russel, FGM; Schalkwijk, S; Ter Heine, R, 2019
)
0.83
"To evaluate the proportion of children with lopinavir Cmin ≥1 mg/L when receiving a novel 8-hourly lopinavir/ritonavir dosing strategy during rifampicin co-treatment."( Pharmacokinetics of adjusted-dose 8-hourly lopinavir/ritonavir in HIV-infected children co-treated with rifampicin.
McIlleron, H; Rabie, H; Rawizza, H; Van Rie, A; Wiesner, L; Winckler, J; Zar, H; Zuidewind, P, 2019
)
0.98
" Rifampicin was dosed at 10-20 mg/kg/day."( Pharmacokinetics of adjusted-dose 8-hourly lopinavir/ritonavir in HIV-infected children co-treated with rifampicin.
McIlleron, H; Rabie, H; Rawizza, H; Van Rie, A; Wiesner, L; Winckler, J; Zar, H; Zuidewind, P, 2019
)
0.76
" The method was validated according to International Conference on Harmonization guidelines, and it was successively applied for the determination of the studied drugs in their different pharmaceutical dosage forms and gave excellent percent of recovery."( Silver Nanoparticles Synthesis for Sensitive Spectrophotometric Determination of Sofosbuvir, Lamivudine, and Ritonavir in Pure Forms and Pharmaceutical Dosage Forms.
Elhenawee, M; Saleh, H; Saraya, RE, 2020
)
0.77
" The final analysis data set contained 17,725 observations from 1,054 subjects, including healthy subjects and subjects with drug-sensitive, multidrug-resistant, or extensively drug-resistant pulmonary tuberculosis dosed pretomanid in monotherapy or combination therapy for up to 6 months."( Population Pharmacokinetics of the Antituberculosis Agent Pretomanid.
Everitt, D; Nedelman, JR; Salinger, DH; Subramoney, V, 2019
)
0.51
" The results showed that the steady-state trough concentration of multiple dosing of GLS4 alone was significantly lower than the 90% effective concentration of 55."( A First-in-Human Trial of GLS4, a Novel Inhibitor of Hepatitis B Virus Capsid Assembly, following Single- and Multiple-Ascending-Oral-Dose Studies with or without Ritonavir in Healthy Adult Volunteers.
Cui, Y; Huang, Z; Jia, B; Luo, L; Ren, Q; Sheng, X; Wang, X; Xu, J; Zhang, Y; Zhao, H; Zhao, N; Zhao, X, 2019
)
0.71
" The objective of the study was to apply physiologically-based pharmacokinetic (PBPK) modeling to predict optimal dosage regimens of quinine when coadministered with lopinavir/ritonavir in malaria and HIV coinfected patients with different conditions."( Physiologically-Based Pharmacokinetic Modeling for Optimal Dosage Prediction of Quinine Coadministered With Ritonavir-Boosted Lopinavir.
Karbwang, J; Na-Bangchang, K; Rajoli, RKR; Saeheng, T; Siccardi, M, 2020
)
0.96
" With the assistance of therapeutic drug monitoring, dolutegravir dosing was increased to 150 mg daily."( Successful use of once-daily high-dose darunavir and dolutegravir in multidrug-resistant HIV.
Fulco, PP; Gomes, D; Leibrand Kaczmar, CR; Smith, T, 2020
)
0.56
"Solid dosage forms of amorphous solid dispersions (ASDs) have rarely been assessed for their crushability, although it might possibly be a more frequent practice than thought to facilitate oral administration in several clinical conditions (e."( The influence of crushing amorphous solid dispersion dosage forms on the in-vitro dissolution kinetics.
Appeltans, B; Pas, T; Van den Mooter, G; Verbert, S, 2020
)
0.56
" These models were then used to assess the magnitude of interaction at therapeutic venetoclax doses and to explore various clinical dosing strategies that maintain venetoclax and digoxin concentrations within their respective therapeutic windows."( Semimechanistic Modeling to Guide Venetoclax Coadministration with Ritonavir and Digoxin.
Alhadab, AA; Freise, KJ; Salem, AH, 2020
)
0.79
" In dose-escalation trials, the maximum tolerated doses for two different dosing regimens were established and dose-limiting toxicities (DLTs) were recorded."( Quantification of the pharmacokinetic-toxicodynamic relationship of oral docetaxel co-administered with ritonavir.
Beijnen, JH; de Weger, VA; Dorlo, TPC; Huitema, ADR; Janssen, JM; Marchetti, S; Nuijen, B; Schellens, JHM; Stuurman, RE; Yu, H, 2020
)
0.77
" This study aimed to develop a mechanistic absorption and disposition model to describe exposure to ritonavir following oral dosing of the commercial amorphous solid dispersion tablet, Norvir, under fasted and fed conditions."( Biopharmaceutic
Arora, S; Gardner, I; Jamei, M; Kilford, P; Pansari, A; Turner, DB, 2020
)
0.77
"This paper summarizes efforts to (i) better understand the behavior of amorphous solid dispersions (ASDs) under real-life dosing conditions and (ii) evaluate the capability of in vitro methodologies to capture gastro-intestinal drug disposition."( The effect of reduced gastric acid secretion on the gastrointestinal disposition of a ritonavir amorphous solid dispersion in fasted healthy volunteers: an in vivo - in vitro investigation.
Augustijns, P; Barker, R; Beato, S; Kostantini, C; Kourentas, A; Mann, JC; Reppas, C; Tack, J; Van Den Abeele, J; Vertzoni, M, 2020
)
0.78
" Based on pharmacokinetics, safety, and efficacy data, and PBPK modeling, dosing modifications for ribociclib recommend avoiding concurrent use of strong CYP3A inhibitors/inducers, and caution regarding using CYP3A substrates with narrow therapeutic indices."( Ribociclib Drug-Drug Interactions: Clinical Evaluations and Physiologically-Based Pharmacokinetic Modeling to Guide Drug Labeling.
Chakraborty, A; Dhuria, SV; Elmeliegy, M; He, H; Heimbach, T; Huth, F; Ji, Y; Miller, M; Samant, TS; Schiller, H; Umehara, K, 2020
)
0.56
" Current dosage guidelines recommend using cobicistat- or ritonavir-boosted darunavir 800 mg every 24 h (q24h) in protease inhibitor-naïve patients, or ritonavir-boosted darunavir 600 mg q12h in experienced patients."( Exploration of Reduced Doses and Short-Cycle Therapy for Darunavir/Cobicistat in Patients with HIV Using Population Pharmacokinetic Modeling and Simulations.
Belkhir, L; Elens, L; Haufroid, V; Stillemans, G; Vandercam, B; Vincent, A, 2021
)
0.87
"Diverging physicochemical properties of HIV drug combinations are challenging to formulate as a single dosage form."( Controlled Solvent Removal from Antiviral Drugs and Excipients in Solution Enables the Formation of Novel Combination Multi-Drug-Motifs in Pharmaceutical Powders Composed of Lopinavir, Ritonavir and Tenofovir.
Ho, RJY; Lane, S; McConnachie, L; Yu, D; Yu, J, 2020
)
0.75
"Pharmacokinetic boosting of antiretroviral (ARV) therapies with either ritonavir or cobicistat is used to achieve target drug exposure, lower pill burden, and provide simplified dosing schedules."( Pharmacokinetic Enhancement of HIV Antiretroviral Therapy During Pregnancy.
Best, BM; Eke, AC; Mirochnick, M; Momper, JD; Salama, E, 2020
)
0.79
" Surges in volume of patients requiring mechanical ventilation coupled with prolonged ventilator days and the high sedative dosing requirements observed quickly led to the depletion of "just-in-time" inventories typically maintained by institutions."( It Takes a Village…: Contending With Drug Shortages During Disasters.
Barletta, JF; Burry, LD; Christian, MD; Dichter, J; Erstad, BL; Geiling, J; Kanji, S; Maves, RC; Williamson, D, 2020
)
0.56
" (2) The patients were divided into normal dose group (500 mg, twice a day, n = 19) and reduced dose group (250 mg, twice a day, n = 5) according to the dosage of kaletra."( [Clinical characteristics of liver damage in 30 patients with severe coronavirus disease 2019 in Sichuan area].
Chen, H; Deng, C; Du, Q; Guo, Y; Huang, X; Li, T; Lu, S; Yue, R, 2020
)
0.56
" Dosage of ritonavir inhibits hepatic metabolization of the two prodrugs to different extents and decreases their efficacy in acting on TuMTs."( Predicting Metabolism-Related Drug-Drug Interactions Using a Microphysiological Multitissue System.
Bonanini, F; Frey, O; Hierlemann, A; Hoelting, L; Lohasz, C; Renggli, K, 2020
)
0.95
" Chloroquine (CQ) had a 100% discharge rate compared to 50% with lopinavir-ritonavir at day 14, however a trial has recommended against a high dosage due to cardiotoxic events."( Repurposing of drugs for COVID-19: a systematic review and meta-analysis.
Amparore, D; Checcucci, E; Dasgupta, P; Elhage, O; Fiori, C; Kotecha, P; Light, A; Porpiglia, F, 2022
)
0.95
" Therefore, in the absence of adverse drug reaction, lopinavir dosage should not be reduced."( Plasma Concentrations and Safety of Lopinavir/Ritonavir in COVID-19 Patients.
Azoulay, C; Batista, R; Benaboud, S; Boujaafar, S; Canouï, E; Carlier, N; Chouchana, L; Gana, I; Kernéis, S; Legendre, P; Preta, LH; Regard, L; Terrier, B; Treluyer, JM; Zerbit, J; Zheng, Y, 2021
)
0.88
"7%)) and incorrect dosing (n=6 (10."( Prescribing practices of lopinavir/ritonavir, hydroxychloroquine and azithromycin during the COVID-19 epidemic crisis and pharmaceutical interventions in a French teaching hospital.
Belmas, AS; Danion, F; Fourtage, M; Gourieux, B; Michel, B; Nai, T; Nivoix, Y; Reisz, F; Reiter-Schatz, A; Ruch, Y; Walther, J, 2021
)
0.9
" This review summarizes the current data for the most commonly used drugs for coronavirus disease 2019 and will cover the unique factors that may affect the dosing of these medications in patients with CKD."( Treatment Options for Coronavirus Disease 2019 in Patients With Reduced or Absent Kidney Function.
Govil, A; Luckett, K; Miller-Handley, H, 2020
)
0.56
" Lower white blood cell count and lactate dehydrogenase at the time of drug administration as well as shorter time from supplemental oxygen initiation to dosing were significantly associated with clinical improvement in the univariate analysis."( Early clinical outcomes with tocilizumab for severe COVID-19: a two-centre retrospective study.
Daniel, NM; Hilden, P; Raja, K; Smoke, SM, 2021
)
0.62
" DRV/r dosing was continued based on original study protocols."( The DIANA Study: Continued Access to Darunavir/Ritonavir (DRV/r) and Long-Term Safety Follow-Up in HIV-1-Infected Pediatric Patients Aged 3 to < 18 Years.
Blanche, S; Chetty, P; Hufkens, V; Masenya, M; Opsomer, M; Vanveggel, S; Violari, A, 2021
)
0.88
"PK and safety findings for ZDV, 3TC, and LPV/r support current WHO weight band dosing of syrup formulations in children with SAM."( Pharmacokinetics and Safety of Zidovudine, Lamivudine, and Lopinavir/Ritonavir in HIV-infected Children With Severe Acute Malnutrition in Sub-Saharan Africa: IMPAACT Protocol P1092.
Aweeka, F; Bradford, S; Browning, R; Coletti, A; Costello, D; Graham, B; Hughes, E; Kamthunzi, P; Kawalazira, R; Mmbaga, BT; Moye, J; Musoke, P; Nathoo, K; Norman, J; Owor, M; Purdue, L; Reding, C; Tierney, C; Whalen, ME; Wiesner, L; Ziemba, L, 2021
)
0.86
"Despite a collaborative effort towards developing suitable oral drug products for pediatrics over the past decade, appropriate pediatric dosage forms have remained lacking due to special considerations in dose flexibility, swallowability, palatability, and safety of excipients for pediatrics."( Development of nanoparticle-based orodispersible palatable pediatric formulations.
Deng, Y; Shen, J; Shen, L; Yang, Y, 2021
)
0.62
" However, should some hospitalized patients have dosage escalation to intermediate dose? Should some be considered for full-dose anticoagulation without a measurable thromboembolic event and how should that anticoagulation be monitored? Should patients receive postdischarge anticoagulation and with what medication and for how long? What thrombotic issues are related to the various medications being used to treat this coagulopathy? Is antiphospholipid antibody part of this syndrome? What is the significance of isolated ischemic stroke and limb ischemia in this disorder and how does this interface with the rest of the clinical and laboratory features of this disorder? The aims of this article are to explore these questions and interpret the available data based on the current evidence."( COVID-19 and Its Implications for Thrombosis and Anticoagulation.
Berkman, SA; Tapson, VF, 2021
)
0.62
" The mosquitoes could be dosed with a high precision (%CV: ≤13."( The pharmacokinetics and drug-drug interactions of ivermectin in Aedes aegypti mosquitoes.
Chaccour, C; Duthaler, U; Hammann, F; Hofer, L; Krähenbühl, S; Maia, M; Müller, P; Weber, M, 2021
)
0.62
" Trough plasma concentrations of lopinavir and ritonavir were higher than those expected, while those of hydroxychloroquine were those expected with the dosing regimen."( An open-label randomized controlled trial of the effect of lopinavir/ritonavir, lopinavir/ritonavir plus IFN-β-1a and hydroxychloroquine in hospitalized patients with COVID-19.
Ader, F; Alfaiate, T; Andrejak, C; Belhadi, D; Botelho-Nevers, E; Bouadma, L; Bouiller, K; Bouscambert-Duchamp, M; Burdet, C; Cabié, A; Clere-Jehl, R; Costagliola, D; Courjon, J; Danion, F; Dechanet, A; Delmas, C; Diallo, A; Dubost, C; Dupont, A; Epaulard, O; Faure, E; Gaci, R; Gagneux-Brunon, A; Gallien, S; Goehringer, F; Hites, M; Jaureguiberry, S; Kimmoun, A; Lacombe, K; Lanoix, JP; Launay, O; Lê, MP; Leroy, S; Lescure, FX; Lina, B; Makinson, A; Martin-Blondel, G; Mentré, F; Mercier, N; Mootien, J; Mourvillier, B; Navellou, JC; Noret, M; Nseir, S; Peiffer-Smadja, N; Peytavin, G; Piroth, L; Poissy, J; Pourcher, V; Raffi, F; Reignier, J; Reuter, J; Richard, JC; Saillard, J; Staub, T; Tolsma, V; Wallet, F; Yazdanpanah, Y, 2021
)
1.11
"1 ng/mL), resulting in contraception failure, when depot medroxyprogesterone is dosed at 12-week intervals."( A Semimechanistic Pharmacokinetic Model for Depot Medroxyprogesterone Acetate and Drug-Drug Interactions With Antiretroviral and Antituberculosis Treatment.
Cohn, SE; Denti, P; Dooley, KE; Firnhaber, C; Francis, J; Godfrey, C; Kendall, MA; McIlleron, H; Mngqibisa, R; Wu, X, 2021
)
0.62
"We conclude that despite availability of clinical evidence, double dosing LPV/r in patients receiving rifampicin-based TB treatment is low in Uganda's public HIV clinics but this does not seem to affect patient survival and viral suppression."( Adoption of evidence-informed guidelines in prescribing protease inhibitors for HIV-Tuberculosis co-infected patients on rifampicin and effects on HIV treatment outcomes in Uganda.
Castelnuovo, B; Haguma, P; Kalibbala, D; Kirenga, B; Muddu, M; Mulindwa, F; Semitala, FC, 2021
)
0.62
"This is an open-label, randomized, fixed sequence single intravenous dosing study to assess pharmacokinetic interactions between remdesivir and TDF/3TC (Study A, crossover design) or TDF/3TC plus ATV/r (Study B)."( An open-label, randomized, single intravenous dosing study to investigate the effect of fixed-dose combinations of tenofovir/lamivudine or atazanavir/ritonavir on the pharmacokinetics of remdesivir in Ugandan healthy volunteers (RemTLAR).
Byakika-Kibwika, P; D'Avolio, A; Kaboggoza, JP; Lamorde, M; Waitt, C; Walimbwa, SI, 2021
)
0.82
"The clinical guidelines suggest that the dosing of cyclosporine (CsA), during combination therapy with paritaprevir/ritonavir-ombitasvir and dasabuvir (PrOD), would be only one-fifth of the pre-PrOD total daily dose to be administered once daily."( Drug-Drug Interactions With Cyclosporine in the Anti-Hepatitis C Viral PrOD Combination Regimen of Paritaprevir/Ritonavir-Ombitasvir and Dasabuvir in Organ Transplant Recipients With Severe Hepatic Fibrosis or Cirrhosis.
Chang, YL; Chou, YC; Hsu, CC; Huang, YH; Huang, YY; Loong, CC; Wu, TH, 2022
)
1.14
" The authors proposed reducing the CsA dosage during PrOD treatment to one-seventh of that of the pre-PrOD treatment of the total daily dose to maintain target CsA levels."( Drug-Drug Interactions With Cyclosporine in the Anti-Hepatitis C Viral PrOD Combination Regimen of Paritaprevir/Ritonavir-Ombitasvir and Dasabuvir in Organ Transplant Recipients With Severe Hepatic Fibrosis or Cirrhosis.
Chang, YL; Chou, YC; Hsu, CC; Huang, YH; Huang, YY; Loong, CC; Wu, TH, 2022
)
0.93
" Our results show the clinically significant interaction between NR and immunosuppressive agents can be reasonably managed with a standardized dosing protocol."( Early clinical experience with nirmatrelvir/ritonavir for the treatment of COVID-19 in solid organ transplant recipients.
Brown, RS; Chen, JK; Hedvat, J; Jennings, DL; Kovac, DB; Lange, NW; Pereira, MR; Salerno, DM; Scheffert, J; Shertel, T, 2022
)
0.98
" Dosing strategies to reduce treatment costs are warranted."( Ritonavir-Boosted Exposure of Kinase Inhibitors: an Open Label, Cross-over Pharmacokinetic Proof-of-Concept Trial with Erlotinib.
Baas, P; Beijnen, JH; Boosman, RJ; Burgers, JA; de Gooijer, CJ; Groenland, SL; Huitema, ADR; Steeghs, N; van der Noort, V, 2022
)
2.16
" Complete pharmacokinetic profiles at steady-state were taken up to 24 h after erlotinib intake for both dosing strategies."( Ritonavir-Boosted Exposure of Kinase Inhibitors: an Open Label, Cross-over Pharmacokinetic Proof-of-Concept Trial with Erlotinib.
Baas, P; Beijnen, JH; Boosman, RJ; Burgers, JA; de Gooijer, CJ; Groenland, SL; Huitema, ADR; Steeghs, N; van der Noort, V, 2022
)
2.16
" Similar results regarding safety in both dosing strategies were observed, no grade 3 or higher adverse event was reported."( Ritonavir-Boosted Exposure of Kinase Inhibitors: an Open Label, Cross-over Pharmacokinetic Proof-of-Concept Trial with Erlotinib.
Baas, P; Beijnen, JH; Boosman, RJ; Burgers, JA; de Gooijer, CJ; Groenland, SL; Huitema, ADR; Steeghs, N; van der Noort, V, 2022
)
2.16
" We describe the population pharmacokinetics of ethambutol in children with TB to guide dosing in this population."( Population pharmacokinetics of ethambutol in African children: a pooled analysis.
Abdelwahab, MT; Andrieux-Meyer, I; Bekker, A; Chabala, C; Cotton, MF; Davies, G; Denti, P; Hesseling, A; Lee, J; McIlleron, H; Rabie, H; Tikiso, T; Wiesner, L; Zar, HJ, 2022
)
0.72
" Options for mitigating risk from DDIs with nirmatrelvir/ritonavir are limited due to the clinical illness, the short window for intervention, and the related difficulty of implementing clinical monitoring or dosage adjustment of the comedication."( Recommendations for the Management of Drug-Drug Interactions Between the COVID-19 Antiviral Nirmatrelvir/Ritonavir (Paxlovid) and Comedications.
Back, DJ; Boffito, M; Boyle, A; Burger, D; Flexner, C; Gibbons, S; Khoo, S; Kuritzkes, DR; Marra, F; Marzolini, C; Pozniak, A; Waters, L, 2022
)
1.18
" However, local safety profile should also be evaluated depending on the new indication, administration route and dosage form."( Antivirals and the Potential Benefits of Orally Inhaled Drug Administration in COVID-19 Treatment.
Akbal-Dagistan, O; Basarir, NS; Culha, M; Erturk, A; Sahin, G; Sancar, S; Yildiz-Pekoz, A, 2022
)
0.72
" Nirmatrelvir is the active component against the SARS-CoV-2 virus, whereas ritonavir, a potent CYP3A inhibitor, is intended to boost the activity of nirmatrelvir by increasing its concentration in plasma to ensure persistence of antiviral concentrations during the 12-hour dosing interval."( Clinically Relevant Interactions Between Ritonavir-Boosted Nirmatrelvir and Concomitant Antiseizure Medications: Implications for the Management of COVID-19 in Patients with Epilepsy.
Bialer, M; Caraco, Y; Levy, RH; Perucca, E; Wanounou, M, 2022
)
1.22
" Although a decrease in the dosage of ISDs can prevent toxicity, an inappropriate dosage regimen may also result in insufficient exposure and a risk of rejection."( Therapeutic Drug Monitoring and Dosage Adjustments of Immunosuppressive Drugs When Combined With Nirmatrelvir/Ritonavir in Patients With COVID-19.
Bergan, S; Brunet, M; Budde, K; De Winter, BCM; Elens, L; Hesselink, DA; Johnson-Davis, KL; Langman, LJ; Lawson, R; Lemaitre, F; Masuda, S; Moes, DJAR; Noceti, O; Pattanaik, S; Pawinski, T; Van Gelder, T; Venkataramanan, R, 2023
)
1.12
" The established correlation equations can be useful in therapeutic drug monitoring (TDM) and dosing regimen optimization."( The investigation of the complex population-drug-drug interaction between ritonavir-boosted lopinavir and chloroquine or ivermectin using physiologically-based pharmacokinetic modeling.
Alsmadi, MM, 2023
)
1.14
", a hyperimmune anti-COVID-19 intravenous immunoglobulin), methylprednisolone, interferon-beta/standard-of-care (SOC), interferon-beta-1b, convalescent plasma, remdesivir, lopinavir/ritonavir, immunoglobulin gamma, high dosage sarilumab (HS), auxora, and imatinib were effective when compared with placebo or SOC group."( Comparative efficacy and safety of pharmacological interventions for severe COVID-19 patients: An updated network meta-analysis of 48 randomized controlled trials.
Chen, J; Cheng, Q; Fang, Z; Jia, Q; Zhao, G, 2022
)
0.91
" This study aimed to develop a population pharmacokinetic model of LPV/r and provide dosage optimization in Thai PLWH."( Dose optimization with population pharmacokinetics of ritonavir-boosted lopinavir for Thai people living with HIV with and without active tuberculosis.
Avihingsanon, A; Chaivichacharn, P; Gatechompol, S; Punyawudho, B; Ubolyam, S, 2022
)
0.97
" The reduced LPV/r dosage offers sufficient LPV exposure for Thai PLWH."( Dose optimization with population pharmacokinetics of ritonavir-boosted lopinavir for Thai people living with HIV with and without active tuberculosis.
Avihingsanon, A; Chaivichacharn, P; Gatechompol, S; Punyawudho, B; Ubolyam, S, 2022
)
0.97
"6% probability of improving 6-month survival across varying hydrocortisone dosing strategies."( Long-term (180-Day) Outcomes in Critically Ill Patients With COVID-19 in the REMAP-CAP Randomized Clinical Trial.
Al-Beidh, F; Angus, DC; Annane, D; Arabi, YM; Beane, A; Berry, LR; Berry, S; Bhimani, Z; Bonten, MJM; Bradbury, CA; Brunkhorst, FM; Burrell, A; Buxton, M; Buzgau, A; Charles, WN; Cove, M; Derde, LPG; Detry, MA; Estcourt, LJ; Fagbodun, EO; Fitzgerald, M; Girard, TD; Goligher, EC; Goossens, H; Gordon, AC; Green, C; Haniffa, R; Higgins, AM; Hills, T; Horvat, CM; Huang, DT; Ichihara, N; Lamontagne, F; Lawler, PR; Lewis, RJ; Lorenzi, E; Marshall, JC; McArthur, CJ; McAuley, DF; McGlothlin, A; McGuinness, SP; McQuilten, Z; McVerry, BJ; Mouncey, PR; Murthy, S; Neal, MD; Nichol, AD; Parke, RL; Parker, JC; Parry-Billings, K; Peters, SEC; Reyes, LF; Rowan, KM; Saito, H; Santos, MS; Saunders, CT; Serpa-Neto, A; Seymour, CW; Shankar-Hari, M; Stronach, LM; Turgeon, AF; Turner, AM; van Bentum-Puijk, W; van de Veerdonk, FL; Webb, SA; Zarychanski, R, 2023
)
0.91
"The formulation development of amorphous solid dispersions (ASDs) towards a patient-friendly oral solid dosage form is proving to be still challenging."( Downstream processing of amorphous solid dispersions into orodispersible tablets.
Breitkreutz, J; Klinken, S; Kokott, M; Wiedey, R, 2023
)
0.91
"In order to overcome potentially suboptimal therapeutic exposures, nirmatrelvir is dosed in combination with ritonavir to boost the pharmacokinetics of the active product."( A commentary on the use of pharmacoenhancers in the pharmaceutical industry and the implication for DMPK drug discovery strategies.
Fazal, L; Martins, V; Oganesian, A; Shah, A; Stow, J; Walton, H; Wilsher, N, 2022
)
0.93
"NMV/r has significant DDIs with many common dermatological medications, which may require temporary discontinuation, dosage adjustment or substitution with other anti-COVID-19 agents such as molnupiravir."( Drug interactions between common dermatological medications and the oral anti-COVID-19 agents nirmatrelvir-ritonavir and molnupiravir.
Huang, X; Oon, HH; Quah, KSE; Renia, L, 2022
)
0.93
"0 μg/mL in the pure form and in pharmaceutical dosage form."( Adjusted green HPLC determination of nirmatrelvir and ritonavir in the new FDA approved co-packaged pharmaceutical dosage using supported computational calculations.
Abdelazim, AH; Almrasy, AA; Batubara, AS; Gamal, M; Imam, MS; Ramzy, S, 2023
)
1.16
"Although super-boosted lopinavir/ritonavir (LPV/r; ratio 4:4 instead of 4:1) is recommended for infants living with HIV and receiving concomitant rifampicin, in clinical practice, many different LPV/r dosing strategies are applied due to poor availability of pediatric separate ritonavir formulations needed to superboost."( Brief Report: Suboptimal Lopinavir Exposure in Infants on Rifampicin Treatment Receiving Double-dosed or Semisuperboosted Lopinavir/Ritonavir: Time for a Change.
Ballesteros, A; Buck, WC; Burger, DM; Chabala, C; Chitsamatanga, M; Colbers, A; Domínguez-Rodríguez, S; Jacobs, TG; Madrid, L; Moraleda, C; Mujuru, HA; Mumbiro, V; Namuziya, N; Nathoo, KJ; Nduna, B; Passanduca, A; Rojo, P; Tagarro, A, 2023
)
1.4
" These data may support clinicians to make more informed dosing decisions for patients with cancer undergoing treatment with KIs who require treatment with ritonavir-containing COVID-19 antiviral therapies."( Understanding the Risk of Drug Interactions Between Ritonavir-Containing COVID-19 Therapies and Small-Molecule Kinase Inhibitors in Patients With Cancer.
Hopkins, AM; Karapetis, CS; McLachlan, AJ; Miners, JO; Rowland, A; Sorich, MJ; van Dyk, M, 2023
)
1.36
" Nirmatrelvir plus ritonavir was recently approved in the US, the UK and Europe as a new co-packaged dosage form for the treatment of COVID-19."( Simultaneous green TLC determination of nirmatrelvir and ritonavir in the pharmaceutical dosage form and spiked human plasma.
Abdelazim, AH; Almrasy, AA; Batubara, AS; Gamal, M; Hasanin, THA; Imam, MS; Khojah, H; Ramzy, S, 2023
)
1.48
" Therefore, optimization of tacrolimus dosing is urgently needed in transplant recipients receiving nirmatrelvir/ritonavir treatment."( Nirmatrelvir/ritonavir treatment in SARS-CoV-2 positive kidney transplant recipients - a case series with four patients.
Kühn, W; Schneider, J; Tanriver, Y; Wagner, D; Walz, G; Wobser, R, 2023
)
1.49
" Further studies are needed to determine the optimal dosing strategies of nirmatrelvir/ritonavir, immunosuppressant adjustment, and monitoring in this patient population."( Optimizing the use of nirmatrelvir/ritonavir in solid organ transplant recipients with COVID-19: A review of immunosuppressant adjustment strategies.
Li, Y; Song, T; Tang, Y, 2023
)
1.41
" Once submitted, the pharmacist would identify which oral COVID-19 medication and dosage was most appropriate."( Facilitating oral COVID-19 therapy utilization through a pharmacy consult service.
Portman, DB; Scolese, CJ,
)
0.13
" Nirmatrelvir/ritonavir was the predominant agent used with 16% requiring renal dosage adjustment."( Facilitating oral COVID-19 therapy utilization through a pharmacy consult service.
Portman, DB; Scolese, CJ,
)
0.49
" Clearance and volume of distribution were derived from nirmatrelvir PK obtained using a spray-dried dispersion (SDD) formulation where it is considered to be dosed as an oral solution, and absorption is near complete."( Physiologically-Based Pharmacokinetic Modeling of PAXLOVID™ with First-Order Absorption Kinetics.
Di, L; Jaini, R; Lin, J; Sagawa, K, 2023
)
0.91
" Daily emtricitabine/TAF was dosed according to World Health Organization (WHO)-recommended weight bands: 120/15 mg in children weighing 14 to <25 kg and 200/25 mg in those weighing ≥25 kg."( First Pharmacokinetic Data of Tenofovir Alafenamide Fumarate and Tenofovir With Dolutegravir or Boosted Protease Inhibitors in African Children: A Substudy of the CHAPAS-4 Trial.
Bamford, A; Burger, DM; Bwakura-Dangarembizi, M; Chabala, C; Colbers, A; Denti, P; Doerholt, K; Gibb, DM; Griffiths, AL; Makumbi, S; McIlleron, HM; Monkiewicz, LN; Mulenga, V; Mumbiro, V; Musiime, V; Nangiya, J; Szubert, AJ; Waalewijn, H; Wasmann, RE; Wiesner, L, 2023
)
0.91
"In children, TAF combined with boosted PIs or dolutegravir and dosed according to WHO-recommended weight bands provides TAF and tenofovir concentrations previously demonstrated to be well tolerated and effective in adults."( First Pharmacokinetic Data of Tenofovir Alafenamide Fumarate and Tenofovir With Dolutegravir or Boosted Protease Inhibitors in African Children: A Substudy of the CHAPAS-4 Trial.
Bamford, A; Burger, DM; Bwakura-Dangarembizi, M; Chabala, C; Colbers, A; Denti, P; Doerholt, K; Gibb, DM; Griffiths, AL; Makumbi, S; McIlleron, HM; Monkiewicz, LN; Mulenga, V; Mumbiro, V; Musiime, V; Nangiya, J; Szubert, AJ; Waalewijn, H; Wasmann, RE; Wiesner, L, 2023
)
0.91
"Determining the appropriate dosing regimens of antiretroviral (ARV) drugs for pregnant individuals living with HIV-1 infection is critical to maximize maternal health and prevent perinatal HIV transmission."( Pharmacokinetics of Antiretroviral Agents in Pregnant Individuals Living With HIV: Current Status and Considerations for Study Design and Interpretation.
Belew, Y; Choi, SY; Reynolds, K; Struble, K; Yang, X, 2023
)
0.91
" We evaluated whether increased daily dosing with modified 8-hourly lopinavir/ritonavir 4:1 would maintain therapeutic plasma concentrations of lopinavir in children living with HIV receiving rifampicin-based antituberculosis treatment."( Inadequate Lopinavir Concentrations With Modified 8-Hourly Lopinavir/Ritonavir 4:1 Dosing During Rifampicin-based Tuberculosis Treatment in Children Living With HIV.
Chabala, C; Choo, L; Chungu, C; Crook, A; Gibb, D; Kapasa, M; LeBeau, K; Lungu, J; McIlleron, H; Mulenga, V; Tembo, CH; Turkova, A; Weisner, L; Zimba, K; Zyambo, K, 2023
)
1.37
"9 kg, on rifampicin-based antituberculosis treatment were commenced or switched to 8-hourly liquid lopinavir/ritonavir 4:1 with increased daily dosing using weight-band dosing approach."( Inadequate Lopinavir Concentrations With Modified 8-Hourly Lopinavir/Ritonavir 4:1 Dosing During Rifampicin-based Tuberculosis Treatment in Children Living With HIV.
Chabala, C; Choo, L; Chungu, C; Crook, A; Gibb, D; Kapasa, M; LeBeau, K; Lungu, J; McIlleron, H; Mulenga, V; Tembo, CH; Turkova, A; Weisner, L; Zimba, K; Zyambo, K, 2023
)
1.36
"7 mg/L]) were lower than on standard dosing without rifampicin (n = 12; area under the curve 24 121."( Inadequate Lopinavir Concentrations With Modified 8-Hourly Lopinavir/Ritonavir 4:1 Dosing During Rifampicin-based Tuberculosis Treatment in Children Living With HIV.
Chabala, C; Choo, L; Chungu, C; Crook, A; Gibb, D; Kapasa, M; LeBeau, K; Lungu, J; McIlleron, H; Mulenga, V; Tembo, CH; Turkova, A; Weisner, L; Zimba, K; Zyambo, K, 2023
)
1.14
"Modified 8-hourly dosing of lopinavir/ritonavir failed to achieve adequate lopinavir concentrations with concurrent antituberculosis treatment."( Inadequate Lopinavir Concentrations With Modified 8-Hourly Lopinavir/Ritonavir 4:1 Dosing During Rifampicin-based Tuberculosis Treatment in Children Living With HIV.
Chabala, C; Choo, L; Chungu, C; Crook, A; Gibb, D; Kapasa, M; LeBeau, K; Lungu, J; McIlleron, H; Mulenga, V; Tembo, CH; Turkova, A; Weisner, L; Zimba, K; Zyambo, K, 2023
)
1.42
" Paxlovid, a co-packaged dosage form of two antiviral drugs (nirmatrelvir and ritonavir) developed by Pfizer, received its first FDA Emergency Use Authorization (EUA) and conditional marketing by European Medical Agency in patients at high risk of developing severe COVID-19."( Nirmatrelvir and ritonavir combination: an antiviral therapy for COVID-19.
Jogvanshi, A; Naikwadi, S; Navitha Reddy, G; Sonti, R,
)
0.7
" Female ferrets dosed with 20 or 100 mg/kg nirmatrelvir/ritonavir twice-daily show 1-2 log order reduction of viral RNA copies and infectious titers, which correlates with low nirmatrelvir exposure in nasal turbinates."( Comparing molnupiravir and nirmatrelvir/ritonavir efficacy and the effects on SARS-CoV-2 transmission in animal models.
Andrews, MK; Cox, RM; Greninger, AL; Karimi, A; Kolykhalov, AA; Krueger, RE; Lieber, CM; Lieberman, NAP; Natchus, MG; Painter, GR; Plemper, RK; Roychoudhury, P; Sticher, ZM; Wolf, JD, 2023
)
1.42
" The prospective simulations not only provided scientific evidence regarding rational dosing management strategies when initiating nirmatrelvir/ritonavir therapy but also provided a reference for the design of clinical DDIs study that may save resources and time."( Drug-drug interactions and dose management of BTK inhibitors when initiating nirmatrelvir/ritonavir (paxlovid) based on physiologically-based pharmacokinetic models.
Chen, L; Chen, W; Li, C; Li, L, 2023
)
1.33
" These drugs are typically available in liquid formulations to aid in dosing for children who cannot swallow tablets."( Anti-HIV drugs lopinavir/ritonavir activate bitter taste receptors.
Chen, S; Cui, M; Lu, Y; Wen, J; Xu, K; Zhou, X, 2023
)
1.21
" Rational dosage adjustment obtained good tolerance but did not influence the efficacy."( Nirmatrelvir/ritonavir use in patients with COVID-19 on hemodialysis: a case series.
Diao, ZL; Guan, YM; Huang, HD; Liu, WH; Liu, YJ; Tian, DL; Wang, G; Zhang, AH; Zhao, YC, 2023
)
1.28
"The therapeutic approach involved a 5-day regimen of nirmatrelvir/ritonavir at a dosage of 300/100 mg administered twice daily, along with a daily dosage of 6 mg of dexamethasone."( Successful utilization of nirmatrelvir/ritonavir and dexamethasone in a patient with total artificial heart and COVID-19: A case report.
Ahmed, H; Alghamdi, AA; Alowais, SA; AlQahtani, H; Badreldin, HA; Bosaeed, M; Hussain, A; Saleh, KB; Selimovic, N, 2023
)
1.42
"The current World Health Organization (WHO) pediatric tuberculosis dosing guidelines lead to suboptimal drug exposures."( Evaluating pediatric tuberculosis dosing guidelines: A model-based individual data pooled analysis.
Aarnoutse, R; Chabala, C; Cotton, MF; Denti, P; Galileya, LT; Gibb, D; Hesseling, A; Lee, J; McIlleron, H; Njahira Mukui, I; Rabie, H; Turkova, A; Wasmann, RE; Zar, H, 2023
)
0.91
" The importance of the drug-drug interactions with lopinavir/ritonavir and efavirenz should be evaluated further and considered in future dosing guidance."( Evaluating pediatric tuberculosis dosing guidelines: A model-based individual data pooled analysis.
Aarnoutse, R; Chabala, C; Cotton, MF; Denti, P; Galileya, LT; Gibb, D; Hesseling, A; Lee, J; McIlleron, H; Njahira Mukui, I; Rabie, H; Turkova, A; Wasmann, RE; Zar, H, 2023
)
1.15
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Roles (4)

RoleDescription
antiviral drugA substance used in the prophylaxis or therapy of virus diseases.
HIV protease inhibitorAn inhibitor of HIV protease, an enzyme required for production of proteins needed for viral assembly.
environmental contaminantAny minor or unwanted substance introduced into the environment that can have undesired effects.
xenobioticA xenobiotic (Greek, xenos "foreign"; bios "life") is a compound that is foreign to a living organism. Principal xenobiotics include: drugs, carcinogens and various compounds that have been introduced into the environment by artificial means.
[role information is derived from Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Drug Classes (5)

ClassDescription
1,3-thiazoles
L-valine derivativeA proteinogenic amino acid derivative resulting from reaction of L-valine at the amino group or the carboxy group, or from the replacement of any hydrogen of L-valine by a heteroatom.
carbamate esterAny ester of carbamic acid or its N-substituted derivatives.
ureas
carboxamideAn amide of a carboxylic acid, having the structure RC(=O)NR2. The term is used as a suffix in systematic name formation to denote the -C(=O)NH2 group including its carbon atom.
[compound class information is derived from Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Protein Targets (100)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Chain A, Beta-lactamaseEscherichia coli K-12Potency7.07950.044717.8581100.0000AID485341
Chain A, Ferritin light chainEquus caballus (horse)Potency31.62285.623417.292931.6228AID485281
Chain A, CruzipainTrypanosoma cruziPotency39.81070.002014.677939.8107AID1476
LuciferasePhotinus pyralis (common eastern firefly)Potency19.69320.007215.758889.3584AID588342; AID624030
SMAD family member 2Homo sapiens (human)Potency23.91450.173734.304761.8120AID1346924
Fumarate hydrataseHomo sapiens (human)Potency37.22120.00308.794948.0869AID1347053
GLS proteinHomo sapiens (human)Potency31.62280.35487.935539.8107AID624170
SMAD family member 3Homo sapiens (human)Potency23.91450.173734.304761.8120AID1346924
TDP1 proteinHomo sapiens (human)Potency18.96250.000811.382244.6684AID686978; AID686979
GLI family zinc finger 3Homo sapiens (human)Potency5.62260.000714.592883.7951AID1259392
Microtubule-associated protein tauHomo sapiens (human)Potency39.81070.180013.557439.8107AID1460
AR proteinHomo sapiens (human)Potency28.32890.000221.22318,912.5098AID743035; AID743036; AID743042; AID743054; AID743063
aldehyde dehydrogenase 1 family, member A1Homo sapiens (human)Potency39.81070.011212.4002100.0000AID1030
estrogen receptor 2 (ER beta)Homo sapiens (human)Potency16.89680.000657.913322,387.1992AID1259377; AID1259378
progesterone receptorHomo sapiens (human)Potency9.43920.000417.946075.1148AID1346795
cytochrome P450 family 3 subfamily A polypeptide 4Homo sapiens (human)Potency0.01740.01237.983543.2770AID1645841
EWS/FLI fusion proteinHomo sapiens (human)Potency17.45840.001310.157742.8575AID1259253; AID1259255; AID1259256
glucocorticoid receptor [Homo sapiens]Homo sapiens (human)Potency23.91450.000214.376460.0339AID720692
retinoid X nuclear receptor alphaHomo sapiens (human)Potency15.79690.000817.505159.3239AID1159527; AID1159531
estrogen-related nuclear receptor alphaHomo sapiens (human)Potency22.90830.001530.607315,848.9004AID1224841; AID1224842; AID1224848; AID1224849; AID1259401; AID1259403
farnesoid X nuclear receptorHomo sapiens (human)Potency33.48890.375827.485161.6524AID743217
pregnane X nuclear receptorHomo sapiens (human)Potency5.95570.005428.02631,258.9301AID1346982
estrogen nuclear receptor alphaHomo sapiens (human)Potency18.49590.000229.305416,493.5996AID743069; AID743075; AID743078; AID743079
GVesicular stomatitis virusPotency0.61660.01238.964839.8107AID1645842
cytochrome P450 2D6Homo sapiens (human)Potency4.89750.00108.379861.1304AID1645840
polyproteinZika virusPotency37.22120.00308.794948.0869AID1347053
peroxisome proliferator-activated receptor deltaHomo sapiens (human)Potency5.95530.001024.504861.6448AID743212
peroxisome proliferator activated receptor gammaHomo sapiens (human)Potency21.35740.001019.414170.9645AID743094; AID743140; AID743191
alpha-galactosidaseHomo sapiens (human)Potency50.11874.466818.391635.4813AID1467
aryl hydrocarbon receptorHomo sapiens (human)Potency13.33320.000723.06741,258.9301AID743085
cytochrome P450, family 19, subfamily A, polypeptide 1, isoform CRA_aHomo sapiens (human)Potency33.49150.001723.839378.1014AID743083
activating transcription factor 6Homo sapiens (human)Potency26.83250.143427.612159.8106AID1159516
vitamin D3 receptor isoform VDRAHomo sapiens (human)Potency28.18380.354828.065989.1251AID504847
thyroid hormone receptor beta isoform 2Rattus norvegicus (Norway rat)Potency16.81820.000323.4451159.6830AID743065; AID743067
mitogen-activated protein kinase 1Homo sapiens (human)Potency3.98110.039816.784239.8107AID1454
nuclear factor erythroid 2-related factor 2 isoform 1Homo sapiens (human)Potency20.86470.000627.21521,122.0200AID743202; AID743219
nuclear receptor ROR-gamma isoform 1Mus musculus (house mouse)Potency14.12540.00798.23321,122.0200AID2551
DNA polymerase kappa isoform 1Homo sapiens (human)Potency39.81070.031622.3146100.0000AID588579
histone acetyltransferase KAT2A isoform 1Homo sapiens (human)Potency28.18380.251215.843239.8107AID504327
neuropeptide S receptor isoform AHomo sapiens (human)Potency12.58930.015812.3113615.5000AID1461
Interferon betaHomo sapiens (human)Potency0.61660.00339.158239.8107AID1645842
HLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)Potency0.61660.01238.964839.8107AID1645842
Cellular tumor antigen p53Homo sapiens (human)Potency7.49780.002319.595674.0614AID651631
Inositol hexakisphosphate kinase 1Homo sapiens (human)Potency0.61660.01238.964839.8107AID1645842
ATPase family AAA domain-containing protein 5Homo sapiens (human)Potency9.43920.011917.942071.5630AID651632
Ataxin-2Homo sapiens (human)Potency9.43920.011912.222168.7989AID651632
cytochrome P450 2C9, partialHomo sapiens (human)Potency0.61660.01238.964839.8107AID1645842
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Inhibition Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Chain A, HIV-1 PROTEASEHuman immunodeficiency virus 1Ki0.00000.00000.00000.0000AID977610
Chain B, HIV-1 PROTEASEHuman immunodeficiency virus 1Ki0.00000.00000.00000.0000AID977610
Chain A, POL polyproteinHuman immunodeficiency virus 1Ki0.93200.93202.58354.2350AID977610
Chain B, POL polyproteinHuman immunodeficiency virus 1Ki0.93200.93202.58354.2350AID977610
Chain A, POL polyproteinHuman immunodeficiency virus 1Ki0.93200.93202.58354.2350AID977610
Chain B, POL polyproteinHuman immunodeficiency virus 1Ki0.93200.93202.58354.2350AID977610
Chain A, EndothiapepsinCryphonectria parasitica (chestnut blight fungus)Ki0.01500.01500.01500.0150AID977610
Chain A, EndothiapepsinCryphonectria parasitica (chestnut blight fungus)Ki0.01500.01500.01500.0150AID977610
Solute carrier family 22 member 2Homo sapiens (human)IC50 (µMol)24.80000.40003.10009.7000AID721751
Solute carrier family 22 member 1 Homo sapiens (human)IC50 (µMol)33.90000.21005.553710.0000AID721750
ATP-binding cassette sub-family C member 3Homo sapiens (human)IC50 (µMol)11.10000.63154.45319.3000AID1473740
Multidrug resistance-associated protein 4Homo sapiens (human)IC50 (µMol)34.00000.20005.677410.0000AID1473741
Solute carrier family 22 member 3Homo sapiens (human)IC50 (µMol)300.00000.09003.72779.5000AID721749
Solute carrier organic anion transporter family member 2B1 Homo sapiens (human)IC50 (µMol)6.10000.55003.70836.1000AID699544
Solute carrier organic anion transporter family member 2B1 Homo sapiens (human)Ki5.90000.53004.11578.4800AID699544
Bile salt export pumpHomo sapiens (human)IC50 (µMol)2.08500.11007.190310.0000AID1443980; AID1443989; AID1473738; AID1674183
Gag-Pol polyproteinHuman immunodeficiency virus type 1 (BRU ISOLATE)Ki0.00430.00000.08283.3000AID1795271; AID1796305; AID1797110
Gag-Pol polyproteinHIV-1 M:B_ARV2/SF2Ki0.00210.00000.01090.0895AID1797666
Gag-Pol polyproteinHIV-1 M:B_HXB2RKi0.39320.00000.51449.0000AID1795276; AID1797107
Cytochrome P450 1A2Homo sapiens (human)IC50 (µMol)25.00000.00011.774010.0000AID1069010
Gag-Pol polyproteinHuman immunodeficiency virus type 1 (RF/HAT ISOLATE)Ki0.00690.00000.05051.6160AID1797110
ATP-dependent translocase ABCB1Mus musculus (house mouse)IC50 (µMol)50.00000.06404.012610.0000AID150754; AID681128
ATP-dependent translocase ABCB1Homo sapiens (human)IC50 (µMol)14.10000.00022.318510.0000AID150752; AID150755; AID416864; AID679892; AID681122; AID681358
Cytochrome P450 3A4Homo sapiens (human)IC50 (µMol)0.16230.00011.753610.0000AID1069013; AID1657049; AID470602; AID54752; AID54923; AID625251; AID738134
Cytochrome P450 3A4Homo sapiens (human)Ki0.10000.00011.41629.9000AID520315; AID589111
Replicase polyprotein 1abSevere acute respiratory syndrome coronavirus 2IC50 (µMol)20.00000.00022.45859.9600AID1845236
Cytochrome P450 2D6Homo sapiens (human)IC50 (µMol)26.15000.00002.015110.0000AID1069009; AID625249
Cytochrome P450 2C9 Homo sapiens (human)IC50 (µMol)2.45800.00002.800510.0000AID1069008; AID625248
Gag-Pol polyproteinHuman immunodeficiency virus type 1 (NEW YORK-5 ISOLATE)IC50 (µMol)0.06700.00020.10421.7000AID1796876
Alpha-1B adrenergic receptorRattus norvegicus (Norway rat)IC50 (µMol)39.60000.00021.874210.0000AID416864
Cytochrome P450 3A5Homo sapiens (human)Ki0.12000.02202.60407.6000AID589167
ATP-dependent translocase ABCB1Mus musculus (house mouse)IC50 (µMol)50.00000.20004.713010.0000AID150753; AID681119
Substance-K receptorHomo sapiens (human)IC50 (µMol)2.62200.00013.12109.5530AID625227
Substance-K receptorHomo sapiens (human)Ki0.87400.00011.92429.7930AID625227
UDP-glucuronosyltransferase 1A1 Homo sapiens (human)IC50 (µMol)2.35000.30003.25807.3000AID1222388; AID1222389
Alpha-1D adrenergic receptorRattus norvegicus (Norway rat)IC50 (µMol)39.60000.00021.270410.0000AID416864
Thromboxane-A synthase Homo sapiens (human)IC50 (µMol)0.07600.00091.230410.0000AID625229
Gag-Pol polyproteinHuman immunodeficiency virus type 1 (STRAIN UGANDAN / ISOLATE U455)Ki0.00690.00000.05051.6160AID1797110
Cytochrome P450 2C19Homo sapiens (human)IC50 (µMol)12.70000.00002.398310.0000AID1069011
Mu-type opioid receptorHomo sapiens (human)IC50 (µMol)65.54200.00010.813310.0000AID625163
Mu-type opioid receptorHomo sapiens (human)Ki26.60600.00000.419710.0000AID625163
Vasopressin V1a receptorHomo sapiens (human)IC50 (µMol)12.41600.00060.38352.0000AID625233
Vasopressin V1a receptorHomo sapiens (human)Ki4.98100.00020.62357.0300AID625233
Kappa-type opioid receptorHomo sapiens (human)IC50 (µMol)13.65500.00001.201110.0000AID625162
Kappa-type opioid receptorHomo sapiens (human)Ki5.46200.00000.362410.0000AID625162
Alpha-1A adrenergic receptorRattus norvegicus (Norway rat)IC50 (µMol)39.60000.00001.819410.0000AID416864
Protease Human immunodeficiency virus 1Ki0.00900.00000.04433.1000AID160300; AID160316; AID160455; AID160481; AID162710; AID163487; AID219260; AID225562; AID225563; AID225564; AID238682; AID82295; AID82944; AID82946
Multidrug and toxin extrusion protein 2Homo sapiens (human)IC50 (µMol)14.05000.16003.95718.6000AID721748; AID721752
Canalicular multispecific organic anion transporter 1Homo sapiens (human)IC50 (µMol)133.00002.41006.343310.0000AID1473739
Multidrug and toxin extrusion protein 1Homo sapiens (human)IC50 (µMol)2.19330.01002.765610.0000AID721746; AID721747; AID721754
Solute carrier organic anion transporter family member 1B3Homo sapiens (human)IC50 (µMol)4.40000.10472.71957.0795AID699543
Solute carrier organic anion transporter family member 1B3Homo sapiens (human)Ki4.00000.08002.46889.8000AID699543
Broad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)IC50 (µMol)19.50000.00401.966610.0000AID679171
Solute carrier organic anion transporter family member 1B1Homo sapiens (human)IC50 (µMol)2.00000.05002.37979.7000AID1218863; AID1218864; AID1218865; AID699542
Solute carrier organic anion transporter family member 1B1Homo sapiens (human)Ki1.30000.04401.36305.0000AID699542
Protease Human immunodeficiency virus 1IC50 (µMol)4.92530.00000.81769.8500AID1069006; AID738316
Protease Human immunodeficiency virus 1Ki0.00030.00000.02841.1000AID321660; AID374591; AID443165
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Activation Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Nuclear receptor subfamily 1 group I member 2Homo sapiens (human)EC50 (µMol)4.00000.00203.519610.0000AID1215086
Protease Human immunodeficiency virus 1Kd0.00060.00010.04120.5770AID162706; AID238043
Protease Human immunodeficiency virus 1EC50 (µMol)0.15000.00070.69422.7300AID1498796
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Other Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
AlbuminHomo sapiens (human)KD2189.00006.00006.00006.0000AID239810
Cytochrome P450 3A4Homo sapiens (human)Ks0.09000.01900.21000.6900AID1657045; AID738133; AID738136; AID738139
Thromboxane-A synthaseRattus norvegicus (Norway rat)IC90 (µMol)0.07600.01040.07030.1250AID210277
Protease Human immunodeficiency virus 1ED500.81400.00140.41032.4700AID104301; AID104302; AID104303; AID104304; AID104312
Protease Human immunodeficiency virus 1IC90 (µMol)0.12530.00200.67847.3000AID105206; AID210277; AID82294
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (436)

Processvia Protein(s)Taxonomy
activation of cysteine-type endopeptidase activity involved in apoptotic processSolute carrier family 22 member 2Homo sapiens (human)
positive regulation of gene expressionSolute carrier family 22 member 2Homo sapiens (human)
organic cation transportSolute carrier family 22 member 2Homo sapiens (human)
monoatomic cation transportSolute carrier family 22 member 2Homo sapiens (human)
neurotransmitter transportSolute carrier family 22 member 2Homo sapiens (human)
serotonin transportSolute carrier family 22 member 2Homo sapiens (human)
body fluid secretionSolute carrier family 22 member 2Homo sapiens (human)
organic cation transportSolute carrier family 22 member 2Homo sapiens (human)
quaternary ammonium group transportSolute carrier family 22 member 2Homo sapiens (human)
prostaglandin transportSolute carrier family 22 member 2Homo sapiens (human)
amine transportSolute carrier family 22 member 2Homo sapiens (human)
putrescine transportSolute carrier family 22 member 2Homo sapiens (human)
spermidine transportSolute carrier family 22 member 2Homo sapiens (human)
acetylcholine transportSolute carrier family 22 member 2Homo sapiens (human)
choline transportSolute carrier family 22 member 2Homo sapiens (human)
dopamine transportSolute carrier family 22 member 2Homo sapiens (human)
norepinephrine transportSolute carrier family 22 member 2Homo sapiens (human)
xenobiotic transportSolute carrier family 22 member 2Homo sapiens (human)
epinephrine transportSolute carrier family 22 member 2Homo sapiens (human)
histamine transportSolute carrier family 22 member 2Homo sapiens (human)
serotonin uptakeSolute carrier family 22 member 2Homo sapiens (human)
histamine uptakeSolute carrier family 22 member 2Homo sapiens (human)
norepinephrine uptakeSolute carrier family 22 member 2Homo sapiens (human)
thiamine transmembrane transportSolute carrier family 22 member 2Homo sapiens (human)
purine-containing compound transmembrane transportSolute carrier family 22 member 2Homo sapiens (human)
amino acid import across plasma membraneSolute carrier family 22 member 2Homo sapiens (human)
dopamine uptakeSolute carrier family 22 member 2Homo sapiens (human)
L-arginine import across plasma membraneSolute carrier family 22 member 2Homo sapiens (human)
export across plasma membraneSolute carrier family 22 member 2Homo sapiens (human)
transport across blood-brain barrierSolute carrier family 22 member 2Homo sapiens (human)
L-alpha-amino acid transmembrane transportSolute carrier family 22 member 2Homo sapiens (human)
spermidine transmembrane transportSolute carrier family 22 member 2Homo sapiens (human)
L-arginine transmembrane transportSolute carrier family 22 member 2Homo sapiens (human)
cellular detoxificationSolute carrier family 22 member 2Homo sapiens (human)
xenobiotic transport across blood-brain barrierSolute carrier family 22 member 2Homo sapiens (human)
xenobiotic metabolic processSolute carrier family 22 member 1 Homo sapiens (human)
neurotransmitter transportSolute carrier family 22 member 1 Homo sapiens (human)
serotonin transportSolute carrier family 22 member 1 Homo sapiens (human)
establishment or maintenance of transmembrane electrochemical gradientSolute carrier family 22 member 1 Homo sapiens (human)
organic cation transportSolute carrier family 22 member 1 Homo sapiens (human)
quaternary ammonium group transportSolute carrier family 22 member 1 Homo sapiens (human)
prostaglandin transportSolute carrier family 22 member 1 Homo sapiens (human)
monoamine transportSolute carrier family 22 member 1 Homo sapiens (human)
putrescine transportSolute carrier family 22 member 1 Homo sapiens (human)
spermidine transportSolute carrier family 22 member 1 Homo sapiens (human)
acetylcholine transportSolute carrier family 22 member 1 Homo sapiens (human)
dopamine transportSolute carrier family 22 member 1 Homo sapiens (human)
norepinephrine transportSolute carrier family 22 member 1 Homo sapiens (human)
thiamine transportSolute carrier family 22 member 1 Homo sapiens (human)
xenobiotic transportSolute carrier family 22 member 1 Homo sapiens (human)
epinephrine transportSolute carrier family 22 member 1 Homo sapiens (human)
serotonin uptakeSolute carrier family 22 member 1 Homo sapiens (human)
norepinephrine uptakeSolute carrier family 22 member 1 Homo sapiens (human)
thiamine transmembrane transportSolute carrier family 22 member 1 Homo sapiens (human)
metanephric proximal tubule developmentSolute carrier family 22 member 1 Homo sapiens (human)
purine-containing compound transmembrane transportSolute carrier family 22 member 1 Homo sapiens (human)
dopamine uptakeSolute carrier family 22 member 1 Homo sapiens (human)
monoatomic cation transmembrane transportSolute carrier family 22 member 1 Homo sapiens (human)
transport across blood-brain barrierSolute carrier family 22 member 1 Homo sapiens (human)
(R)-carnitine transmembrane transportSolute carrier family 22 member 1 Homo sapiens (human)
acyl carnitine transmembrane transportSolute carrier family 22 member 1 Homo sapiens (human)
spermidine transmembrane transportSolute carrier family 22 member 1 Homo sapiens (human)
cellular detoxificationSolute carrier family 22 member 1 Homo sapiens (human)
xenobiotic transport across blood-brain barrierSolute carrier family 22 member 1 Homo sapiens (human)
xenobiotic metabolic processATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
bile acid and bile salt transportATP-binding cassette sub-family C member 3Homo sapiens (human)
glucuronoside transportATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transportATP-binding cassette sub-family C member 3Homo sapiens (human)
transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
leukotriene transportATP-binding cassette sub-family C member 3Homo sapiens (human)
monoatomic anion transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
transport across blood-brain barrierATP-binding cassette sub-family C member 3Homo sapiens (human)
prostaglandin secretionMultidrug resistance-associated protein 4Homo sapiens (human)
cilium assemblyMultidrug resistance-associated protein 4Homo sapiens (human)
platelet degranulationMultidrug resistance-associated protein 4Homo sapiens (human)
xenobiotic metabolic processMultidrug resistance-associated protein 4Homo sapiens (human)
xenobiotic transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
bile acid and bile salt transportMultidrug resistance-associated protein 4Homo sapiens (human)
prostaglandin transportMultidrug resistance-associated protein 4Homo sapiens (human)
urate transportMultidrug resistance-associated protein 4Homo sapiens (human)
glutathione transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
cAMP transportMultidrug resistance-associated protein 4Homo sapiens (human)
leukotriene transportMultidrug resistance-associated protein 4Homo sapiens (human)
monoatomic anion transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
export across plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
transport across blood-brain barrierMultidrug resistance-associated protein 4Homo sapiens (human)
guanine nucleotide transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
negative regulation of DNA-templated transcriptionNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
regulation of DNA-templated transcriptionNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
xenobiotic metabolic processNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
signal transductionNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
steroid metabolic processNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
positive regulation of gene expressionNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
intracellular receptor signaling pathwayNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
xenobiotic catabolic processNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
xenobiotic transportNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
positive regulation of DNA-templated transcriptionNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
positive regulation of transcription by RNA polymerase IINuclear receptor subfamily 1 group I member 2Homo sapiens (human)
cell differentiationNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
negative regulation of transcription by RNA polymerase IINuclear receptor subfamily 1 group I member 2Homo sapiens (human)
histamine metabolic processSolute carrier family 22 member 3Homo sapiens (human)
organic cation transportSolute carrier family 22 member 3Homo sapiens (human)
quaternary ammonium group transportSolute carrier family 22 member 3Homo sapiens (human)
monoatomic ion transportSolute carrier family 22 member 3Homo sapiens (human)
neurotransmitter transportSolute carrier family 22 member 3Homo sapiens (human)
serotonin transportSolute carrier family 22 member 3Homo sapiens (human)
organic cation transportSolute carrier family 22 member 3Homo sapiens (human)
quaternary ammonium group transportSolute carrier family 22 member 3Homo sapiens (human)
organic anion transportSolute carrier family 22 member 3Homo sapiens (human)
monocarboxylic acid transportSolute carrier family 22 member 3Homo sapiens (human)
monoamine transportSolute carrier family 22 member 3Homo sapiens (human)
spermidine transportSolute carrier family 22 member 3Homo sapiens (human)
dopamine transportSolute carrier family 22 member 3Homo sapiens (human)
norepinephrine transportSolute carrier family 22 member 3Homo sapiens (human)
regulation of appetiteSolute carrier family 22 member 3Homo sapiens (human)
xenobiotic transportSolute carrier family 22 member 3Homo sapiens (human)
epinephrine transportSolute carrier family 22 member 3Homo sapiens (human)
histamine transportSolute carrier family 22 member 3Homo sapiens (human)
serotonin uptakeSolute carrier family 22 member 3Homo sapiens (human)
histamine uptakeSolute carrier family 22 member 3Homo sapiens (human)
norepinephrine uptakeSolute carrier family 22 member 3Homo sapiens (human)
epinephrine uptakeSolute carrier family 22 member 3Homo sapiens (human)
purine-containing compound transmembrane transportSolute carrier family 22 member 3Homo sapiens (human)
dopamine uptakeSolute carrier family 22 member 3Homo sapiens (human)
transport across blood-brain barrierSolute carrier family 22 member 3Homo sapiens (human)
spermidine transmembrane transportSolute carrier family 22 member 3Homo sapiens (human)
cellular detoxificationSolute carrier family 22 member 3Homo sapiens (human)
xenobiotic metabolic processSolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
monoatomic ion transportSolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
organic anion transportSolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
prostaglandin transportSolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
heme catabolic processSolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
sodium-independent organic anion transportSolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
transmembrane transportSolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
transport across blood-brain barrierSolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
bile acid and bile salt transportSolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
fatty acid metabolic processBile salt export pumpHomo sapiens (human)
bile acid biosynthetic processBile salt export pumpHomo sapiens (human)
xenobiotic metabolic processBile salt export pumpHomo sapiens (human)
xenobiotic transmembrane transportBile salt export pumpHomo sapiens (human)
response to oxidative stressBile salt export pumpHomo sapiens (human)
bile acid metabolic processBile salt export pumpHomo sapiens (human)
response to organic cyclic compoundBile salt export pumpHomo sapiens (human)
bile acid and bile salt transportBile salt export pumpHomo sapiens (human)
canalicular bile acid transportBile salt export pumpHomo sapiens (human)
protein ubiquitinationBile salt export pumpHomo sapiens (human)
regulation of fatty acid beta-oxidationBile salt export pumpHomo sapiens (human)
carbohydrate transmembrane transportBile salt export pumpHomo sapiens (human)
bile acid signaling pathwayBile salt export pumpHomo sapiens (human)
cholesterol homeostasisBile salt export pumpHomo sapiens (human)
response to estrogenBile salt export pumpHomo sapiens (human)
response to ethanolBile salt export pumpHomo sapiens (human)
xenobiotic export from cellBile salt export pumpHomo sapiens (human)
lipid homeostasisBile salt export pumpHomo sapiens (human)
phospholipid homeostasisBile salt export pumpHomo sapiens (human)
positive regulation of bile acid secretionBile salt export pumpHomo sapiens (human)
regulation of bile acid metabolic processBile salt export pumpHomo sapiens (human)
transmembrane transportBile salt export pumpHomo sapiens (human)
cell surface receptor signaling pathway via JAK-STATInterferon betaHomo sapiens (human)
response to exogenous dsRNAInterferon betaHomo sapiens (human)
B cell activation involved in immune responseInterferon betaHomo sapiens (human)
cell surface receptor signaling pathwayInterferon betaHomo sapiens (human)
cell surface receptor signaling pathway via JAK-STATInterferon betaHomo sapiens (human)
response to virusInterferon betaHomo sapiens (human)
positive regulation of autophagyInterferon betaHomo sapiens (human)
cytokine-mediated signaling pathwayInterferon betaHomo sapiens (human)
natural killer cell activationInterferon betaHomo sapiens (human)
positive regulation of peptidyl-serine phosphorylation of STAT proteinInterferon betaHomo sapiens (human)
cellular response to interferon-betaInterferon betaHomo sapiens (human)
B cell proliferationInterferon betaHomo sapiens (human)
negative regulation of viral genome replicationInterferon betaHomo sapiens (human)
innate immune responseInterferon betaHomo sapiens (human)
positive regulation of innate immune responseInterferon betaHomo sapiens (human)
regulation of MHC class I biosynthetic processInterferon betaHomo sapiens (human)
negative regulation of T cell differentiationInterferon betaHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIInterferon betaHomo sapiens (human)
defense response to virusInterferon betaHomo sapiens (human)
type I interferon-mediated signaling pathwayInterferon betaHomo sapiens (human)
neuron cellular homeostasisInterferon betaHomo sapiens (human)
cellular response to exogenous dsRNAInterferon betaHomo sapiens (human)
cellular response to virusInterferon betaHomo sapiens (human)
negative regulation of Lewy body formationInterferon betaHomo sapiens (human)
negative regulation of T-helper 2 cell cytokine productionInterferon betaHomo sapiens (human)
positive regulation of apoptotic signaling pathwayInterferon betaHomo sapiens (human)
response to exogenous dsRNAInterferon betaHomo sapiens (human)
B cell differentiationInterferon betaHomo sapiens (human)
natural killer cell activation involved in immune responseInterferon betaHomo sapiens (human)
adaptive immune responseInterferon betaHomo sapiens (human)
T cell activation involved in immune responseInterferon betaHomo sapiens (human)
humoral immune responseInterferon betaHomo sapiens (human)
positive regulation of T cell mediated cytotoxicityHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
adaptive immune responseHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
antigen processing and presentation of endogenous peptide antigen via MHC class I via ER pathway, TAP-independentHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
regulation of T cell anergyHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
defense responseHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
immune responseHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
detection of bacteriumHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
regulation of interleukin-12 productionHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
regulation of interleukin-6 productionHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
protection from natural killer cell mediated cytotoxicityHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
innate immune responseHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
regulation of dendritic cell differentiationHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
antigen processing and presentation of endogenous peptide antigen via MHC class IbHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
cellular response to starvationAlbuminHomo sapiens (human)
negative regulation of mitochondrial depolarizationAlbuminHomo sapiens (human)
cellular response to calcium ion starvationAlbuminHomo sapiens (human)
cellular oxidant detoxificationAlbuminHomo sapiens (human)
transportAlbuminHomo sapiens (human)
viral life cycleGag-Pol polyproteinHIV-1 M:B_HXB2R
establishment of integrated proviral latencyGag-Pol polyproteinHIV-1 M:B_HXB2R
negative regulation of cell population proliferationCellular tumor antigen p53Homo sapiens (human)
regulation of cell cycleCellular tumor antigen p53Homo sapiens (human)
regulation of cell cycle G2/M phase transitionCellular tumor antigen p53Homo sapiens (human)
DNA damage responseCellular tumor antigen p53Homo sapiens (human)
ER overload responseCellular tumor antigen p53Homo sapiens (human)
cellular response to glucose starvationCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
regulation of apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
positive regulation of miRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
negative regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
mitophagyCellular tumor antigen p53Homo sapiens (human)
in utero embryonic developmentCellular tumor antigen p53Homo sapiens (human)
somitogenesisCellular tumor antigen p53Homo sapiens (human)
release of cytochrome c from mitochondriaCellular tumor antigen p53Homo sapiens (human)
hematopoietic progenitor cell differentiationCellular tumor antigen p53Homo sapiens (human)
T cell proliferation involved in immune responseCellular tumor antigen p53Homo sapiens (human)
B cell lineage commitmentCellular tumor antigen p53Homo sapiens (human)
T cell lineage commitmentCellular tumor antigen p53Homo sapiens (human)
response to ischemiaCellular tumor antigen p53Homo sapiens (human)
nucleotide-excision repairCellular tumor antigen p53Homo sapiens (human)
double-strand break repairCellular tumor antigen p53Homo sapiens (human)
regulation of DNA-templated transcriptionCellular tumor antigen p53Homo sapiens (human)
regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
protein import into nucleusCellular tumor antigen p53Homo sapiens (human)
autophagyCellular tumor antigen p53Homo sapiens (human)
DNA damage responseCellular tumor antigen p53Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediator resulting in cell cycle arrestCellular tumor antigen p53Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediator resulting in transcription of p21 class mediatorCellular tumor antigen p53Homo sapiens (human)
transforming growth factor beta receptor signaling pathwayCellular tumor antigen p53Homo sapiens (human)
Ras protein signal transductionCellular tumor antigen p53Homo sapiens (human)
gastrulationCellular tumor antigen p53Homo sapiens (human)
neuroblast proliferationCellular tumor antigen p53Homo sapiens (human)
negative regulation of neuroblast proliferationCellular tumor antigen p53Homo sapiens (human)
protein localizationCellular tumor antigen p53Homo sapiens (human)
negative regulation of DNA replicationCellular tumor antigen p53Homo sapiens (human)
negative regulation of cell population proliferationCellular tumor antigen p53Homo sapiens (human)
determination of adult lifespanCellular tumor antigen p53Homo sapiens (human)
mRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
rRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
response to salt stressCellular tumor antigen p53Homo sapiens (human)
response to inorganic substanceCellular tumor antigen p53Homo sapiens (human)
response to X-rayCellular tumor antigen p53Homo sapiens (human)
response to gamma radiationCellular tumor antigen p53Homo sapiens (human)
positive regulation of gene expressionCellular tumor antigen p53Homo sapiens (human)
cardiac muscle cell apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of cardiac muscle cell apoptotic processCellular tumor antigen p53Homo sapiens (human)
glial cell proliferationCellular tumor antigen p53Homo sapiens (human)
viral processCellular tumor antigen p53Homo sapiens (human)
glucose catabolic process to lactate via pyruvateCellular tumor antigen p53Homo sapiens (human)
cerebellum developmentCellular tumor antigen p53Homo sapiens (human)
negative regulation of cell growthCellular tumor antigen p53Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
negative regulation of transforming growth factor beta receptor signaling pathwayCellular tumor antigen p53Homo sapiens (human)
mitotic G1 DNA damage checkpoint signalingCellular tumor antigen p53Homo sapiens (human)
negative regulation of telomere maintenance via telomeraseCellular tumor antigen p53Homo sapiens (human)
T cell differentiation in thymusCellular tumor antigen p53Homo sapiens (human)
tumor necrosis factor-mediated signaling pathwayCellular tumor antigen p53Homo sapiens (human)
regulation of tissue remodelingCellular tumor antigen p53Homo sapiens (human)
cellular response to UVCellular tumor antigen p53Homo sapiens (human)
multicellular organism growthCellular tumor antigen p53Homo sapiens (human)
positive regulation of mitochondrial membrane permeabilityCellular tumor antigen p53Homo sapiens (human)
cellular response to glucose starvationCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
positive regulation of apoptotic processCellular tumor antigen p53Homo sapiens (human)
negative regulation of apoptotic processCellular tumor antigen p53Homo sapiens (human)
entrainment of circadian clock by photoperiodCellular tumor antigen p53Homo sapiens (human)
mitochondrial DNA repairCellular tumor antigen p53Homo sapiens (human)
regulation of DNA damage response, signal transduction by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
positive regulation of neuron apoptotic processCellular tumor antigen p53Homo sapiens (human)
transcription initiation-coupled chromatin remodelingCellular tumor antigen p53Homo sapiens (human)
negative regulation of proteolysisCellular tumor antigen p53Homo sapiens (human)
negative regulation of DNA-templated transcriptionCellular tumor antigen p53Homo sapiens (human)
positive regulation of DNA-templated transcriptionCellular tumor antigen p53Homo sapiens (human)
positive regulation of RNA polymerase II transcription preinitiation complex assemblyCellular tumor antigen p53Homo sapiens (human)
positive regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
response to antibioticCellular tumor antigen p53Homo sapiens (human)
fibroblast proliferationCellular tumor antigen p53Homo sapiens (human)
negative regulation of fibroblast proliferationCellular tumor antigen p53Homo sapiens (human)
circadian behaviorCellular tumor antigen p53Homo sapiens (human)
bone marrow developmentCellular tumor antigen p53Homo sapiens (human)
embryonic organ developmentCellular tumor antigen p53Homo sapiens (human)
positive regulation of peptidyl-tyrosine phosphorylationCellular tumor antigen p53Homo sapiens (human)
protein stabilizationCellular tumor antigen p53Homo sapiens (human)
negative regulation of helicase activityCellular tumor antigen p53Homo sapiens (human)
protein tetramerizationCellular tumor antigen p53Homo sapiens (human)
chromosome organizationCellular tumor antigen p53Homo sapiens (human)
neuron apoptotic processCellular tumor antigen p53Homo sapiens (human)
regulation of cell cycleCellular tumor antigen p53Homo sapiens (human)
hematopoietic stem cell differentiationCellular tumor antigen p53Homo sapiens (human)
negative regulation of glial cell proliferationCellular tumor antigen p53Homo sapiens (human)
type II interferon-mediated signaling pathwayCellular tumor antigen p53Homo sapiens (human)
cardiac septum morphogenesisCellular tumor antigen p53Homo sapiens (human)
positive regulation of programmed necrotic cell deathCellular tumor antigen p53Homo sapiens (human)
protein-containing complex assemblyCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to endoplasmic reticulum stressCellular tumor antigen p53Homo sapiens (human)
thymocyte apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of thymocyte apoptotic processCellular tumor antigen p53Homo sapiens (human)
necroptotic processCellular tumor antigen p53Homo sapiens (human)
cellular response to hypoxiaCellular tumor antigen p53Homo sapiens (human)
cellular response to xenobiotic stimulusCellular tumor antigen p53Homo sapiens (human)
cellular response to ionizing radiationCellular tumor antigen p53Homo sapiens (human)
cellular response to gamma radiationCellular tumor antigen p53Homo sapiens (human)
cellular response to UV-CCellular tumor antigen p53Homo sapiens (human)
stem cell proliferationCellular tumor antigen p53Homo sapiens (human)
signal transduction by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
reactive oxygen species metabolic processCellular tumor antigen p53Homo sapiens (human)
cellular response to actinomycin DCellular tumor antigen p53Homo sapiens (human)
positive regulation of release of cytochrome c from mitochondriaCellular tumor antigen p53Homo sapiens (human)
cellular senescenceCellular tumor antigen p53Homo sapiens (human)
replicative senescenceCellular tumor antigen p53Homo sapiens (human)
oxidative stress-induced premature senescenceCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathwayCellular tumor antigen p53Homo sapiens (human)
oligodendrocyte apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of execution phase of apoptosisCellular tumor antigen p53Homo sapiens (human)
negative regulation of mitophagyCellular tumor antigen p53Homo sapiens (human)
regulation of mitochondrial membrane permeability involved in apoptotic processCellular tumor antigen p53Homo sapiens (human)
regulation of intrinsic apoptotic signaling pathway by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
positive regulation of miRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
negative regulation of G1 to G0 transitionCellular tumor antigen p53Homo sapiens (human)
negative regulation of miRNA processingCellular tumor antigen p53Homo sapiens (human)
negative regulation of glucose catabolic process to lactate via pyruvateCellular tumor antigen p53Homo sapiens (human)
negative regulation of pentose-phosphate shuntCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to hypoxiaCellular tumor antigen p53Homo sapiens (human)
regulation of fibroblast apoptotic processCellular tumor antigen p53Homo sapiens (human)
negative regulation of reactive oxygen species metabolic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of reactive oxygen species metabolic processCellular tumor antigen p53Homo sapiens (human)
negative regulation of stem cell proliferationCellular tumor antigen p53Homo sapiens (human)
positive regulation of cellular senescenceCellular tumor antigen p53Homo sapiens (human)
positive regulation of intrinsic apoptotic signaling pathwayCellular tumor antigen p53Homo sapiens (human)
steroid catabolic processCytochrome P450 1A2Homo sapiens (human)
porphyrin-containing compound metabolic processCytochrome P450 1A2Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 1A2Homo sapiens (human)
cholesterol metabolic processCytochrome P450 1A2Homo sapiens (human)
estrogen metabolic processCytochrome P450 1A2Homo sapiens (human)
toxin biosynthetic processCytochrome P450 1A2Homo sapiens (human)
post-embryonic developmentCytochrome P450 1A2Homo sapiens (human)
alkaloid metabolic processCytochrome P450 1A2Homo sapiens (human)
regulation of gene expressionCytochrome P450 1A2Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 1A2Homo sapiens (human)
dibenzo-p-dioxin metabolic processCytochrome P450 1A2Homo sapiens (human)
epoxygenase P450 pathwayCytochrome P450 1A2Homo sapiens (human)
lung developmentCytochrome P450 1A2Homo sapiens (human)
methylationCytochrome P450 1A2Homo sapiens (human)
monocarboxylic acid metabolic processCytochrome P450 1A2Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 1A2Homo sapiens (human)
retinol metabolic processCytochrome P450 1A2Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 1A2Homo sapiens (human)
cellular respirationCytochrome P450 1A2Homo sapiens (human)
aflatoxin metabolic processCytochrome P450 1A2Homo sapiens (human)
hydrogen peroxide biosynthetic processCytochrome P450 1A2Homo sapiens (human)
oxidative demethylationCytochrome P450 1A2Homo sapiens (human)
cellular response to cadmium ionCytochrome P450 1A2Homo sapiens (human)
omega-hydroxylase P450 pathwayCytochrome P450 1A2Homo sapiens (human)
G2/M transition of mitotic cell cycleATP-dependent translocase ABCB1Homo sapiens (human)
xenobiotic metabolic processATP-dependent translocase ABCB1Homo sapiens (human)
response to xenobiotic stimulusATP-dependent translocase ABCB1Homo sapiens (human)
phospholipid translocationATP-dependent translocase ABCB1Homo sapiens (human)
terpenoid transportATP-dependent translocase ABCB1Homo sapiens (human)
regulation of response to osmotic stressATP-dependent translocase ABCB1Homo sapiens (human)
transmembrane transportATP-dependent translocase ABCB1Homo sapiens (human)
transepithelial transportATP-dependent translocase ABCB1Homo sapiens (human)
stem cell proliferationATP-dependent translocase ABCB1Homo sapiens (human)
ceramide translocationATP-dependent translocase ABCB1Homo sapiens (human)
export across plasma membraneATP-dependent translocase ABCB1Homo sapiens (human)
transport across blood-brain barrierATP-dependent translocase ABCB1Homo sapiens (human)
positive regulation of anion channel activityATP-dependent translocase ABCB1Homo sapiens (human)
carboxylic acid transmembrane transportATP-dependent translocase ABCB1Homo sapiens (human)
xenobiotic detoxification by transmembrane export across the plasma membraneATP-dependent translocase ABCB1Homo sapiens (human)
xenobiotic transport across blood-brain barrierATP-dependent translocase ABCB1Homo sapiens (human)
regulation of chloride transportATP-dependent translocase ABCB1Homo sapiens (human)
lipid hydroxylationCytochrome P450 3A4Homo sapiens (human)
lipid metabolic processCytochrome P450 3A4Homo sapiens (human)
steroid catabolic processCytochrome P450 3A4Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 3A4Homo sapiens (human)
steroid metabolic processCytochrome P450 3A4Homo sapiens (human)
cholesterol metabolic processCytochrome P450 3A4Homo sapiens (human)
androgen metabolic processCytochrome P450 3A4Homo sapiens (human)
estrogen metabolic processCytochrome P450 3A4Homo sapiens (human)
alkaloid catabolic processCytochrome P450 3A4Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 3A4Homo sapiens (human)
calcitriol biosynthetic process from calciolCytochrome P450 3A4Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 3A4Homo sapiens (human)
vitamin D metabolic processCytochrome P450 3A4Homo sapiens (human)
vitamin D catabolic processCytochrome P450 3A4Homo sapiens (human)
retinol metabolic processCytochrome P450 3A4Homo sapiens (human)
retinoic acid metabolic processCytochrome P450 3A4Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 3A4Homo sapiens (human)
aflatoxin metabolic processCytochrome P450 3A4Homo sapiens (human)
oxidative demethylationCytochrome P450 3A4Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 2D6Homo sapiens (human)
steroid metabolic processCytochrome P450 2D6Homo sapiens (human)
cholesterol metabolic processCytochrome P450 2D6Homo sapiens (human)
estrogen metabolic processCytochrome P450 2D6Homo sapiens (human)
coumarin metabolic processCytochrome P450 2D6Homo sapiens (human)
alkaloid metabolic processCytochrome P450 2D6Homo sapiens (human)
alkaloid catabolic processCytochrome P450 2D6Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 2D6Homo sapiens (human)
isoquinoline alkaloid metabolic processCytochrome P450 2D6Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 2D6Homo sapiens (human)
retinol metabolic processCytochrome P450 2D6Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 2D6Homo sapiens (human)
negative regulation of bindingCytochrome P450 2D6Homo sapiens (human)
oxidative demethylationCytochrome P450 2D6Homo sapiens (human)
negative regulation of cellular organofluorine metabolic processCytochrome P450 2D6Homo sapiens (human)
arachidonic acid metabolic processCytochrome P450 2D6Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 2C9 Homo sapiens (human)
steroid metabolic processCytochrome P450 2C9 Homo sapiens (human)
cholesterol metabolic processCytochrome P450 2C9 Homo sapiens (human)
estrogen metabolic processCytochrome P450 2C9 Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 2C9 Homo sapiens (human)
epoxygenase P450 pathwayCytochrome P450 2C9 Homo sapiens (human)
urea metabolic processCytochrome P450 2C9 Homo sapiens (human)
monocarboxylic acid metabolic processCytochrome P450 2C9 Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 2C9 Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 2C9 Homo sapiens (human)
amide metabolic processCytochrome P450 2C9 Homo sapiens (human)
icosanoid biosynthetic processCytochrome P450 2C9 Homo sapiens (human)
oxidative demethylationCytochrome P450 2C9 Homo sapiens (human)
omega-hydroxylase P450 pathwayCytochrome P450 2C9 Homo sapiens (human)
lipid hydroxylationCytochrome P450 3A5Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 3A5Homo sapiens (human)
steroid metabolic processCytochrome P450 3A5Homo sapiens (human)
estrogen metabolic processCytochrome P450 3A5Homo sapiens (human)
alkaloid catabolic processCytochrome P450 3A5Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 3A5Homo sapiens (human)
retinol metabolic processCytochrome P450 3A5Homo sapiens (human)
retinoic acid metabolic processCytochrome P450 3A5Homo sapiens (human)
aflatoxin metabolic processCytochrome P450 3A5Homo sapiens (human)
oxidative demethylationCytochrome P450 3A5Homo sapiens (human)
muscle contractionSubstance-K receptorHomo sapiens (human)
tachykinin receptor signaling pathwaySubstance-K receptorHomo sapiens (human)
positive regulation of acetylcholine secretion, neurotransmissionSubstance-K receptorHomo sapiens (human)
intestine smooth muscle contractionSubstance-K receptorHomo sapiens (human)
negative regulation of luteinizing hormone secretionSubstance-K receptorHomo sapiens (human)
operant conditioningSubstance-K receptorHomo sapiens (human)
positive regulation of vascular permeabilitySubstance-K receptorHomo sapiens (human)
positive regulation of monoatomic ion transportSubstance-K receptorHomo sapiens (human)
positive regulation of smooth muscle contractionSubstance-K receptorHomo sapiens (human)
response to electrical stimulusSubstance-K receptorHomo sapiens (human)
prolactin secretionSubstance-K receptorHomo sapiens (human)
positive regulation of uterine smooth muscle contractionSubstance-K receptorHomo sapiens (human)
positive regulation of flagellated sperm motilitySubstance-K receptorHomo sapiens (human)
liver developmentUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
bilirubin conjugationUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
xenobiotic metabolic processUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
acute-phase responseUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
response to nutrientUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
steroid metabolic processUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
estrogen metabolic processUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
animal organ regenerationUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
response to lipopolysaccharideUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
retinoic acid metabolic processUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
response to starvationUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
negative regulation of steroid metabolic processUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
flavone metabolic processUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
cellular glucuronidationUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
flavonoid glucuronidationUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
xenobiotic glucuronidationUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
biphenyl catabolic processUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
cellular response to ethanolUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
cellular response to glucocorticoid stimulusUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
cellular response to estradiol stimulusUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
prostaglandin biosynthetic processThromboxane-A synthase Homo sapiens (human)
icosanoid metabolic processThromboxane-A synthase Homo sapiens (human)
cyclooxygenase pathwayThromboxane-A synthase Homo sapiens (human)
intracellular chloride ion homeostasisThromboxane-A synthase Homo sapiens (human)
response to ethanolThromboxane-A synthase Homo sapiens (human)
positive regulation of vasoconstrictionThromboxane-A synthase Homo sapiens (human)
response to fatty acidThromboxane-A synthase Homo sapiens (human)
long-chain fatty acid metabolic processCytochrome P450 2C19Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 2C19Homo sapiens (human)
steroid metabolic processCytochrome P450 2C19Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 2C19Homo sapiens (human)
epoxygenase P450 pathwayCytochrome P450 2C19Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 2C19Homo sapiens (human)
omega-hydroxylase P450 pathwayCytochrome P450 2C19Homo sapiens (human)
G protein-coupled receptor signaling pathway, coupled to cyclic nucleotide second messengerMu-type opioid receptorHomo sapiens (human)
adenylate cyclase-inhibiting G protein-coupled acetylcholine receptor signaling pathwayMu-type opioid receptorHomo sapiens (human)
phospholipase C-activating G protein-coupled receptor signaling pathwayMu-type opioid receptorHomo sapiens (human)
sensory perceptionMu-type opioid receptorHomo sapiens (human)
negative regulation of cell population proliferationMu-type opioid receptorHomo sapiens (human)
sensory perception of painMu-type opioid receptorHomo sapiens (human)
G protein-coupled opioid receptor signaling pathwayMu-type opioid receptorHomo sapiens (human)
behavioral response to ethanolMu-type opioid receptorHomo sapiens (human)
positive regulation of neurogenesisMu-type opioid receptorHomo sapiens (human)
negative regulation of Wnt protein secretionMu-type opioid receptorHomo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeMu-type opioid receptorHomo sapiens (human)
calcium ion transmembrane transportMu-type opioid receptorHomo sapiens (human)
cellular response to morphineMu-type opioid receptorHomo sapiens (human)
regulation of cellular response to stressMu-type opioid receptorHomo sapiens (human)
regulation of NMDA receptor activityMu-type opioid receptorHomo sapiens (human)
neuropeptide signaling pathwayMu-type opioid receptorHomo sapiens (human)
maternal aggressive behaviorVasopressin V1a receptorHomo sapiens (human)
positive regulation of systemic arterial blood pressureVasopressin V1a receptorHomo sapiens (human)
generation of precursor metabolites and energyVasopressin V1a receptorHomo sapiens (human)
activation of phospholipase C activityVasopressin V1a receptorHomo sapiens (human)
positive regulation of cytosolic calcium ion concentrationVasopressin V1a receptorHomo sapiens (human)
negative regulation of female receptivityVasopressin V1a receptorHomo sapiens (human)
grooming behaviorVasopressin V1a receptorHomo sapiens (human)
blood circulationVasopressin V1a receptorHomo sapiens (human)
positive regulation of cell population proliferationVasopressin V1a receptorHomo sapiens (human)
positive regulation of heart rateVasopressin V1a receptorHomo sapiens (human)
positive regulation of glutamate secretionVasopressin V1a receptorHomo sapiens (human)
myotube differentiationVasopressin V1a receptorHomo sapiens (human)
calcium-mediated signalingVasopressin V1a receptorHomo sapiens (human)
telencephalon developmentVasopressin V1a receptorHomo sapiens (human)
positive regulation of cell growthVasopressin V1a receptorHomo sapiens (human)
positive regulation of prostaglandin biosynthetic processVasopressin V1a receptorHomo sapiens (human)
positive regulation of cellular pH reductionVasopressin V1a receptorHomo sapiens (human)
social behaviorVasopressin V1a receptorHomo sapiens (human)
cellular response to water deprivationVasopressin V1a receptorHomo sapiens (human)
maternal behaviorVasopressin V1a receptorHomo sapiens (human)
sperm ejaculationVasopressin V1a receptorHomo sapiens (human)
response to corticosteroneVasopressin V1a receptorHomo sapiens (human)
negative regulation of transmission of nerve impulseVasopressin V1a receptorHomo sapiens (human)
transport across blood-brain barrierVasopressin V1a receptorHomo sapiens (human)
G protein-coupled receptor signaling pathwayVasopressin V1a receptorHomo sapiens (human)
positive regulation of vasoconstrictionVasopressin V1a receptorHomo sapiens (human)
cellular response to hormone stimulusVasopressin V1a receptorHomo sapiens (human)
regulation of systemic arterial blood pressure by vasopressinVasopressin V1a receptorHomo sapiens (human)
immune responseKappa-type opioid receptorHomo sapiens (human)
adenylate cyclase-inhibiting G protein-coupled receptor signaling pathwayKappa-type opioid receptorHomo sapiens (human)
phospholipase C-activating G protein-coupled receptor signaling pathwayKappa-type opioid receptorHomo sapiens (human)
chemical synaptic transmissionKappa-type opioid receptorHomo sapiens (human)
sensory perceptionKappa-type opioid receptorHomo sapiens (human)
locomotory behaviorKappa-type opioid receptorHomo sapiens (human)
sensory perception of painKappa-type opioid receptorHomo sapiens (human)
adenylate cyclase-inhibiting opioid receptor signaling pathwayKappa-type opioid receptorHomo sapiens (human)
response to insulinKappa-type opioid receptorHomo sapiens (human)
positive regulation of dopamine secretionKappa-type opioid receptorHomo sapiens (human)
negative regulation of luteinizing hormone secretionKappa-type opioid receptorHomo sapiens (human)
response to nicotineKappa-type opioid receptorHomo sapiens (human)
G protein-coupled opioid receptor signaling pathwayKappa-type opioid receptorHomo sapiens (human)
maternal behaviorKappa-type opioid receptorHomo sapiens (human)
eating behaviorKappa-type opioid receptorHomo sapiens (human)
response to estrogenKappa-type opioid receptorHomo sapiens (human)
estrous cycleKappa-type opioid receptorHomo sapiens (human)
response to ethanolKappa-type opioid receptorHomo sapiens (human)
regulation of saliva secretionKappa-type opioid receptorHomo sapiens (human)
behavioral response to cocaineKappa-type opioid receptorHomo sapiens (human)
sensory perception of temperature stimulusKappa-type opioid receptorHomo sapiens (human)
defense response to virusKappa-type opioid receptorHomo sapiens (human)
cellular response to lipopolysaccharideKappa-type opioid receptorHomo sapiens (human)
cellular response to glucose stimulusKappa-type opioid receptorHomo sapiens (human)
positive regulation of p38MAPK cascadeKappa-type opioid receptorHomo sapiens (human)
positive regulation of potassium ion transmembrane transportKappa-type opioid receptorHomo sapiens (human)
response to acrylamideKappa-type opioid receptorHomo sapiens (human)
positive regulation of eating behaviorKappa-type opioid receptorHomo sapiens (human)
conditioned place preferenceKappa-type opioid receptorHomo sapiens (human)
neuropeptide signaling pathwayKappa-type opioid receptorHomo sapiens (human)
sodium ion transportSodium/bile acid cotransporterHomo sapiens (human)
response to organic cyclic compoundSodium/bile acid cotransporterHomo sapiens (human)
bile acid and bile salt transportSodium/bile acid cotransporterHomo sapiens (human)
response to nutrient levelsSodium/bile acid cotransporterHomo sapiens (human)
bile acid signaling pathwaySodium/bile acid cotransporterHomo sapiens (human)
response to estrogenSodium/bile acid cotransporterHomo sapiens (human)
response to ethanolSodium/bile acid cotransporterHomo sapiens (human)
symbiont entry into host cellSodium/bile acid cotransporterHomo sapiens (human)
transmembrane transportSodium/bile acid cotransporterHomo sapiens (human)
cellular response to xenobiotic stimulusSodium/bile acid cotransporterHomo sapiens (human)
regulation of bile acid secretionSodium/bile acid cotransporterHomo sapiens (human)
organic cation transportMultidrug and toxin extrusion protein 2Homo sapiens (human)
transmembrane transportMultidrug and toxin extrusion protein 2Homo sapiens (human)
proton transmembrane transportMultidrug and toxin extrusion protein 2Homo sapiens (human)
xenobiotic detoxification by transmembrane export across the plasma membraneMultidrug and toxin extrusion protein 2Homo sapiens (human)
inositol phosphate metabolic processInositol hexakisphosphate kinase 1Homo sapiens (human)
phosphatidylinositol phosphate biosynthetic processInositol hexakisphosphate kinase 1Homo sapiens (human)
negative regulation of cold-induced thermogenesisInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol phosphate biosynthetic processInositol hexakisphosphate kinase 1Homo sapiens (human)
xenobiotic metabolic processCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
negative regulation of gene expressionCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bile acid and bile salt transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bilirubin transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
heme catabolic processCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic export from cellCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transepithelial transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
leukotriene transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
monoatomic anion transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transport across blood-brain barrierCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transport across blood-brain barrierCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transmembrane transportMultidrug and toxin extrusion protein 1Homo sapiens (human)
organic cation transportMultidrug and toxin extrusion protein 1Homo sapiens (human)
putrescine transportMultidrug and toxin extrusion protein 1Homo sapiens (human)
xenobiotic transportMultidrug and toxin extrusion protein 1Homo sapiens (human)
transmembrane transportMultidrug and toxin extrusion protein 1Homo sapiens (human)
thiamine transmembrane transportMultidrug and toxin extrusion protein 1Homo sapiens (human)
amino acid import across plasma membraneMultidrug and toxin extrusion protein 1Homo sapiens (human)
L-arginine import across plasma membraneMultidrug and toxin extrusion protein 1Homo sapiens (human)
L-alpha-amino acid transmembrane transportMultidrug and toxin extrusion protein 1Homo sapiens (human)
proton transmembrane transportMultidrug and toxin extrusion protein 1Homo sapiens (human)
L-arginine transmembrane transportMultidrug and toxin extrusion protein 1Homo sapiens (human)
xenobiotic detoxification by transmembrane export across the plasma membraneMultidrug and toxin extrusion protein 1Homo sapiens (human)
cell population proliferationATPase family AAA domain-containing protein 5Homo sapiens (human)
positive regulation of B cell proliferationATPase family AAA domain-containing protein 5Homo sapiens (human)
nuclear DNA replicationATPase family AAA domain-containing protein 5Homo sapiens (human)
signal transduction in response to DNA damageATPase family AAA domain-containing protein 5Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorATPase family AAA domain-containing protein 5Homo sapiens (human)
isotype switchingATPase family AAA domain-containing protein 5Homo sapiens (human)
positive regulation of DNA replicationATPase family AAA domain-containing protein 5Homo sapiens (human)
positive regulation of isotype switching to IgG isotypesATPase family AAA domain-containing protein 5Homo sapiens (human)
DNA clamp unloadingATPase family AAA domain-containing protein 5Homo sapiens (human)
regulation of mitotic cell cycle phase transitionATPase family AAA domain-containing protein 5Homo sapiens (human)
negative regulation of intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorATPase family AAA domain-containing protein 5Homo sapiens (human)
positive regulation of cell cycle G2/M phase transitionATPase family AAA domain-containing protein 5Homo sapiens (human)
negative regulation of receptor internalizationAtaxin-2Homo sapiens (human)
regulation of translationAtaxin-2Homo sapiens (human)
RNA metabolic processAtaxin-2Homo sapiens (human)
P-body assemblyAtaxin-2Homo sapiens (human)
stress granule assemblyAtaxin-2Homo sapiens (human)
RNA transportAtaxin-2Homo sapiens (human)
xenobiotic metabolic processSolute carrier organic anion transporter family member 1B3Homo sapiens (human)
monoatomic ion transportSolute carrier organic anion transporter family member 1B3Homo sapiens (human)
organic anion transportSolute carrier organic anion transporter family member 1B3Homo sapiens (human)
bile acid and bile salt transportSolute carrier organic anion transporter family member 1B3Homo sapiens (human)
heme catabolic processSolute carrier organic anion transporter family member 1B3Homo sapiens (human)
sodium-independent organic anion transportSolute carrier organic anion transporter family member 1B3Homo sapiens (human)
transmembrane transportSolute carrier organic anion transporter family member 1B3Homo sapiens (human)
lipid transportBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
organic anion transportBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
urate transportBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
biotin transportBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
sphingolipid biosynthetic processBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
riboflavin transportBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
urate metabolic processBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
transmembrane transportBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
transepithelial transportBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
renal urate salt excretionBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
export across plasma membraneBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
transport across blood-brain barrierBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
cellular detoxificationBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
xenobiotic transport across blood-brain barrierBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
xenobiotic metabolic processSolute carrier organic anion transporter family member 1B1Homo sapiens (human)
monoatomic ion transportSolute carrier organic anion transporter family member 1B1Homo sapiens (human)
organic anion transportSolute carrier organic anion transporter family member 1B1Homo sapiens (human)
bile acid and bile salt transportSolute carrier organic anion transporter family member 1B1Homo sapiens (human)
prostaglandin transportSolute carrier organic anion transporter family member 1B1Homo sapiens (human)
heme catabolic processSolute carrier organic anion transporter family member 1B1Homo sapiens (human)
sodium-independent organic anion transportSolute carrier organic anion transporter family member 1B1Homo sapiens (human)
transmembrane transportSolute carrier organic anion transporter family member 1B1Homo sapiens (human)
thyroid hormone transportSolute carrier organic anion transporter family member 1B1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (188)

Processvia Protein(s)Taxonomy
amine transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
acetylcholine transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
neurotransmitter transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
monoamine transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
organic anion transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
organic cation transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
prostaglandin transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
L-amino acid transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
pyrimidine nucleoside transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
choline transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
thiamine transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
putrescine transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
efflux transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
spermidine transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
quaternary ammonium group transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
toxin transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
xenobiotic transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
L-arginine transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
acetylcholine transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
neurotransmitter transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
dopamine:sodium symporter activitySolute carrier family 22 member 1 Homo sapiens (human)
norepinephrine:sodium symporter activitySolute carrier family 22 member 1 Homo sapiens (human)
protein bindingSolute carrier family 22 member 1 Homo sapiens (human)
monoamine transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
secondary active organic cation transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
organic anion transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
organic cation transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
prostaglandin transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
pyrimidine nucleoside transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
thiamine transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
putrescine transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
spermidine transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
quaternary ammonium group transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
toxin transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
identical protein bindingSolute carrier family 22 member 1 Homo sapiens (human)
xenobiotic transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
(R)-carnitine transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
ATP bindingATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type xenobiotic transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
glucuronoside transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type bile acid transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATP hydrolysis activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATPase-coupled transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
icosanoid transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
guanine nucleotide transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
protein bindingMultidrug resistance-associated protein 4Homo sapiens (human)
ATP bindingMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type xenobiotic transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
prostaglandin transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
urate transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
purine nucleotide transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type bile acid transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
efflux transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
15-hydroxyprostaglandin dehydrogenase (NAD+) activityMultidrug resistance-associated protein 4Homo sapiens (human)
ATP hydrolysis activityMultidrug resistance-associated protein 4Homo sapiens (human)
glutathione transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ATPase-coupled transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
xenobiotic transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
RNA polymerase II transcription regulatory region sequence-specific DNA bindingNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
DNA-binding transcription factor activity, RNA polymerase II-specificNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
DNA-binding transcription activator activity, RNA polymerase II-specificNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
nuclear receptor activityNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
protein bindingNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
zinc ion bindingNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
nuclear receptor bindingNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
sequence-specific double-stranded DNA bindingNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
neurotransmitter transmembrane transporter activitySolute carrier family 22 member 3Homo sapiens (human)
protein bindingSolute carrier family 22 member 3Homo sapiens (human)
monoamine transmembrane transporter activitySolute carrier family 22 member 3Homo sapiens (human)
organic anion transmembrane transporter activitySolute carrier family 22 member 3Homo sapiens (human)
organic cation transmembrane transporter activitySolute carrier family 22 member 3Homo sapiens (human)
spermidine transmembrane transporter activitySolute carrier family 22 member 3Homo sapiens (human)
quaternary ammonium group transmembrane transporter activitySolute carrier family 22 member 3Homo sapiens (human)
toxin transmembrane transporter activitySolute carrier family 22 member 3Homo sapiens (human)
organic anion transmembrane transporter activitySolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
prostaglandin transmembrane transporter activitySolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
sodium-independent organic anion transmembrane transporter activitySolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
transmembrane transporter activitySolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
bile acid transmembrane transporter activitySolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
protein bindingBile salt export pumpHomo sapiens (human)
ATP bindingBile salt export pumpHomo sapiens (human)
ABC-type xenobiotic transporter activityBile salt export pumpHomo sapiens (human)
bile acid transmembrane transporter activityBile salt export pumpHomo sapiens (human)
canalicular bile acid transmembrane transporter activityBile salt export pumpHomo sapiens (human)
carbohydrate transmembrane transporter activityBile salt export pumpHomo sapiens (human)
ABC-type bile acid transporter activityBile salt export pumpHomo sapiens (human)
ATP hydrolysis activityBile salt export pumpHomo sapiens (human)
cytokine activityInterferon betaHomo sapiens (human)
cytokine receptor bindingInterferon betaHomo sapiens (human)
type I interferon receptor bindingInterferon betaHomo sapiens (human)
protein bindingInterferon betaHomo sapiens (human)
chloramphenicol O-acetyltransferase activityInterferon betaHomo sapiens (human)
TAP bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
signaling receptor bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
protein bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
peptide antigen bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
TAP bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
protein-folding chaperone bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
oxygen bindingAlbuminHomo sapiens (human)
DNA bindingAlbuminHomo sapiens (human)
fatty acid bindingAlbuminHomo sapiens (human)
copper ion bindingAlbuminHomo sapiens (human)
protein bindingAlbuminHomo sapiens (human)
toxic substance bindingAlbuminHomo sapiens (human)
antioxidant activityAlbuminHomo sapiens (human)
pyridoxal phosphate bindingAlbuminHomo sapiens (human)
identical protein bindingAlbuminHomo sapiens (human)
protein-folding chaperone bindingAlbuminHomo sapiens (human)
exogenous protein bindingAlbuminHomo sapiens (human)
enterobactin bindingAlbuminHomo sapiens (human)
peptidase activityGag-Pol polyproteinHIV-1 M:B_HXB2R
integrase activityGag-Pol polyproteinHIV-1 M:B_HXB2R
transcription cis-regulatory region bindingCellular tumor antigen p53Homo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription factor activity, RNA polymerase II-specificCellular tumor antigen p53Homo sapiens (human)
cis-regulatory region sequence-specific DNA bindingCellular tumor antigen p53Homo sapiens (human)
core promoter sequence-specific DNA bindingCellular tumor antigen p53Homo sapiens (human)
TFIID-class transcription factor complex bindingCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription repressor activity, RNA polymerase II-specificCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription activator activity, RNA polymerase II-specificCellular tumor antigen p53Homo sapiens (human)
protease bindingCellular tumor antigen p53Homo sapiens (human)
p53 bindingCellular tumor antigen p53Homo sapiens (human)
DNA bindingCellular tumor antigen p53Homo sapiens (human)
chromatin bindingCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription factor activityCellular tumor antigen p53Homo sapiens (human)
mRNA 3'-UTR bindingCellular tumor antigen p53Homo sapiens (human)
copper ion bindingCellular tumor antigen p53Homo sapiens (human)
protein bindingCellular tumor antigen p53Homo sapiens (human)
zinc ion bindingCellular tumor antigen p53Homo sapiens (human)
enzyme bindingCellular tumor antigen p53Homo sapiens (human)
receptor tyrosine kinase bindingCellular tumor antigen p53Homo sapiens (human)
ubiquitin protein ligase bindingCellular tumor antigen p53Homo sapiens (human)
histone deacetylase regulator activityCellular tumor antigen p53Homo sapiens (human)
ATP-dependent DNA/DNA annealing activityCellular tumor antigen p53Homo sapiens (human)
identical protein bindingCellular tumor antigen p53Homo sapiens (human)
histone deacetylase bindingCellular tumor antigen p53Homo sapiens (human)
protein heterodimerization activityCellular tumor antigen p53Homo sapiens (human)
protein-folding chaperone bindingCellular tumor antigen p53Homo sapiens (human)
protein phosphatase 2A bindingCellular tumor antigen p53Homo sapiens (human)
RNA polymerase II-specific DNA-binding transcription factor bindingCellular tumor antigen p53Homo sapiens (human)
14-3-3 protein bindingCellular tumor antigen p53Homo sapiens (human)
MDM2/MDM4 family protein bindingCellular tumor antigen p53Homo sapiens (human)
disordered domain specific bindingCellular tumor antigen p53Homo sapiens (human)
general transcription initiation factor bindingCellular tumor antigen p53Homo sapiens (human)
molecular function activator activityCellular tumor antigen p53Homo sapiens (human)
promoter-specific chromatin bindingCellular tumor antigen p53Homo sapiens (human)
monooxygenase activityCytochrome P450 1A2Homo sapiens (human)
iron ion bindingCytochrome P450 1A2Homo sapiens (human)
protein bindingCytochrome P450 1A2Homo sapiens (human)
electron transfer activityCytochrome P450 1A2Homo sapiens (human)
oxidoreductase activityCytochrome P450 1A2Homo sapiens (human)
oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen, reduced flavin or flavoprotein as one donor, and incorporation of one atom of oxygenCytochrome P450 1A2Homo sapiens (human)
enzyme bindingCytochrome P450 1A2Homo sapiens (human)
heme bindingCytochrome P450 1A2Homo sapiens (human)
demethylase activityCytochrome P450 1A2Homo sapiens (human)
caffeine oxidase activityCytochrome P450 1A2Homo sapiens (human)
aromatase activityCytochrome P450 1A2Homo sapiens (human)
estrogen 16-alpha-hydroxylase activityCytochrome P450 1A2Homo sapiens (human)
estrogen 2-hydroxylase activityCytochrome P450 1A2Homo sapiens (human)
hydroperoxy icosatetraenoate dehydratase activityCytochrome P450 1A2Homo sapiens (human)
protein bindingATP-dependent translocase ABCB1Homo sapiens (human)
ATP bindingATP-dependent translocase ABCB1Homo sapiens (human)
ABC-type xenobiotic transporter activityATP-dependent translocase ABCB1Homo sapiens (human)
efflux transmembrane transporter activityATP-dependent translocase ABCB1Homo sapiens (human)
ATP hydrolysis activityATP-dependent translocase ABCB1Homo sapiens (human)
transmembrane transporter activityATP-dependent translocase ABCB1Homo sapiens (human)
ubiquitin protein ligase bindingATP-dependent translocase ABCB1Homo sapiens (human)
ATPase-coupled transmembrane transporter activityATP-dependent translocase ABCB1Homo sapiens (human)
xenobiotic transmembrane transporter activityATP-dependent translocase ABCB1Homo sapiens (human)
carboxylic acid transmembrane transporter activityATP-dependent translocase ABCB1Homo sapiens (human)
phosphatidylcholine floppase activityATP-dependent translocase ABCB1Homo sapiens (human)
phosphatidylethanolamine flippase activityATP-dependent translocase ABCB1Homo sapiens (human)
ceramide floppase activityATP-dependent translocase ABCB1Homo sapiens (human)
floppase activityATP-dependent translocase ABCB1Homo sapiens (human)
monooxygenase activityCytochrome P450 3A4Homo sapiens (human)
steroid bindingCytochrome P450 3A4Homo sapiens (human)
iron ion bindingCytochrome P450 3A4Homo sapiens (human)
protein bindingCytochrome P450 3A4Homo sapiens (human)
steroid hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
retinoic acid 4-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
oxidoreductase activityCytochrome P450 3A4Homo sapiens (human)
oxygen bindingCytochrome P450 3A4Homo sapiens (human)
enzyme bindingCytochrome P450 3A4Homo sapiens (human)
heme bindingCytochrome P450 3A4Homo sapiens (human)
vitamin D3 25-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
caffeine oxidase activityCytochrome P450 3A4Homo sapiens (human)
quinine 3-monooxygenase activityCytochrome P450 3A4Homo sapiens (human)
testosterone 6-beta-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
1-alpha,25-dihydroxyvitamin D3 23-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
anandamide 8,9 epoxidase activityCytochrome P450 3A4Homo sapiens (human)
anandamide 11,12 epoxidase activityCytochrome P450 3A4Homo sapiens (human)
anandamide 14,15 epoxidase activityCytochrome P450 3A4Homo sapiens (human)
aromatase activityCytochrome P450 3A4Homo sapiens (human)
vitamin D 24-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
estrogen 16-alpha-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
estrogen 2-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
1,8-cineole 2-exo-monooxygenase activityCytochrome P450 3A4Homo sapiens (human)
3'-5'-RNA exonuclease activityReplicase polyprotein 1abSevere acute respiratory syndrome coronavirus 2
RNA-dependent RNA polymerase activityReplicase polyprotein 1abSevere acute respiratory syndrome coronavirus 2
cysteine-type endopeptidase activityReplicase polyprotein 1abSevere acute respiratory syndrome coronavirus 2
mRNA 5'-cap (guanine-N7-)-methyltransferase activityReplicase polyprotein 1abSevere acute respiratory syndrome coronavirus 2
mRNA (nucleoside-2'-O-)-methyltransferase activityReplicase polyprotein 1abSevere acute respiratory syndrome coronavirus 2
mRNA guanylyltransferase activityReplicase polyprotein 1abSevere acute respiratory syndrome coronavirus 2
RNA endonuclease activity, producing 3'-phosphomonoestersReplicase polyprotein 1abSevere acute respiratory syndrome coronavirus 2
ISG15-specific peptidase activityReplicase polyprotein 1abSevere acute respiratory syndrome coronavirus 2
5'-3' RNA helicase activityReplicase polyprotein 1abSevere acute respiratory syndrome coronavirus 2
protein guanylyltransferase activityReplicase polyprotein 1abSevere acute respiratory syndrome coronavirus 2
monooxygenase activityCytochrome P450 2D6Homo sapiens (human)
iron ion bindingCytochrome P450 2D6Homo sapiens (human)
oxidoreductase activityCytochrome P450 2D6Homo sapiens (human)
oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen, reduced flavin or flavoprotein as one donor, and incorporation of one atom of oxygenCytochrome P450 2D6Homo sapiens (human)
heme bindingCytochrome P450 2D6Homo sapiens (human)
anandamide 8,9 epoxidase activityCytochrome P450 2D6Homo sapiens (human)
anandamide 11,12 epoxidase activityCytochrome P450 2D6Homo sapiens (human)
anandamide 14,15 epoxidase activityCytochrome P450 2D6Homo sapiens (human)
monooxygenase activityCytochrome P450 2C9 Homo sapiens (human)
iron ion bindingCytochrome P450 2C9 Homo sapiens (human)
arachidonic acid epoxygenase activityCytochrome P450 2C9 Homo sapiens (human)
steroid hydroxylase activityCytochrome P450 2C9 Homo sapiens (human)
arachidonic acid 14,15-epoxygenase activityCytochrome P450 2C9 Homo sapiens (human)
arachidonic acid 11,12-epoxygenase activityCytochrome P450 2C9 Homo sapiens (human)
oxidoreductase activityCytochrome P450 2C9 Homo sapiens (human)
(S)-limonene 6-monooxygenase activityCytochrome P450 2C9 Homo sapiens (human)
(S)-limonene 7-monooxygenase activityCytochrome P450 2C9 Homo sapiens (human)
caffeine oxidase activityCytochrome P450 2C9 Homo sapiens (human)
(R)-limonene 6-monooxygenase activityCytochrome P450 2C9 Homo sapiens (human)
aromatase activityCytochrome P450 2C9 Homo sapiens (human)
heme bindingCytochrome P450 2C9 Homo sapiens (human)
oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen, reduced flavin or flavoprotein as one donor, and incorporation of one atom of oxygenCytochrome P450 2C9 Homo sapiens (human)
monooxygenase activityCytochrome P450 3A5Homo sapiens (human)
iron ion bindingCytochrome P450 3A5Homo sapiens (human)
protein bindingCytochrome P450 3A5Homo sapiens (human)
retinoic acid 4-hydroxylase activityCytochrome P450 3A5Homo sapiens (human)
oxidoreductase activityCytochrome P450 3A5Homo sapiens (human)
oxygen bindingCytochrome P450 3A5Homo sapiens (human)
heme bindingCytochrome P450 3A5Homo sapiens (human)
aromatase activityCytochrome P450 3A5Homo sapiens (human)
estrogen 16-alpha-hydroxylase activityCytochrome P450 3A5Homo sapiens (human)
testosterone 6-beta-hydroxylase activityCytochrome P450 3A5Homo sapiens (human)
ATPase-coupled transmembrane transporter activityATP-dependent translocase ABCB1Mus musculus (house mouse)
tachykinin receptor activitySubstance-K receptorHomo sapiens (human)
protein bindingSubstance-K receptorHomo sapiens (human)
substance K receptor activitySubstance-K receptorHomo sapiens (human)
retinoic acid bindingUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
enzyme inhibitor activityUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
steroid bindingUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
glucuronosyltransferase activityUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
enzyme bindingUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
protein homodimerization activityUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
protein heterodimerization activityUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
monooxygenase activityThromboxane-A synthase Homo sapiens (human)
thromboxane-A synthase activityThromboxane-A synthase Homo sapiens (human)
iron ion bindingThromboxane-A synthase Homo sapiens (human)
oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygenThromboxane-A synthase Homo sapiens (human)
heme bindingThromboxane-A synthase Homo sapiens (human)
12-hydroxyheptadecatrienoic acid synthase activityThromboxane-A synthase Homo sapiens (human)
hydroperoxy icosatetraenoate dehydratase activityThromboxane-A synthase Homo sapiens (human)
monooxygenase activityCytochrome P450 2C19Homo sapiens (human)
iron ion bindingCytochrome P450 2C19Homo sapiens (human)
steroid hydroxylase activityCytochrome P450 2C19Homo sapiens (human)
oxidoreductase activityCytochrome P450 2C19Homo sapiens (human)
(S)-limonene 6-monooxygenase activityCytochrome P450 2C19Homo sapiens (human)
(S)-limonene 7-monooxygenase activityCytochrome P450 2C19Homo sapiens (human)
oxygen bindingCytochrome P450 2C19Homo sapiens (human)
enzyme bindingCytochrome P450 2C19Homo sapiens (human)
heme bindingCytochrome P450 2C19Homo sapiens (human)
(R)-limonene 6-monooxygenase activityCytochrome P450 2C19Homo sapiens (human)
aromatase activityCytochrome P450 2C19Homo sapiens (human)
long-chain fatty acid omega-1 hydroxylase activityCytochrome P450 2C19Homo sapiens (human)
arachidonic acid epoxygenase activityCytochrome P450 2C19Homo sapiens (human)
oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen, reduced flavin or flavoprotein as one donor, and incorporation of one atom of oxygenCytochrome P450 2C19Homo sapiens (human)
G-protein alpha-subunit bindingMu-type opioid receptorHomo sapiens (human)
G protein-coupled receptor activityMu-type opioid receptorHomo sapiens (human)
beta-endorphin receptor activityMu-type opioid receptorHomo sapiens (human)
voltage-gated calcium channel activityMu-type opioid receptorHomo sapiens (human)
protein bindingMu-type opioid receptorHomo sapiens (human)
morphine receptor activityMu-type opioid receptorHomo sapiens (human)
G-protein beta-subunit bindingMu-type opioid receptorHomo sapiens (human)
neuropeptide bindingMu-type opioid receptorHomo sapiens (human)
vasopressin receptor activityVasopressin V1a receptorHomo sapiens (human)
protein kinase C bindingVasopressin V1a receptorHomo sapiens (human)
protein bindingVasopressin V1a receptorHomo sapiens (human)
peptide hormone bindingVasopressin V1a receptorHomo sapiens (human)
V1A vasopressin receptor bindingVasopressin V1a receptorHomo sapiens (human)
peptide bindingVasopressin V1a receptorHomo sapiens (human)
G protein-coupled opioid receptor activityKappa-type opioid receptorHomo sapiens (human)
protein bindingKappa-type opioid receptorHomo sapiens (human)
receptor serine/threonine kinase bindingKappa-type opioid receptorHomo sapiens (human)
dynorphin receptor activityKappa-type opioid receptorHomo sapiens (human)
neuropeptide bindingKappa-type opioid receptorHomo sapiens (human)
virus receptor activitySodium/bile acid cotransporterHomo sapiens (human)
protein bindingSodium/bile acid cotransporterHomo sapiens (human)
bile acid:sodium symporter activitySodium/bile acid cotransporterHomo sapiens (human)
organic cation transmembrane transporter activityMultidrug and toxin extrusion protein 2Homo sapiens (human)
antiporter activityMultidrug and toxin extrusion protein 2Homo sapiens (human)
transmembrane transporter activityMultidrug and toxin extrusion protein 2Homo sapiens (human)
xenobiotic transmembrane transporter activityMultidrug and toxin extrusion protein 2Homo sapiens (human)
polyspecific organic cation:proton antiporter activityMultidrug and toxin extrusion protein 2Homo sapiens (human)
inositol-1,3,4,5,6-pentakisphosphate kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol hexakisphosphate kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol heptakisphosphate kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol hexakisphosphate 5-kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
protein bindingInositol hexakisphosphate kinase 1Homo sapiens (human)
ATP bindingInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol hexakisphosphate 1-kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol hexakisphosphate 3-kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol 5-diphosphate pentakisphosphate 5-kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol diphosphate tetrakisphosphate kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
protein bindingCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATP bindingCanalicular multispecific organic anion transporter 1Homo sapiens (human)
organic anion transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type xenobiotic transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bilirubin transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATP hydrolysis activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATPase-coupled transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
protein bindingMultidrug and toxin extrusion protein 1Homo sapiens (human)
organic cation transmembrane transporter activityMultidrug and toxin extrusion protein 1Homo sapiens (human)
L-amino acid transmembrane transporter activityMultidrug and toxin extrusion protein 1Homo sapiens (human)
thiamine transmembrane transporter activityMultidrug and toxin extrusion protein 1Homo sapiens (human)
antiporter activityMultidrug and toxin extrusion protein 1Homo sapiens (human)
putrescine transmembrane transporter activityMultidrug and toxin extrusion protein 1Homo sapiens (human)
transmembrane transporter activityMultidrug and toxin extrusion protein 1Homo sapiens (human)
xenobiotic transmembrane transporter activityMultidrug and toxin extrusion protein 1Homo sapiens (human)
L-arginine transmembrane transporter activityMultidrug and toxin extrusion protein 1Homo sapiens (human)
polyspecific organic cation:proton antiporter activityMultidrug and toxin extrusion protein 1Homo sapiens (human)
protein bindingATPase family AAA domain-containing protein 5Homo sapiens (human)
ATP bindingATPase family AAA domain-containing protein 5Homo sapiens (human)
ATP hydrolysis activityATPase family AAA domain-containing protein 5Homo sapiens (human)
DNA clamp unloader activityATPase family AAA domain-containing protein 5Homo sapiens (human)
DNA bindingATPase family AAA domain-containing protein 5Homo sapiens (human)
RNA bindingAtaxin-2Homo sapiens (human)
epidermal growth factor receptor bindingAtaxin-2Homo sapiens (human)
protein bindingAtaxin-2Homo sapiens (human)
mRNA bindingAtaxin-2Homo sapiens (human)
serine-type endopeptidase inhibitor activitySolute carrier organic anion transporter family member 1B3Homo sapiens (human)
organic anion transmembrane transporter activitySolute carrier organic anion transporter family member 1B3Homo sapiens (human)
bile acid transmembrane transporter activitySolute carrier organic anion transporter family member 1B3Homo sapiens (human)
sodium-independent organic anion transmembrane transporter activitySolute carrier organic anion transporter family member 1B3Homo sapiens (human)
protein bindingBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
ATP bindingBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
organic anion transmembrane transporter activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
ABC-type xenobiotic transporter activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
urate transmembrane transporter activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
biotin transmembrane transporter activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
efflux transmembrane transporter activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
ATP hydrolysis activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
riboflavin transmembrane transporter activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
ATPase-coupled transmembrane transporter activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
identical protein bindingBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
protein homodimerization activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
xenobiotic transmembrane transporter activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
sphingolipid transporter activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
organic anion transmembrane transporter activitySolute carrier organic anion transporter family member 1B1Homo sapiens (human)
bile acid transmembrane transporter activitySolute carrier organic anion transporter family member 1B1Homo sapiens (human)
prostaglandin transmembrane transporter activitySolute carrier organic anion transporter family member 1B1Homo sapiens (human)
sodium-independent organic anion transmembrane transporter activitySolute carrier organic anion transporter family member 1B1Homo sapiens (human)
thyroid hormone transmembrane transporter activitySolute carrier organic anion transporter family member 1B1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (82)

Processvia Protein(s)Taxonomy
plasma membraneSolute carrier family 22 member 2Homo sapiens (human)
basal plasma membraneSolute carrier family 22 member 2Homo sapiens (human)
membraneSolute carrier family 22 member 2Homo sapiens (human)
basolateral plasma membraneSolute carrier family 22 member 2Homo sapiens (human)
apical plasma membraneSolute carrier family 22 member 2Homo sapiens (human)
extracellular exosomeSolute carrier family 22 member 2Homo sapiens (human)
presynapseSolute carrier family 22 member 2Homo sapiens (human)
plasma membraneSolute carrier family 22 member 1 Homo sapiens (human)
basal plasma membraneSolute carrier family 22 member 1 Homo sapiens (human)
membraneSolute carrier family 22 member 1 Homo sapiens (human)
basolateral plasma membraneSolute carrier family 22 member 1 Homo sapiens (human)
apical plasma membraneSolute carrier family 22 member 1 Homo sapiens (human)
lateral plasma membraneSolute carrier family 22 member 1 Homo sapiens (human)
presynapseSolute carrier family 22 member 1 Homo sapiens (human)
plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
basal plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
basolateral plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
nucleolusMultidrug resistance-associated protein 4Homo sapiens (human)
Golgi apparatusMultidrug resistance-associated protein 4Homo sapiens (human)
plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
membraneMultidrug resistance-associated protein 4Homo sapiens (human)
basolateral plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
apical plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
platelet dense granule membraneMultidrug resistance-associated protein 4Homo sapiens (human)
external side of apical plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
nucleoplasmNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
transcription regulator complexNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
nuclear bodyNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
intermediate filament cytoskeletonNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
chromatinNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
nucleusNuclear receptor subfamily 1 group I member 2Homo sapiens (human)
nuclear outer membraneSolute carrier family 22 member 3Homo sapiens (human)
plasma membraneSolute carrier family 22 member 3Homo sapiens (human)
endomembrane systemSolute carrier family 22 member 3Homo sapiens (human)
membraneSolute carrier family 22 member 3Homo sapiens (human)
basolateral plasma membraneSolute carrier family 22 member 3Homo sapiens (human)
apical plasma membraneSolute carrier family 22 member 3Homo sapiens (human)
mitochondrial membraneSolute carrier family 22 member 3Homo sapiens (human)
neuronal cell bodySolute carrier family 22 member 3Homo sapiens (human)
presynapseSolute carrier family 22 member 3Homo sapiens (human)
plasma membraneSolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
basal plasma membraneSolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
apical plasma membraneSolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
basolateral plasma membraneSolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
apical plasma membraneSolute carrier organic anion transporter family member 2B1 Homo sapiens (human)
basolateral plasma membraneBile salt export pumpHomo sapiens (human)
Golgi membraneBile salt export pumpHomo sapiens (human)
endosomeBile salt export pumpHomo sapiens (human)
plasma membraneBile salt export pumpHomo sapiens (human)
cell surfaceBile salt export pumpHomo sapiens (human)
apical plasma membraneBile salt export pumpHomo sapiens (human)
intercellular canaliculusBile salt export pumpHomo sapiens (human)
intracellular canaliculusBile salt export pumpHomo sapiens (human)
recycling endosomeBile salt export pumpHomo sapiens (human)
recycling endosome membraneBile salt export pumpHomo sapiens (human)
extracellular exosomeBile salt export pumpHomo sapiens (human)
membraneBile salt export pumpHomo sapiens (human)
extracellular spaceInterferon betaHomo sapiens (human)
extracellular regionInterferon betaHomo sapiens (human)
Golgi membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
endoplasmic reticulumHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
Golgi apparatusHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
plasma membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
cell surfaceHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
ER to Golgi transport vesicle membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
secretory granule membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
phagocytic vesicle membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
early endosome membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
recycling endosome membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
extracellular exosomeHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
lumenal side of endoplasmic reticulum membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
MHC class I protein complexHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
extracellular spaceHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
external side of plasma membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
extracellular regionAlbuminHomo sapiens (human)
extracellular spaceAlbuminHomo sapiens (human)
nucleusAlbuminHomo sapiens (human)
endoplasmic reticulumAlbuminHomo sapiens (human)
endoplasmic reticulum lumenAlbuminHomo sapiens (human)
Golgi apparatusAlbuminHomo sapiens (human)
platelet alpha granule lumenAlbuminHomo sapiens (human)
extracellular exosomeAlbuminHomo sapiens (human)
blood microparticleAlbuminHomo sapiens (human)
protein-containing complexAlbuminHomo sapiens (human)
cytoplasmAlbuminHomo sapiens (human)
nuclear bodyCellular tumor antigen p53Homo sapiens (human)
nucleusCellular tumor antigen p53Homo sapiens (human)
nucleoplasmCellular tumor antigen p53Homo sapiens (human)
replication forkCellular tumor antigen p53Homo sapiens (human)
nucleolusCellular tumor antigen p53Homo sapiens (human)
cytoplasmCellular tumor antigen p53Homo sapiens (human)
mitochondrionCellular tumor antigen p53Homo sapiens (human)
mitochondrial matrixCellular tumor antigen p53Homo sapiens (human)
endoplasmic reticulumCellular tumor antigen p53Homo sapiens (human)
centrosomeCellular tumor antigen p53Homo sapiens (human)
cytosolCellular tumor antigen p53Homo sapiens (human)
nuclear matrixCellular tumor antigen p53Homo sapiens (human)
PML bodyCellular tumor antigen p53Homo sapiens (human)
transcription repressor complexCellular tumor antigen p53Homo sapiens (human)
site of double-strand breakCellular tumor antigen p53Homo sapiens (human)
germ cell nucleusCellular tumor antigen p53Homo sapiens (human)
chromatinCellular tumor antigen p53Homo sapiens (human)
transcription regulator complexCellular tumor antigen p53Homo sapiens (human)
protein-containing complexCellular tumor antigen p53Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 1A2Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 1A2Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 1A2Homo sapiens (human)
cytoplasmATP-dependent translocase ABCB1Homo sapiens (human)
plasma membraneATP-dependent translocase ABCB1Homo sapiens (human)
cell surfaceATP-dependent translocase ABCB1Homo sapiens (human)
membraneATP-dependent translocase ABCB1Homo sapiens (human)
apical plasma membraneATP-dependent translocase ABCB1Homo sapiens (human)
extracellular exosomeATP-dependent translocase ABCB1Homo sapiens (human)
external side of apical plasma membraneATP-dependent translocase ABCB1Homo sapiens (human)
plasma membraneATP-dependent translocase ABCB1Homo sapiens (human)
cytoplasmCytochrome P450 3A4Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 3A4Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 3A4Homo sapiens (human)
double membrane vesicle viral factory outer membraneReplicase polyprotein 1abSevere acute respiratory syndrome coronavirus 2
mitochondrionCytochrome P450 2D6Homo sapiens (human)
endoplasmic reticulumCytochrome P450 2D6Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 2D6Homo sapiens (human)
cytoplasmCytochrome P450 2D6Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2D6Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 2C9 Homo sapiens (human)
plasma membraneCytochrome P450 2C9 Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2C9 Homo sapiens (human)
cytoplasmCytochrome P450 2C9 Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2C9 Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 3A5Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 3A5Homo sapiens (human)
plasma membraneATP-dependent translocase ABCB1Mus musculus (house mouse)
plasma membraneSubstance-K receptorHomo sapiens (human)
sperm flagellumSubstance-K receptorHomo sapiens (human)
sperm headSubstance-K receptorHomo sapiens (human)
sperm midpieceSubstance-K receptorHomo sapiens (human)
sperm midpieceSubstance-K receptorHomo sapiens (human)
plasma membraneSubstance-K receptorHomo sapiens (human)
endoplasmic reticulumUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
endoplasmic reticulum membraneUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
plasma membraneUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
perinuclear region of cytoplasmUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
endoplasmic reticulum chaperone complexUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
cytochrome complexUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
endoplasmic reticulumUDP-glucuronosyltransferase 1A1 Homo sapiens (human)
endoplasmic reticulumThromboxane-A synthase Homo sapiens (human)
endoplasmic reticulum membraneThromboxane-A synthase Homo sapiens (human)
cytosolThromboxane-A synthase Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 2C19Homo sapiens (human)
plasma membraneCytochrome P450 2C19Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2C19Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2C19Homo sapiens (human)
cytoplasmCytochrome P450 2C19Homo sapiens (human)
endosomeMu-type opioid receptorHomo sapiens (human)
endoplasmic reticulumMu-type opioid receptorHomo sapiens (human)
Golgi apparatusMu-type opioid receptorHomo sapiens (human)
plasma membraneMu-type opioid receptorHomo sapiens (human)
axonMu-type opioid receptorHomo sapiens (human)
dendriteMu-type opioid receptorHomo sapiens (human)
perikaryonMu-type opioid receptorHomo sapiens (human)
synapseMu-type opioid receptorHomo sapiens (human)
plasma membraneMu-type opioid receptorHomo sapiens (human)
neuron projectionMu-type opioid receptorHomo sapiens (human)
endosomeVasopressin V1a receptorHomo sapiens (human)
plasma membraneVasopressin V1a receptorHomo sapiens (human)
endocytic vesicleVasopressin V1a receptorHomo sapiens (human)
plasma membraneVasopressin V1a receptorHomo sapiens (human)
nucleoplasmKappa-type opioid receptorHomo sapiens (human)
mitochondrionKappa-type opioid receptorHomo sapiens (human)
cytosolKappa-type opioid receptorHomo sapiens (human)
plasma membraneKappa-type opioid receptorHomo sapiens (human)
membraneKappa-type opioid receptorHomo sapiens (human)
sarcoplasmic reticulumKappa-type opioid receptorHomo sapiens (human)
T-tubuleKappa-type opioid receptorHomo sapiens (human)
dendriteKappa-type opioid receptorHomo sapiens (human)
synaptic vesicle membraneKappa-type opioid receptorHomo sapiens (human)
presynaptic membraneKappa-type opioid receptorHomo sapiens (human)
perikaryonKappa-type opioid receptorHomo sapiens (human)
axon terminusKappa-type opioid receptorHomo sapiens (human)
postsynaptic membraneKappa-type opioid receptorHomo sapiens (human)
plasma membraneKappa-type opioid receptorHomo sapiens (human)
neuron projectionKappa-type opioid receptorHomo sapiens (human)
plasma membraneSodium/bile acid cotransporterHomo sapiens (human)
basolateral plasma membraneSodium/bile acid cotransporterHomo sapiens (human)
plasma membraneMultidrug and toxin extrusion protein 2Homo sapiens (human)
apical plasma membraneMultidrug and toxin extrusion protein 2Homo sapiens (human)
membraneMultidrug and toxin extrusion protein 2Homo sapiens (human)
fibrillar centerInositol hexakisphosphate kinase 1Homo sapiens (human)
nucleoplasmInositol hexakisphosphate kinase 1Homo sapiens (human)
cytosolInositol hexakisphosphate kinase 1Homo sapiens (human)
nucleusInositol hexakisphosphate kinase 1Homo sapiens (human)
cytoplasmInositol hexakisphosphate kinase 1Homo sapiens (human)
plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
cell surfaceCanalicular multispecific organic anion transporter 1Homo sapiens (human)
apical plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
intercellular canaliculusCanalicular multispecific organic anion transporter 1Homo sapiens (human)
apical plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
plasma membraneMultidrug and toxin extrusion protein 1Homo sapiens (human)
basolateral plasma membraneMultidrug and toxin extrusion protein 1Homo sapiens (human)
apical plasma membraneMultidrug and toxin extrusion protein 1Homo sapiens (human)
membraneMultidrug and toxin extrusion protein 1Homo sapiens (human)
Elg1 RFC-like complexATPase family AAA domain-containing protein 5Homo sapiens (human)
nucleusATPase family AAA domain-containing protein 5Homo sapiens (human)
cytoplasmAtaxin-2Homo sapiens (human)
Golgi apparatusAtaxin-2Homo sapiens (human)
trans-Golgi networkAtaxin-2Homo sapiens (human)
cytosolAtaxin-2Homo sapiens (human)
cytoplasmic stress granuleAtaxin-2Homo sapiens (human)
membraneAtaxin-2Homo sapiens (human)
perinuclear region of cytoplasmAtaxin-2Homo sapiens (human)
ribonucleoprotein complexAtaxin-2Homo sapiens (human)
cytoplasmic stress granuleAtaxin-2Homo sapiens (human)
plasma membraneSolute carrier organic anion transporter family member 1B3Homo sapiens (human)
basal plasma membraneSolute carrier organic anion transporter family member 1B3Homo sapiens (human)
basolateral plasma membraneSolute carrier organic anion transporter family member 1B3Homo sapiens (human)
nucleoplasmBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
plasma membraneBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
apical plasma membraneBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
brush border membraneBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
mitochondrial membraneBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
membrane raftBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
external side of apical plasma membraneBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
plasma membraneBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
plasma membraneSolute carrier organic anion transporter family member 1B1Homo sapiens (human)
basal plasma membraneSolute carrier organic anion transporter family member 1B1Homo sapiens (human)
membraneSolute carrier organic anion transporter family member 1B1Homo sapiens (human)
basolateral plasma membraneSolute carrier organic anion transporter family member 1B1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (793)

Assay IDTitleYearJournalArticle
AID1347160Primary screen NINDS Rhodamine qHTS for Zika virus inhibitors2020Proceedings of the National Academy of Sciences of the United States of America, 12-08, Volume: 117, Issue:49
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors.
AID1296008Cytotoxic Profiling of Annotated Libraries Using Quantitative High-Throughput Screening2020SLAS discovery : advancing life sciences R & D, 01, Volume: 25, Issue:1
Cytotoxic Profiling of Annotated and Diverse Chemical Libraries Using Quantitative High-Throughput Screening.
AID1346987P-glycoprotein substrates identified in KB-8-5-11 adenocarcinoma cell line, qHTS therapeutic library screen2019Molecular pharmacology, 11, Volume: 96, Issue:5
A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
AID1508591NCATS Rat Liver Microsome Stability Profiling2020Scientific reports, 11-26, Volume: 10, Issue:1
Retrospective assessment of rat liver microsomal stability at NCATS: data and QSAR models.
AID1346986P-glycoprotein substrates identified in KB-3-1 adenocarcinoma cell line, qHTS therapeutic library screen2019Molecular pharmacology, 11, Volume: 96, Issue:5
A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
AID1645871NCATS Parallel Artificial Membrane Permeability Assay (PAMPA) Profiling in pH 5 buffer2022Bioorganic & medicinal chemistry, 02-15, Volume: 56Using in vitro ADME data for lead compound selection: An emphasis on PAMPA pH 5 permeability and oral bioavailability.
AID1347159Primary screen GU Rhodamine qHTS for Zika virus inhibitors: Unlinked NS2B-NS3 protease assay2020Proceedings of the National Academy of Sciences of the United States of America, 12-08, Volume: 117, Issue:49
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors.
AID1645848NCATS Kinetic Aqueous Solubility Profiling2019Bioorganic & medicinal chemistry, 07-15, Volume: 27, Issue:14
Predictive models of aqueous solubility of organic compounds built on A large dataset of high integrity.
AID353748Cmax in po dosed Beagle dog at dose molar equivalent to 5 mg/kg RTV coadministered with LPV dosed as 5% dextrose containing solution2009Journal of medicinal chemistry, May-14, Volume: 52, Issue:9
Water-soluble prodrugs of the human immunodeficiency virus protease inhibitors lopinavir and ritonavir.
AID1210949Ratio of Kinact to Ki for CYP3A4 (unknown origin)2011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Inactivation of cytochrome P450 (P450) 3A4 but not P450 3A5 by OSI-930, a thiophene-containing anticancer drug.
AID679071TP_TRANSPORTER: transepithelial transport of Ritonavir at a concentration of 0.1 uM in Caco-2 cells2003Pharmaceutical research, Aug, Volume: 20, Issue:8
Novel experimental parameters to quantify the modulation of absorptive and secretory transport of compounds by P-glycoprotein in cell culture models of intestinal epithelium.
AID372212Tmax in human at 100 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 102007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID525108Antiviral activity against HIV 1 RIN harboring integrase gene infected in MT-4 cells assessed as inhibition of virus-induced cytopathic effect by MTT assay after 20 passages selected in presence of compound2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Mutations in human immunodeficiency virus type 1 integrase confer resistance to the naphthyridine L-870,810 and cross-resistance to the clinical trial drug GS-9137.
AID321698Ratio of EC50 for HIV1 mutant strain 6 to EC50 for wild type HIV12008Bioorganic & medicinal chemistry, Feb-01, Volume: 16, Issue:3
Structure-activity relationships of novel HIV-1 protease inhibitors containing the 3-amino-2-chlorobenzoyl-allophenylnorstatine structure.
AID322096Antiviral activity against HIV1 LAI in MT2 cells assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID416864Inhibition of human MDR1-dependent accumulation of calcein-AM expressed in MDCK2 cells2007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Effects of human immunodeficiency virus protease inhibitors on the intestinal absorption of tenofovir disoproxil fumarate in vitro.
AID543372AUC (0 to 24 hrs) in HIV-infected patient at 100 mg, qd co-administered with 100 mg minocycline and 250 mg valporic acid, bid for 15 days2008Antimicrobial agents and chemotherapy, Sep, Volume: 52, Issue:9
Effects of minocycline and valproic acid coadministration on atazanavir plasma concentrations in human immunodeficiency virus-infected adults receiving atazanavir-ritonavir.
AID557238Ratio of drug level before intrapartum initiation to 30 days pre-last dose of HIV-infected Thai pregnant women serum at 100 mg, po bid in combination with 400 mg, po bid Lopinavir2009Antimicrobial agents and chemotherapy, May, Volume: 53, Issue:5
Early postpartum pharmacokinetics of lopinavir initiated intrapartum in Thai women.
AID1889303Permeability in human Caco-2 cells2022Journal of medicinal chemistry, 02-10, Volume: 65, Issue:3
Dose Number as a Tool to Guide Lead Optimization for Orally Bioavailable Compounds in Drug Discovery.
AID705597Time dependent inhibition of CYP3A4-mediated testosterone-6-beta hydroxylation in human liver microsome2012Journal of medicinal chemistry, Jun-14, Volume: 55, Issue:11
Mechanism-based inactivation (MBI) of cytochrome P450 enzymes: structure-activity relationships and discovery strategies to mitigate drug-drug interaction risks.
AID624623Apparent permeability (Papp) from basolateral to apical side determined in MDR1-MDCKII cells2001The Journal of pharmacology and experimental therapeutics, Nov, Volume: 299, Issue:2
Rational use of in vitro P-glycoprotein assays in drug discovery.
AID1221978Transporter substrate index ratio of permeability from apical to basolateral side in human Caco2 cells at 10 uM up to 120 mins by HPLC-MC analysis in presence of 10 uM of MRP2 inhibitor MK5712011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Attenuation of intestinal absorption by major efflux transporters: quantitative tools and strategies using a Caco-2 model.
AID1578465Drug concentration in total heart in Wistar Han rat at 1 to 2 mg/kg, iv infused for 10 to 20 hrs by LC-MS/MS analysis2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID225562Dissociation constant obtained by inhibition of mutant HIV-protease (A-44)2000Journal of medicinal chemistry, Sep-07, Volume: 43, Issue:18
Identification of MK-944a: a second clinical candidate from the hydroxylaminepentanamide isostere series of HIV protease inhibitors.
AID239810Equilibrium dissociation constant for the interaction between the compound and serum albumin2005Journal of medicinal chemistry, May-19, Volume: 48, Issue:10
Early absorption and distribution analysis of antitumor and anti-AIDS drugs: lipid membrane and plasma protein interactions.
AID104303Anti-HIV-1 activity against mutant HIV-1 in MT-4 cells (mutation selected with ritonavir)2001Journal of medicinal chemistry, Sep-13, Volume: 44, Issue:19
Design and synthesis of potent C(2)-symmetric diol-based HIV-1 protease inhibitors: effects of fluoro substitution.
AID738134Inhibition of C-terminal His-tagged wild type CYP3A4 (unknown origin)-mediated hydroxylation of 7-benzyloxy-4-trifluoromethylcoumarin expressed in Escherichia coli after 5 mins by fluorimetric analysis2013Journal of medicinal chemistry, May-09, Volume: 56, Issue:9
Pyridine-substituted desoxyritonavir is a more potent inhibitor of cytochrome P450 3A4 than ritonavir.
AID1474151Ratio of drug concentration at steady state in human at 600 mg, po BID after 12 hrs to IC50 for human MRP4 overexpressed in Sf9 insect cells2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID588215FDA HLAED, alkaline phosphatase increase2004Current drug discovery technologies, Dec, Volume: 1, Issue:4
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
AID518055Toxicity in healthy human assessed as headache at 100 mg/kg, po administered as single dose2010Antimicrobial agents and chemotherapy, Jun, Volume: 54, Issue:6
Effect of vicriviroc on the QT/corrected QT interval and central nervous system in healthy subjects.
AID290996Selectivity index, Ratio of LC50 for MT4 cells to EC50 for HIV1 3B2007Bioorganic & medicinal chemistry letters, Aug-01, Volume: 17, Issue:15
Synthesis and antiviral property of allophenylnorstatine-based HIV protease inhibitors incorporating D-cysteine derivatives as P2/P3 moieties.
AID668005Inhibition of human CYP3A4 in liver microsomes2012Bioorganic & medicinal chemistry letters, May-01, Volume: 22, Issue:9
Preclinical metabolism of LB42908, a novel farnesyl transferase inhibitor, and its effects on the cytochrome P450 isozyme activities.
AID525170Antimicrobial activity against chloroquine-resistant Plasmodium chabaudi ASCQ infected in NIH mice (Mus musculus) assessed as potentiation of 2.5 mg/kg chloroquine-mediated antimalarial activity at 10 to 160 mg/kg, perorally administered after 72 hrs post2008Antimicrobial agents and chemotherapy, Jul, Volume: 52, Issue:7
Synergy of human immunodeficiency virus protease inhibitors with chloroquine against Plasmodium falciparum in vitro and Plasmodium chabaudi in vivo.
AID257273Antiviral activity against Lopinavir resistant HIV A17 mutant strain2005Bioorganic & medicinal chemistry letters, Dec-15, Volume: 15, Issue:24
Synthesis and activity of N-acyl azacyclic urea HIV-1 protease inhibitors with high potency against multiple drug resistant viral strains.
AID588213Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in non-rodents2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID681120TP_TRANSPORTER: inhibition of Rhodamine 123 efflux in Caco-2 cells2003Journal of medicinal chemistry, Apr-24, Volume: 46, Issue:9
Comparison of in vitro P-glycoprotein screening assays: recommendations for their use in drug discovery.
AID572586Half life in HIV-1 infected patient at 100 mg, po bid coadministered with 300 mg, po qd of atazanavir and NRTI2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Decrease of atazanavir and lopinavir plasma concentrations in a boosted double human immunodeficiency virus protease inhibitor salvage regimen.
AID322123Antiviral activity against HIV1 92TH019 R5 subtype E in phytohemagglutininin-activated PBMCs assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID557223Cmax in HIV-infected Thai pregnant women at 100 mg, po bid initiated intrapartum in combination with 400 mg, po bid Lopinavir measured within 72 hrs postpartum2009Antimicrobial agents and chemotherapy, May, Volume: 53, Issue:5
Early postpartum pharmacokinetics of lopinavir initiated intrapartum in Thai women.
AID699541Inhibition of human liver OATP2B1 expressed in HEK293 Flp-In cells assessed as reduction in [3H]E3S uptake at 20 uM incubated for 5 mins by scintillation counting2012Journal of medicinal chemistry, May-24, Volume: 55, Issue:10
Classification of inhibitors of hepatic organic anion transporting polypeptides (OATPs): influence of protein expression on drug-drug interactions.
AID525173Toxicity in NIH mice (Mus musculus) infected with chloroquine-resistant Plasmodium chabaudi ASCQ assessed as change in body weight at 10 to 160 mg/kg, perorally measured on day 4 postinfection2008Antimicrobial agents and chemotherapy, Jul, Volume: 52, Issue:7
Synergy of human immunodeficiency virus protease inhibitors with chloroquine against Plasmodium falciparum in vitro and Plasmodium chabaudi in vivo.
AID738133Binding affinity to CYP3A4 (3-22)-deleted S119A mutant (unknown origin) expressed in Escherichia coli by spectrophotometric analysis2013Journal of medicinal chemistry, May-09, Volume: 56, Issue:9
Pyridine-substituted desoxyritonavir is a more potent inhibitor of cytochrome P450 3A4 than ritonavir.
AID1219732Inhibition of CYP3A4-mediated lopinavir bioactivation in human liver microsomes assessed as inhibition of GSH-conjugated hydroxylated DMP adduct formation adduct formation at 2 uM after after 30 mins by UPLC-TOFMS analysis2012Drug metabolism and disposition: the biological fate of chemicals, Jan, Volume: 40, Issue:1
CYP3A4-mediated lopinavir bioactivation and its inhibition by ritonavir.
AID1610045Antiviral activity against HIV1 NL4.3 infected in human TZM-bl cells measured upto 24 hrs by bright Glo-luciferase reporter gene assay2019European journal of medicinal chemistry, Dec-01, Volume: 183Synthesis and in-vitro anti-HIV-1 evaluation of novel pyrazolo[4,3-c]pyridin-4-one derivatives.
AID372211Cmin in human at 100 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 102007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID470602Inhibition of human recombinant CYP3A4-mediated oxidation of 7-benzyloxyquinoline2009Bioorganic & medicinal chemistry letters, Sep-15, Volume: 19, Issue:18
Synthesis and evaluation of inhibitors of cytochrome P450 3A (CYP3A) for pharmacokinetic enhancement of drugs.
AID1221965Transporter substrate index of efflux ratio in human Caco2 cells at 10 uM up to 120 mins by HPLC-MC analysis in presence of 1 uM of P-gp inhibitor LY3359792011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Attenuation of intestinal absorption by major efflux transporters: quantitative tools and strategies using a Caco-2 model.
AID1267624Antiviral activity against ritonavir-resistant HIV1 3B/RIT infected in human MT4 cells2015Journal of medicinal chemistry, Dec-24, Volume: 58, Issue:24
Design and Synthesis of DiselenoBisBenzamides (DISeBAs) as Nucleocapsid Protein 7 (NCp7) Inhibitors with anti-HIV Activity.
AID1221982Fraction absorbed in human2011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Attenuation of intestinal absorption by major efflux transporters: quantitative tools and strategies using a Caco-2 model.
AID1578468Unbound tissue partition coefficient, ratio of drug level in heart to plasma in Wistar Han rat at 1 to 2 mg/kg, iv infused for 10 to 20 hrs by LC-MS/MS analysis2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID415240Oral bioavailability in HIV1 infected patient at 600 mg administered as single dose coadministered with 100 mg rotonavir bid2009Journal of medicinal chemistry, Apr-23, Volume: 52, Issue:8
Harnessing nature's insight: design of aspartyl protease inhibitors from treatment of drug-resistant HIV to Alzheimer's disease.
AID680009TP_TRANSPORTER: inhibition of Fexofenadine uptake (Fexofenadine: 2 uM, Ritonavir: 100 uM) in Xenopus laevis oocytes1999Drug metabolism and disposition: the biological fate of chemicals, Aug, Volume: 27, Issue:8
OATP and P-glycoprotein transporters mediate the cellular uptake and excretion of fexofenadine.
AID269307Antiviral activity against HIV2 EHO isolate in human MT2 cells2006Journal of medicinal chemistry, Aug-24, Volume: 49, Issue:17
Structure-based design of novel HIV-1 protease inhibitors to combat drug resistance.
AID321660Inhibition of HIV1 protease2008Bioorganic & medicinal chemistry, Feb-01, Volume: 16, Issue:3
Structure-activity relationships of novel HIV-1 protease inhibitors containing the 3-amino-2-chlorobenzoyl-allophenylnorstatine structure.
AID625282Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for cirrhosis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1219727Inhibition of CYP3A4-mediated lopinavir bioactivation in human liver microsomes assessed as inhibition of lopinavir-GSH adduct I and II formation at 2 uM after after 50 mins by UPLC-TOFMS analysis2012Drug metabolism and disposition: the biological fate of chemicals, Jan, Volume: 40, Issue:1
CYP3A4-mediated lopinavir bioactivation and its inhibition by ritonavir.
AID322120Antiviral activity against HIV1 92UG037 subtype A R5 in phytohemagglutininin-activated PBMCs assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID269314Antiviral activity against multi drug-resistant HIV1 TM variant in human PHA-PBMC cells2006Journal of medicinal chemistry, Aug-24, Volume: 49, Issue:17
Structure-based design of novel HIV-1 protease inhibitors to combat drug resistance.
AID369177Apparent elimination half life in HIV1 infected human at 100 mg, po administered twice daily2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
Association of saquinavir plasma concentrations with side effects but not with antiretroviral outcome in patients infected with protease inhibitor-susceptible human immunodeficiency virus type 1.
AID557283Ratio of EC50 for HIV1 G harboring L10I/V11I/T12E/I15V/L19I/R41K/M46L/L63P/A71T/V82A/L90M in protease encoding region to EC50 for HIV1 ERS104pre2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID589163Mechanism based inhibition of human cytochrome P450 3A4 using intestinal microsomes2005Current drug metabolism, Oct, Volume: 6, Issue:5
Cytochrome p450 enzymes mechanism based inhibitors: common sub-structures and reactivity.
AID553574Antiviral activity against HIV1 ERS104pre infected in human PHA-PBC assessed as inhibition of p24 Gag protein production by ELISA2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID589167Mechanism based inhibition of human cytochrome P450 3A52005Current drug metabolism, Oct, Volume: 6, Issue:5
Cytochrome p450 enzymes mechanism based inhibitors: common sub-structures and reactivity.
AID519606Antimicrobial activity against Plasmodium yoelii 265 liver infected in mammalian hepatocytes after 48 hrs2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
New active drugs against liver stages of Plasmodium predicted by molecular topology.
AID588209Literature-mined public compounds from Greene et al multi-species hepatotoxicity modelling dataset2010Chemical research in toxicology, Jul-19, Volume: 23, Issue:7
Developing structure-activity relationships for the prediction of hepatotoxicity.
AID427584Inhibition of SAP4-dependent growth in Candida albicans SAP2MS4B expressing SAP4ex4A at 100 uM incubated in YCB-BSA medium2008Antimicrobial agents and chemotherapy, Jan, Volume: 52, Issue:1
Tetracycline-inducible expression of individual secreted aspartic proteases in Candida albicans allows isoenzyme-specific inhibitor screening.
AID721740Selectivity ratio of IC50 for human OCT2-mediated ASP+ uptake expressed in HEK293 cells to IC50 for human MATE1-mediated ASP+ uptake expressed in HEK293 cells2013Journal of medicinal chemistry, Feb-14, Volume: 56, Issue:3
Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling.
AID1473739Inhibition of human MRP2 overexpressed in Sf9 cell membrane vesicles assessed as uptake of [3H]-estradiol-17beta-D-glucuronide in presence of ATP and GSH measured after 20 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID82944Tested for inhibitor binding of D25N/V82A mutant of HIV PR2004Journal of medicinal chemistry, Apr-08, Volume: 47, Issue:8
A phenylnorstatine inhibitor binding to HIV-1 protease: geometry, protonation, and subsite-pocket interactions analyzed at atomic resolution.
AID150755Inhibition of P-glycoprotein using calcein-AM assay transfected in porcine PBCEC2003Journal of medicinal chemistry, Apr-24, Volume: 46, Issue:9
Comparison of in vitro P-glycoprotein screening assays: recommendations for their use in drug discovery.
AID558384Drug level in HIV-infected pregnant woman amniotic fluid at 100 mg, po BID by HPLC/UV analysis2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Genital tract, cord blood, and amniotic fluid exposures of seven antiretroviral drugs during and after pregnancy in human immunodeficiency virus type 1-infected women.
AID227700Anticonvulsant activity2003Bioorganic & medicinal chemistry letters, Aug-18, Volume: 13, Issue:16
Topological virtual screening: a way to find new anticonvulsant drugs from chemical diversity.
AID88987Antiviral activity was assessed against I54I/V, A71V, V82A, L90L/M viral strains isolated from patient-235 pretreated with ritonavir (4 fold) at 36th week1998Bioorganic & medicinal chemistry letters, Dec-15, Volume: 8, Issue:24
Potent piperazine hydroxyethylamine HIV protease inhibitors containing novel P3 ligands.
AID1069007Activation of human PXR by cell based luciferase reporter gene assay2014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Structure-activity relationships of diamine inhibitors of cytochrome P450 (CYP) 3A as novel pharmacoenhancers, part I: core region.
AID572590AUC in HIV-1 infected patient at 100 mg, po bid coadministered with 400 mg, po bid of Lopinavir and NRTI2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Decrease of atazanavir and lopinavir plasma concentrations in a boosted double human immunodeficiency virus protease inhibitor salvage regimen.
AID1217726Time dependent inhibition of CYP3A4 (unknown origin) at 1 uM by LC/MS system2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Combination of GSH trapping and time-dependent inhibition assays as a predictive method of drugs generating highly reactive metabolites.
AID235093Resistance of constructed mutant 461/47V/50V virus was calculated as (IC90 for a mutant virus / IC90 of wt HXB2 virus). 1997Journal of medicinal chemistry, Jan-17, Volume: 40, Issue:2
Cyclic urea amides: HIV-1 protease inhibitors with low nanomolar potency against both wild type and protease inhibitor resistant mutants of HIV.
AID235099Resistance of constructed mutant MK639 virus was calculated as (IC90 for a mutant virus / IC90 of wt HXB2 virus). 1997Journal of medicinal chemistry, Jan-17, Volume: 40, Issue:2
Cyclic urea amides: HIV-1 protease inhibitors with low nanomolar potency against both wild type and protease inhibitor resistant mutants of HIV.
AID738317Inhibition of HIV1 protease at 50 uM relative to control2013Bioorganic & medicinal chemistry, Apr-01, Volume: 21, Issue:7
Synthesis and evaluation of coumarin derivatives as potential dual-action HIV-1 protease and reverse transcriptase inhibitors.
AID679171TP_TRANSPORTER: increase in mitoxantrone intracellular accumulation in BCRP-expressing HEK cells2004The Journal of pharmacology and experimental therapeutics, Jul, Volume: 310, Issue:1
HIV protease inhibitors are inhibitors but not substrates of the human breast cancer resistance protein (BCRP/ABCG2).
AID721748Inhibition of human MATE2K-mediated ASP+ uptake expressed in HEK293 cells up to 500 uM after 1.5 mins by fluorescence assay2013Journal of medicinal chemistry, Feb-14, Volume: 56, Issue:3
Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling.
AID681122TP_TRANSPORTER: inhibition of Calcein-AM efflux in MDR1-expressing LLC-PK1 cells2003Journal of medicinal chemistry, Apr-24, Volume: 46, Issue:9
Comparison of in vitro P-glycoprotein screening assays: recommendations for their use in drug discovery.
AID575064Antiviral activity against Human immunodeficiency virus 1 harboring M46I and L76V mutations in viral protease assessed as fold change in drug susceptibility relative to wild type2010Antimicrobial agents and chemotherapy, Nov, Volume: 54, Issue:11
Prevalence, mutation patterns, and effects on protease inhibitor susceptibility of the L76V mutation in HIV-1 protease.
AID525113Antiviral activity against HIV 1 NL4.3 integrase E92Q mutant infected in human MT-4 cells assessed as inhibition of virus-induced cytopathic effect by MTT assay2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Mutations in human immunodeficiency virus type 1 integrase confer resistance to the naphthyridine L-870,810 and cross-resistance to the clinical trial drug GS-9137.
AID557290Antiviral activity against HIV1 NL4-3 harboring L10F/M46I/I54V/V82A amino acid substitution in protease encoding region infected in human MT4 cells assessed as inhibition of p24 Gag protein production selected at 1 uM of Lopinavir by ELISA2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID321689Antiviral activity against HIV1 mutant strain 42008Bioorganic & medicinal chemistry, Feb-01, Volume: 16, Issue:3
Structure-activity relationships of novel HIV-1 protease inhibitors containing the 3-amino-2-chlorobenzoyl-allophenylnorstatine structure.
AID321691Antiviral activity against HIV1 mutant strain 62008Bioorganic & medicinal chemistry, Feb-01, Volume: 16, Issue:3
Structure-activity relationships of novel HIV-1 protease inhibitors containing the 3-amino-2-chlorobenzoyl-allophenylnorstatine structure.
AID1065160Inhibition of Plasmodium falciparum DC6 plasmepsin-5 using DABCYL-GNKRTLAQKQG-EDANS as substrate measured every 15 mins of 75 mins by fluorescence assay2014ACS medicinal chemistry letters, Jan-09, Volume: 5, Issue:1
Evaluation of aminohydantoins as a novel class of antimalarial agents.
AID243849Percent inhibition of human immunodeficiency virus type I (HIV-1) protease was determined at 50 uM concentration2004Bioorganic & medicinal chemistry letters, Dec-06, Volume: 14, Issue:23
Identification of peptidomimetic HTLV-I protease inhibitors containing hydroxymethylcarbonyl (HMC) isostere as the transition-state mimic.
AID408213Antiviral activity against ritonavir and nelfinavir-resistant HIV1 NL4322008Journal of medicinal chemistry, May-22, Volume: 51, Issue:10
Combination of non-natural D-amino acid derivatives and allophenylnorstatine-dimethylthioproline scaffold in HIV protease inhibitors have high efficacy in mutant HIV.
AID680736TP_TRANSPORTER: inhibition of E217betaG uptake (E217betaG: 0.5 uM, Ritonavir: 10 uM) in OATP-C-expressing HeLa cells2003The Journal of pharmacology and experimental therapeutics, Jan, Volume: 304, Issue:1
Human organic anion transporting polypeptide-C (SLC21A6) is a major determinant of rifampin-mediated pregnane X receptor activation.
AID427586Inhibition of SAP2-dependent growth in Candida albicans SAP2MS4B expressing SAP2ex4A at 100 uM incubated in YCB-BSA medium2008Antimicrobial agents and chemotherapy, Jan, Volume: 52, Issue:1
Tetracycline-inducible expression of individual secreted aspartic proteases in Candida albicans allows isoenzyme-specific inhibitor screening.
AID1218865Inhibition of OATP1B1 (unknown origin) expressed in HEK293 cells using estrone-3-sulfate substrate2012Drug metabolism and disposition: the biological fate of chemicals, Aug, Volume: 40, Issue:8
The development, characterization, and application of an OATP1B1 inhibition assay in drug discovery.
AID1221973Efflux ratio of permeability from apical to basolateral side over basolateral to apical side of human Caco2 cells at 10 uM up to 120 mins by HPLC-MC analysis in presence of 10 uM of MRP2 inhibitor MK5712011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Attenuation of intestinal absorption by major efflux transporters: quantitative tools and strategies using a Caco-2 model.
AID374592Antiviral activity against wild type HIV1 NL4-3 infected in MT4 cells after 6 days by MTT assay2007Antimicrobial agents and chemotherapy, Nov, Volume: 51, Issue:11
In vitro antiviral activity and cross-resistance profile of PL-100, a novel protease inhibitor of human immunodeficiency virus type 1.
AID1594780Toxicity in human HepG2 cells measured after 48 hrs by calcein-AM/Hoechst 33342 staining based high-content imaging analysis2019Journal of natural products, 06-28, Volume: 82, Issue:6
Natural-Product-Inspired Compounds as Countermeasures against the Liver Carcinogen Aflatoxin B
AID558391Ratio of drug level in HIV-infected pregnant woman cord blood plasma to maternal blood plasma at 100 mg, po BID by HPLC/UV analysis2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Genital tract, cord blood, and amniotic fluid exposures of seven antiretroviral drugs during and after pregnancy in human immunodeficiency virus type 1-infected women.
AID588219FDA HLAED, gamma-glutamyl transferase (GGT) increase2004Current drug discovery technologies, Dec, Volume: 1, Issue:4
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
AID263207Antiviral activity against HIV1 HXB2 in MT4 cells2006Bioorganic & medicinal chemistry letters, Apr-01, Volume: 16, Issue:7
Ultra-potent P1 modified arylsulfonamide HIV protease inhibitors: the discovery of GW0385.
AID88990Antiviral activity was assessed against K20K/R, M36M/I, 154V/I, L63L/P, V82/A viral strains isolated from patient-131 pretreated with ritonavir(20 fold) at 28th week1998Bioorganic & medicinal chemistry letters, Dec-15, Volume: 8, Issue:24
Potent piperazine hydroxyethylamine HIV protease inhibitors containing novel P3 ligands.
AID391268Antiviral activity against multidrug-resistant HIV1 with protease L10I, K14R, L33I, M36I, M46I, F53I, K55R, I62V, L63P, A71V, G73S, V82A, L90M, I93L mutation in phytohemagglutininin-activated PBMC assessed as inhibition of p24G protein production2008Journal of medicinal chemistry, Oct-09, Volume: 51, Issue:19
Flexible cyclic ethers/polyethers as novel P2-ligands for HIV-1 protease inhibitors: design, synthesis, biological evaluation, and protein-ligand X-ray studies.
AID374644Tmax in human at 100 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 10 followed by 100 mg, po of drug coadministered with voriconazole 200 mg, p2007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID105581In vitro anti-HIV activity in MT-4 cells by using an HIV cytopathic assay1998Journal of medicinal chemistry, Sep-24, Volume: 41, Issue:20
Design and synthesis of new potent C2-symmetric HIV-1 protease inhibitors. Use of L-mannaric acid as a peptidomimetic scaffold.
AID353749Cmax in po dosed Beagle dog at dose molar equivalent to 5 mg/kg RTV coadministered with LPV-OMP dosed as 5% dextrose containing solution2009Journal of medicinal chemistry, May-14, Volume: 52, Issue:9
Water-soluble prodrugs of the human immunodeficiency virus protease inhibitors lopinavir and ritonavir.
AID721743Ratio of Cmax unbound to IC50 for human MATE1-mediated [14]-metformin uptake expressed in HEK293 cells2013Journal of medicinal chemistry, Feb-14, Volume: 56, Issue:3
Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling.
AID278956Antiviral activity against HIV1 A4 isolate with protease I84A mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID1604603Antiviral activity against Human immunodeficiency virus 12020Journal of medicinal chemistry, 03-26, Volume: 63, Issue:6
Urea Derivatives in Modern Drug Discovery and Medicinal Chemistry.
AID572570Half life in HIV-1 infected patient at 100 mg, po bid coadministered with 400 mg, po bid of Lopinavir and 300 mg, po qd of atazanavir2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Decrease of atazanavir and lopinavir plasma concentrations in a boosted double human immunodeficiency virus protease inhibitor salvage regimen.
AID738130Binding affinity to CYP3A4 (3-22)-deleted S119A mutant (unknown origin) expressed in Escherichia coli by stopped flow assay2013Journal of medicinal chemistry, May-09, Volume: 56, Issue:9
Pyridine-substituted desoxyritonavir is a more potent inhibitor of cytochrome P450 3A4 than ritonavir.
AID322100Selectivity index, ratio of CC50 for MT2 cells to EC50 for HIV1 LAI2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID297669Inhibition of HIV1 Protease M2 variant by FRET based assay2007Journal of medicinal chemistry, Sep-06, Volume: 50, Issue:18
Design and synthesis of HIV-1 protease inhibitors incorporating oxazolidinones as P2/P2' ligands in pseudosymmetric dipeptide isosteres.
AID557295Ratio of EC50 for HIV1 NL4-3 harboring L10F/L24I/M46I/L63P/A71V/G73S/V82T amino acid substitution in protease encoding region to EC50 for HIV1 NL4-32009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID1220559Fraction unbound in cynomolgus monkey brain homogenates at 1 uM after 6 hrs by equilibrium dialysis method2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Species independence in brain tissue binding using brain homogenates.
AID391272Antiviral activity against HIV1 ERS104 with protease L63P mutation in phytohemagglutininin-activated PBMC assessed as inhibition of p24G protein production2008Journal of medicinal chemistry, Oct-09, Volume: 51, Issue:19
Flexible cyclic ethers/polyethers as novel P2-ligands for HIV-1 protease inhibitors: design, synthesis, biological evaluation, and protein-ligand X-ray studies.
AID1578463Drug concentration in total white adipose tissue in Wistar Han rat at 1 to 2 mg/kg, iv infused for 10 to 20 hrs by LC-MS/MS analysis2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID369174Tlag in HIV1 infected human at 100 mg, po administered twice daily2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
Association of saquinavir plasma concentrations with side effects but not with antiretroviral outcome in patients infected with protease inhibitor-susceptible human immunodeficiency virus type 1.
AID557277Ratio of EC50 for HIV1 TM harboring L10I/K14R/R41K/M46L/I54V/L63P/A71V/V82A/L90M/I93L in protease encoding region to EC50 for HIV1 ERS104pre2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID553568Selectivity index, ratio of CC50 for human MT2 cells to EC50 for HIV1 LAI2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID322107Antiviral activity against lopinavir-resistant HIV1 in MT4 cells assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID225564Dissociation constant obtained by inhibition of mutant HIV-protease (V-18)2000Journal of medicinal chemistry, Sep-07, Volume: 43, Issue:18
Identification of MK-944a: a second clinical candidate from the hydroxylaminepentanamide isostere series of HIV protease inhibitors.
AID408206Inhibition of HIV1 protease at 1 nM2008Journal of medicinal chemistry, May-22, Volume: 51, Issue:10
Combination of non-natural D-amino acid derivatives and allophenylnorstatine-dimethylthioproline scaffold in HIV protease inhibitors have high efficacy in mutant HIV.
AID19468Partition coefficient (logP)2003Journal of medicinal chemistry, Apr-24, Volume: 46, Issue:9
Comparison of in vitro P-glycoprotein screening assays: recommendations for their use in drug discovery.
AID557243Toxicity in HIV-infected Thai pregnant women assessed as adverse effect at 100 mg, po bid initiated intrapartum administered for 30 days in combination with 400 mg, po bid Lopinavir2009Antimicrobial agents and chemotherapy, May, Volume: 53, Issue:5
Early postpartum pharmacokinetics of lopinavir initiated intrapartum in Thai women.
AID557284Antiviral activity against HIV1 NL4-3 infected in human MT4 cells assessed as inhibition of p24 gag protein production by ELISA2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID322121Antiviral activity against HIV1 BaL R5 subtype B in phytohemagglutininin-activated PBMCs assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID242868Association rate constant for the interaction between the compound and serum albumin2005Journal of medicinal chemistry, May-19, Volume: 48, Issue:10
Early absorption and distribution analysis of antitumor and anti-AIDS drugs: lipid membrane and plasma protein interactions.
AID1079931Moderate liver toxicity, defined via clinical-chemistry results: ALT or AST serum activity 6 times the normal upper limit (N) or alkaline phosphatase serum activity of 1.7 N. Value is number of references indexed. [column 'BIOL' in source]
AID427583Inhibition of SAP5-dependent growth in Candida albicans SAP2MS4B expressing SAP5ex4A at 100 uM incubated in YCB-BSA medium2008Antimicrobial agents and chemotherapy, Jan, Volume: 52, Issue:1
Tetracycline-inducible expression of individual secreted aspartic proteases in Candida albicans allows isoenzyme-specific inhibitor screening.
AID557226Drug level in HIV-infected Thai pregnant women serum at 100 mg, po bid initiated intrapartum in combination with 400 mg, po bid Lopinavir measured 2 hrs post last dose2009Antimicrobial agents and chemotherapy, May, Volume: 53, Issue:5
Early postpartum pharmacokinetics of lopinavir initiated intrapartum in Thai women.
AID391269Antiviral activity against multidrug-resistant HIV1 with protease L10I, V11I, T12E, I15V, L19I, R41K, M46L, L63P, A71T, V82A, L90M mutation in phytohemagglutininin-activated PBMC assessed as inhibition of p24G protein production2008Journal of medicinal chemistry, Oct-09, Volume: 51, Issue:19
Flexible cyclic ethers/polyethers as novel P2-ligands for HIV-1 protease inhibitors: design, synthesis, biological evaluation, and protein-ligand X-ray studies.
AID1220560Fraction unbound in human occipital cortex at 1 uM after 6 hrs by equilibrium dialysis method2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Species independence in brain tissue binding using brain homogenates.
AID408217Ratio of EC50 for HIV1 3B in presence of 10% fetal calf serum and alpha1 acid glycoprotein to EC50 for HIV1 3B in presence of 10% fetal calf serum2008Journal of medicinal chemistry, May-22, Volume: 51, Issue:10
Combination of non-natural D-amino acid derivatives and allophenylnorstatine-dimethylthioproline scaffold in HIV protease inhibitors have high efficacy in mutant HIV.
AID278969Antiviral activity against HIV1 C2 isolate with protease I84C mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID558398Ratio of drug level in HIV-infected pregnant woman amniotic fluid to maternal blood plasma at 100 mg, po BID by HPLC/UV analysis2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Genital tract, cord blood, and amniotic fluid exposures of seven antiretroviral drugs during and after pregnancy in human immunodeficiency virus type 1-infected women.
AID1474166Liver toxicity in human assessed as induction of drug-induced liver injury by measuring severity class index2016Drug discovery today, Apr, Volume: 21, Issue:4
DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans.
AID373867Hepatic clearance in human hepatocytes in absence of fetal calf serum2009European journal of medicinal chemistry, Apr, Volume: 44, Issue:4
First-principle, structure-based prediction of hepatic metabolic clearance values in human.
AID374640Tmax in human at 400 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 10 followed by 400 mg, po of drug coadministered with voriconazole 200 mg, p2007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID589196Mechanism based inhibition of human cytochrome P450 3A4, partition ratio2005Current drug metabolism, Oct, Volume: 6, Issue:5
Cytochrome p450 enzymes mechanism based inhibitors: common sub-structures and reactivity.
AID444057Fraction escaping hepatic elimination in human2010Journal of medicinal chemistry, Feb-11, Volume: 53, Issue:3
Physicochemical space for optimum oral bioavailability: contribution of human intestinal absorption and first-pass elimination.
AID572583Cmin in HIV-1 infected patient at 100 mg, po bid coadministered with 400 mg, po bid of Lopinavir and 300 mg, po qd of atazanavir2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Decrease of atazanavir and lopinavir plasma concentrations in a boosted double human immunodeficiency virus protease inhibitor salvage regimen.
AID1578462Total plasma concentration in Wistar Han rat at 1 to 2 mg/kg, iv infused for 10 to 20 hrs by LC-MS/MS analysis2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID353732Cmax in Sprague-Dawley rat plasma at 5 mg/kg, po2009Journal of medicinal chemistry, May-14, Volume: 52, Issue:9
Water-soluble prodrugs of the human immunodeficiency virus protease inhibitors lopinavir and ritonavir.
AID239809Equilibrium dissociation constant for the interaction between the compound and serum albumin2005Journal of medicinal chemistry, May-19, Volume: 48, Issue:10
Early absorption and distribution analysis of antitumor and anti-AIDS drugs: lipid membrane and plasma protein interactions.
AID525112Antiviral activity against HIV 1 NL4.3 harboring integrase L74M mutant infected in human MT-4 cells assessed as inhibition of virus-induced cytopathic effect by MTT assay2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Mutations in human immunodeficiency virus type 1 integrase confer resistance to the naphthyridine L-870,810 and cross-resistance to the clinical trial drug GS-9137.
AID321685Antiviral activity against wild type HIV12008Bioorganic & medicinal chemistry, Feb-01, Volume: 16, Issue:3
Structure-activity relationships of novel HIV-1 protease inhibitors containing the 3-amino-2-chlorobenzoyl-allophenylnorstatine structure.
AID557281Ratio of EC50 for HIV1 B harboring L10I/I15V/E35D/N37E/K45R/I54V/L63P/A71V/V82T/L90M/I93L/C95F in protease encoding region to EC50 for HIV1 ERS104pre2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID278977Antiviral activity against HIV1 C10 isolate with protease I84C mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID525169Antimicrobial activity against chloroquine-resistant Plasmodium chabaudi ASCQ infected in NIH mice (Mus musculus) at 10 to 160 mg/kg, perorally administered after 72 hrs post inoculation measured on day 4 post infection2008Antimicrobial agents and chemotherapy, Jul, Volume: 52, Issue:7
Synergy of human immunodeficiency virus protease inhibitors with chloroquine against Plasmodium falciparum in vitro and Plasmodium chabaudi in vivo.
AID353763AUC in Sprague-Dawley rat plasma at 5 mg/kg, po2009Journal of medicinal chemistry, May-14, Volume: 52, Issue:9
Water-soluble prodrugs of the human immunodeficiency virus protease inhibitors lopinavir and ritonavir.
AID416859Increase in P-glycoprotein-mediated tenofovir disoproxil fumarate permeation from apical to basolateral side of human Caco-2 cells at 20 uM2007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Effects of human immunodeficiency virus protease inhibitors on the intestinal absorption of tenofovir disoproxil fumarate in vitro.
AID625280Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for cholecystitis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID557297Ratio of EC50 for HIV1 NL4-3 harboring M46I/V82F/I84V amino acid substitution in protease encoding region to EC50 for HIV1 NL4-32009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID519580Antiviral activity against HIV1 clone5 infected in HEK293 cells harboring A-790742-selected protease L63P, A71V, and V82G mutation assessed as reduction in viral replication by luciferase reporter gene assay relative to wild type HIV1 RF2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
Characterization of a novel human immunodeficiency virus type 1 protease inhibitor, A-790742.
AID374593Cytotoxicity against human MT4 cells after 6 days by MTT assay2007Antimicrobial agents and chemotherapy, Nov, Volume: 51, Issue:11
In vitro antiviral activity and cross-resistance profile of PL-100, a novel protease inhibitor of human immunodeficiency virus type 1.
AID681154TP_TRANSPORTER: cell accumulation in KB-3-1 and KB-V1 cells2001The Journal of pharmacology and experimental therapeutics, Mar, Volume: 296, Issue:3
In vitro substrate identification studies for p-glycoprotein-mediated transport: species difference and predictability of in vivo results.
AID88997Antiviral activity was assessed against M36M/I, V82F viral strains isolated from patient-313 pretreated with ritonavir(9 fold) at 57th day1998Bioorganic & medicinal chemistry letters, Dec-15, Volume: 8, Issue:24
Potent piperazine hydroxyethylamine HIV protease inhibitors containing novel P3 ligands.
AID391265Antiviral activity against multidrug-resistant HIV1 with protease L10I, K14R, R41K, M46L, I54V, L63P, A71V, V82A, L90M, I93L mutation in phytohemagglutininin-activated PBMC assessed as inhibition of p24Gag protein production2008Journal of medicinal chemistry, Oct-09, Volume: 51, Issue:19
Flexible cyclic ethers/polyethers as novel P2-ligands for HIV-1 protease inhibitors: design, synthesis, biological evaluation, and protein-ligand X-ray studies.
AID519572Antiviral activity against HIV1 P9 infected in human MT4 cells derived from viral passages with A-790742 harboring protease V82V/L and I84V mutation assessed as reduction in viral cytopathogenicity treated 1 hr post infection measured 5 days post infectio2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
Characterization of a novel human immunodeficiency virus type 1 protease inhibitor, A-790742.
AID105722Compound was evaluated for its antiviral inhibition in MT-4 cell culture2000Bioorganic & medicinal chemistry letters, Jun-05, Volume: 10, Issue:11
Novel inhibitors of HIV protease: design, synthesis and biological evaluation of picomolar inhibitors containing cyclic P1/P2 scaffolds.
AID105206Inhibitory concentration against accumulation of viral p24 antigen following infection of MT-4 cells1997Journal of medicinal chemistry, Jan-17, Volume: 40, Issue:2
Cyclic urea amides: HIV-1 protease inhibitors with low nanomolar potency against both wild type and protease inhibitor resistant mutants of HIV.
AID1217728Intrinsic clearance for reactive metabolites formation per mg of protein based on cytochrome P450 (unknown origin) inactivation rate by TDI assay2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Combination of GSH trapping and time-dependent inhibition assays as a predictive method of drugs generating highly reactive metabolites.
AID369171Cmin in HIV1 infected human at 100 mg, po administered twice daily2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
Association of saquinavir plasma concentrations with side effects but not with antiretroviral outcome in patients infected with protease inhibitor-susceptible human immunodeficiency virus type 1.
AID1079943Malignant tumor, proven histopathologically. Value is number of references indexed. [column 'T.MAL' in source]
AID372198AUC (0 to 12 hrs) in human at 400 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 102007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID150754Inhibition of P-glycoprotein, mouse L-mdr1b expressed in LLC-PK1 epithelial cells using calcein-AM polarisation assay2003Journal of medicinal chemistry, Apr-24, Volume: 46, Issue:9
Comparison of in vitro P-glycoprotein screening assays: recommendations for their use in drug discovery.
AID427581Inhibition of SAP8-dependent growth in Candida albicans SAP2MS4B expressing SAP8ex4A at 100 uM incubated in YCB-BSA medium2008Antimicrobial agents and chemotherapy, Jan, Volume: 52, Issue:1
Tetracycline-inducible expression of individual secreted aspartic proteases in Candida albicans allows isoenzyme-specific inhibitor screening.
AID1222388Inhibition of human recombinant UGT1A1 expressed in HEK293 cells assessed as reduction in bilirubin glucuronidation by LC-MS/MS method2011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Correlation between bilirubin glucuronidation and estradiol-3-gluronidation in the presence of model UDP-glucuronosyltransferase 1A1 substrates/inhibitors.
AID322101Antiviral activity against HIV1 NL4-3 in MT4 cells by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID1578460Fraction unbound in Wistar Han rat liver at 2 uM incubated for 6 hrs by equilibrium dialysis method2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID369941Antiviral activity against HIV2 MS infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect after 5 days by MTT assay2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro selection and characterization of human immunodeficiency virus type 2 with decreased susceptibility to lopinavir.
AID322104Antiviral activity against idinavir-resistant HIV1 in MT4 cells assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID553577Antiviral activity against HIV1 MM harboring L10I/K43T/M46L/I54V/L63P/A71V/V82A/L90M/Q92K in protease encoding region infected in human PHA-PBC assessed as inhibition of p24 Gag protein production by ELISA2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID573997Ratio of EC50 for Human immunodeficiency virus 1 3B to EC50 for MK-0518-resistant Human immunodeficiency virus harboring G140S and Q148H mutations in integrase2008Antimicrobial agents and chemotherapy, Aug, Volume: 52, Issue:8
Preclinical evaluation of 1H-benzylindole derivatives as novel human immunodeficiency virus integrase strand transfer inhibitors.
AID557282Ratio of EC50 for HIV1 C harboring L10I/I15V/K20R/L24I/M36I/M46L/I54V/I62V/L63P/K70Q/V82A/L89M in protease encoding region to EC50 for HIV1 ERS104pre2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID322099Cytotoxicity against human MT2 cells assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID679892TP_TRANSPORTER: inhibition of cyclosporin A uptake (cyclosporin A: 10 uM) in MDR1-expressing LLC-PK1 cells2003The Journal of pharmacology and experimental therapeutics, Mar, Volume: 304, Issue:3
Pharmacokinetics and interactions of a novel antagonist of chemokine receptor 5 (CCR5) with ritonavir in rats and monkeys: role of CYP3A and P-glycoprotein.
AID572591Half life in HIV-1 infected patient at 100 mg, po bid coadministered with 400 mg, po bid of Lopinavir and NRTI2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Decrease of atazanavir and lopinavir plasma concentrations in a boosted double human immunodeficiency virus protease inhibitor salvage regimen.
AID373234AUC (0 to 12 hrs) in human at 100 mg, po bid for 10 days pretreated with paroxetin at 20 mg, po qd for 10 days followed by 16 days washout period then coadministered with paroxetin at 20 mg, po qd and fosamprenavir at 700 mg, po bid for 10 days2007Antimicrobial agents and chemotherapy, Nov, Volume: 51, Issue:11
Interaction study of the combined use of paroxetine and fosamprenavir-ritonavir in healthy subjects.
AID322115Antiviral activity against HIV1 MDR/JSL R5 in phytohemagglutininin-activated PBMCs assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID1079947Comments (NB not yet translated). [column 'COMMENTAIRES' in source]
AID369942Antiviral activity against HIV2 CBL-23 infected in human PBMC assessed as inhibition of virus production after 5 days by Lenti-RT activity assay2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro selection and characterization of human immunodeficiency virus type 2 with decreased susceptibility to lopinavir.
AID444055Fraction absorbed in human2010Journal of medicinal chemistry, Feb-11, Volume: 53, Issue:3
Physicochemical space for optimum oral bioavailability: contribution of human intestinal absorption and first-pass elimination.
AID557299Ratio of EC50 for HIV1 NL4-3 harboring L23I/K43I/M46I/I50L/G51A/A71V amino acid substitution in protease encoding region to EC50 for HIV1 NL4-32009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID198434Inhibitory activity against Ritonavir resistant virus mutant strain1999Bioorganic & medicinal chemistry letters, Nov-15, Volume: 9, Issue:22
Unsymmetrical cyclic ureas as HIV-1 protease inhibitors: novel biaryl indazoles as P2/P2' substituents.
AID557274Antiviral activity against HIV1 B harboring L10I/I15V/E35D/N37E/K45R/I54V/L63P/A71V/V82T/L90M/I93L/C95F in protease encoding region infected in human PHA-PBC assessed as inhibition of p24 Gag protein production by ELISA2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID525102Antiviral activity against HIV 1 3B infected in MT-4 cells assessed as inhibition of virus-induced cytopathic effect by MTT assay2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Mutations in human immunodeficiency virus type 1 integrase confer resistance to the naphthyridine L-870,810 and cross-resistance to the clinical trial drug GS-9137.
AID525167Antimicrobial activity against chloroquine-resistant Plasmodium falciparum 3D7 infected in human erythrocytes assessed as potentiation of choloroquine-mediated antimalarial activity by light microscopy2008Antimicrobial agents and chemotherapy, Jul, Volume: 52, Issue:7
Synergy of human immunodeficiency virus protease inhibitors with chloroquine against Plasmodium falciparum in vitro and Plasmodium chabaudi in vivo.
AID444054Oral bioavailability in human2010Journal of medicinal chemistry, Feb-11, Volume: 53, Issue:3
Physicochemical space for optimum oral bioavailability: contribution of human intestinal absorption and first-pass elimination.
AID322118Antiviral activity against HIV1 MDR/G X4 in phytohemagglutininin-activated PBMCs assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID557242Ratio of drug level in 4 hrs to 30 days post last dose of HIV-infected Thai pregnant women serum at 100 mg, po bid initiated intrapartum in combination with 400 mg, po bid Lopinavir2009Antimicrobial agents and chemotherapy, May, Volume: 53, Issue:5
Early postpartum pharmacokinetics of lopinavir initiated intrapartum in Thai women.
AID588218FDA HLAED, lactate dehydrogenase (LDH) increase2004Current drug discovery technologies, Dec, Volume: 1, Issue:4
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
AID1069013Inhibition of human CYP3A4-mediated midazolam 1'-hydroxylase activity2014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Structure-activity relationships of diamine inhibitors of cytochrome P450 (CYP) 3A as novel pharmacoenhancers, part I: core region.
AID1222389Inhibition of human recombinant UGT1A1 expressed in HEK293 cells assessed as reduction in estradiol 3-glucuronidation by LC-MS/MS method2011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Correlation between bilirubin glucuronidation and estradiol-3-gluronidation in the presence of model UDP-glucuronosyltransferase 1A1 substrates/inhibitors.
AID679278TP_TRANSPORTER: transepithelial transport (apical to basal) in MDR1-expressing LLC-PK1 cells2001Pharmaceutical research, Dec, Volume: 18, Issue:12
Comparative studies on in vitro methods for evaluating in vivo function of MDR1 P-glycoprotein.
AID160455Inhibition constant against HIV-1 Protease2002Journal of medicinal chemistry, Dec-05, Volume: 45, Issue:25
Relationships between structure and interaction kinetics for HIV-1 protease inhibitors.
AID269316Antiviral activity against multi drug-resistant HIV1 ES variant in human PHA-PBMC cells2006Journal of medicinal chemistry, Aug-24, Volume: 49, Issue:17
Structure-based design of novel HIV-1 protease inhibitors to combat drug resistance.
AID679998TP_TRANSPORTER: transepithelial transport (basal to apical) in mdr1a-expressing LLC-PK1 cell2001Molecular pharmacology, Apr, Volume: 59, Issue:4
P-glycoprotein limits oral availability, brain, and fetal penetration of saquinavir even with high doses of ritonavir.
AID1267620Selectivity index, ratio of CC50 for human HUT78 cells to EC50 for HIV1 3B2015Journal of medicinal chemistry, Dec-24, Volume: 58, Issue:24
Design and Synthesis of DiselenoBisBenzamides (DISeBAs) as Nucleocapsid Protein 7 (NCp7) Inhibitors with anti-HIV Activity.
AID162679Inhibition of HIV protease at 0.5 nM1998Bioorganic & medicinal chemistry letters, Dec-15, Volume: 8, Issue:24
Potent piperazine hydroxyethylamine HIV protease inhibitors containing novel P3 ligands.
AID374657Toxicity in human assessed as serious adverse events at 400 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 102007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID243931Percent inhibition of synthesized human T-cell leukemia virus type I (HTLV-1) protease was determined at 100 uM concentration2004Bioorganic & medicinal chemistry letters, Dec-06, Volume: 14, Issue:23
Identification of peptidomimetic HTLV-I protease inhibitors containing hydroxymethylcarbonyl (HMC) isostere as the transition-state mimic.
AID1443989Inhibition of recombinant human BSEP expressed in baculovirus infected sf9 cell plasma membrane vesicles assessed as reduction in ATP-dependent [3H]-taurocholate uptake in to vesicles preincubated for 10 mins followed by ATP addition measured after 10 to 2014Hepatology (Baltimore, Md.), Sep, Volume: 60, Issue:3
Human drug-induced liver injury severity is highly associated with dual inhibition of liver mitochondrial function and bile salt export pump.
AID525279Antimicrobial activity against chloroquine-sensitive Plasmodium falciparum 3D7 after 48 hrs2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
Antimalarial asexual stage-specific and gametocytocidal activities of HIV protease inhibitors.
AID520314Inhibition of human CYP2B6 in human liver microsomes2008Antimicrobial agents and chemotherapy, May, Volume: 52, Issue:5
Rapid clinical induction of hepatic cytochrome P4502B6 activity by ritonavir.
AID699542Inhibition of human liver OATP1B1 expressed in HEK293 Flp-In cells assessed as reduction in E17-betaG uptake by scintillation counting2012Journal of medicinal chemistry, May-24, Volume: 55, Issue:10
Classification of inhibitors of hepatic organic anion transporting polypeptides (OATPs): influence of protein expression on drug-drug interactions.
AID1217710Covalent binding in human liver microsomes measured per mg of protein using radiolabelled compound at 10 uM after 1 hr incubation by liquid scintillation counting2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Combination of GSH trapping and time-dependent inhibition assays as a predictive method of drugs generating highly reactive metabolites.
AID372204Cmin in human at 400 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 102007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID680158TP_TRANSPORTER: transepithelial transport (basal to apical) in mdr1b-expressing LLC-PK1 cell2001Molecular pharmacology, Apr, Volume: 59, Issue:4
P-glycoprotein limits oral availability, brain, and fetal penetration of saquinavir even with high doses of ritonavir.
AID738132Binding affinity to CYP3A4 (3-22)-deleted S119A mutant (unknown origin) expressed in Escherichia coli assessed as increase in melting temperature by spectrophotometric analysis (Rvb = 53.5 +/- 0.1 degC)2013Journal of medicinal chemistry, May-09, Volume: 56, Issue:9
Pyridine-substituted desoxyritonavir is a more potent inhibitor of cytochrome P450 3A4 than ritonavir.
AID625291Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for liver function tests abnormal2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID519573Antiviral activity against HIV1 P15 infected in human MT4 cells derived from viral passages with A-790742 harboring protease L33L/F, K45I, V82L, and I84V mutation assessed as reduction in viral cytopathogenicity treated 1 hr post infection measured 5 days2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
Characterization of a novel human immunodeficiency virus type 1 protease inhibitor, A-790742.
AID444053Renal clearance in human2010Journal of medicinal chemistry, Feb-11, Volume: 53, Issue:3
Physicochemical space for optimum oral bioavailability: contribution of human intestinal absorption and first-pass elimination.
AID160300Inhibition of HIV protease2000Bioorganic & medicinal chemistry letters, Jun-05, Volume: 10, Issue:11
Novel inhibitors of HIV protease: design, synthesis and biological evaluation of picomolar inhibitors containing cyclic P1/P2 scaffolds.
AID235096Resistance of constructed mutant 82F virus was calculated as (IC90 for a mutant virus / IC90 of wt HXB2 virus). 1997Journal of medicinal chemistry, Jan-17, Volume: 40, Issue:2
Cyclic urea amides: HIV-1 protease inhibitors with low nanomolar potency against both wild type and protease inhibitor resistant mutants of HIV.
AID525172Antimicrobial activity against chloroquine-sensitive Plasmodium chabaudi ASS infected in NIH mice (Mus musculus) assessed as potentiation of 2.5 mg/kg chloroquine-mediated antimalarial activity at 100 mg/kg, perorally administered after 72 hrs post inocul2008Antimicrobial agents and chemotherapy, Jul, Volume: 52, Issue:7
Synergy of human immunodeficiency virus protease inhibitors with chloroquine against Plasmodium falciparum in vitro and Plasmodium chabaudi in vivo.
AID353747Cmax in po dosed Beagle dog at dose molar equivalent to 5 mg/kg LPV, po dosed as 5% dextrose containing solution2009Journal of medicinal chemistry, May-14, Volume: 52, Issue:9
Water-soluble prodrugs of the human immunodeficiency virus protease inhibitors lopinavir and ritonavir.
AID1267613Antiviral activity against HIV1 3B infected in human MT4 cells assessed as inhibition of viral replication by measuring p24 Ag production at 2 ug/ml measured at 31 hrs post infection by ELISA2015Journal of medicinal chemistry, Dec-24, Volume: 58, Issue:24
Design and Synthesis of DiselenoBisBenzamides (DISeBAs) as Nucleocapsid Protein 7 (NCp7) Inhibitors with anti-HIV Activity.
AID374591Inhibition of HIV1 protease2007Antimicrobial agents and chemotherapy, Nov, Volume: 51, Issue:11
In vitro antiviral activity and cross-resistance profile of PL-100, a novel protease inhibitor of human immunodeficiency virus type 1.
AID278970Antiviral activity against HIV1 C3 isolate with protease I84C mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID540231Dose normalised AUC in dog after po administration2005Xenobiotica; the fate of foreign compounds in biological systems, Feb, Volume: 35, Issue:2
Comparative evaluation of oral systemic exposure of 56 xenobiotics in rat, dog, monkey and human.
AID572584Cmax in HIV-1 infected patient at 100 mg, po bid coadministered with 300 mg, po qd of atazanavir and NRTI2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Decrease of atazanavir and lopinavir plasma concentrations in a boosted double human immunodeficiency virus protease inhibitor salvage regimen.
AID322098Antiviral activity against HIV2 ROD in MT2 cells assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID321692Antiviral activity against HIV1 mutant strain 72008Bioorganic & medicinal chemistry, Feb-01, Volume: 16, Issue:3
Structure-activity relationships of novel HIV-1 protease inhibitors containing the 3-amino-2-chlorobenzoyl-allophenylnorstatine structure.
AID278982Resistance to HIV1 with protease 46I, 54I and I84A mutation in HEK 293 cells relative to similar background2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID699539Inhibition of human liver OATP1B1 expressed in HEK293 Flp-In cells assessed as reduction in E17-betaG uptake at 20 uM by scintillation counting2012Journal of medicinal chemistry, May-24, Volume: 55, Issue:10
Classification of inhibitors of hepatic organic anion transporting polypeptides (OATPs): influence of protein expression on drug-drug interactions.
AID519579Antiviral activity against HIV1 clone4 infected in HEK293 cells harboring A-790742-selected protease L33F, A71V, G73S, V77I, V82L, and I84V mutation assessed as reduction in viral replication by luciferase reporter gene assay relative to wild type HIV1 pN2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
Characterization of a novel human immunodeficiency virus type 1 protease inhibitor, A-790742.
AID278967Antiviral activity against HIV1 A15 isolate with protease I84A mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID671806Antiviral activity against wild type HIV1 3B infected in human MT4 LTR-EGFP cells by EGFP reporter gene assay2012Bioorganic & medicinal chemistry letters, Aug-01, Volume: 22, Issue:15
Benzoxazole and benzothiazole amides as novel pharmacokinetic enhancers of HIV protease inhibitors.
AID105397Anti-HIV activity was determined in MT-4 cells in the presence of 50% human serum using cytopathic effect assay1998Bioorganic & medicinal chemistry letters, Dec-15, Volume: 8, Issue:24
Potent piperazine hydroxyethylamine HIV protease inhibitors containing novel P3 ligands.
AID557288Antiviral activity against HIV1 NL4-3 harboring L10F/D30N/K45I/A71V/T74S amino acid substitution in protease encoding region infected in human MT4 cells assessed as inhibition of p24 Gag protein production selected at 5 uM of nelfinavir by ELISA2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID529759Antimicrobial activity against Plasmodium falciparum at ring stage2008Antimicrobial agents and chemotherapy, Jul, Volume: 52, Issue:7
Stronger activity of human immunodeficiency virus type 1 protease inhibitors against clinical isolates of Plasmodium vivax than against those of P. falciparum.
AID369946Ratio of EC50 for HIV2 CDC310319 infected in human PBMC to EC50 for HIV1 NL4-3 infected in human MT4 cells2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro selection and characterization of human immunodeficiency virus type 2 with decreased susceptibility to lopinavir.
AID278974Antiviral activity against HIV1 C7 isolate with protease I84C mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID738136Binding affinity to C-terminal His-tagged wild type CYP3A4 (unknown origin) expressed in Escherichia coli assessed as spectral changes by spectrophotometric analysis relative to control2013Journal of medicinal chemistry, May-09, Volume: 56, Issue:9
Pyridine-substituted desoxyritonavir is a more potent inhibitor of cytochrome P450 3A4 than ritonavir.
AID82748Fold increase in EC50 for antiviral activity against HIV Protease-Resistant strains with C protease amino acid substitutions2001Journal of medicinal chemistry, Jul-05, Volume: 44, Issue:14
4-Hydroxy-5,6-dihydropyrones as inhibitors of HIV protease: the effect of heterocyclic substituents at C-6 on antiviral potency and pharmacokinetic parameters.
AID372206AUC (0 to 12 hrs) in human at 400 mg, po bid from day 11 to day 20 pretreated with placebo from day 1 to day 3 followed by washout from day 4 to day 102007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID372396Cmin in human at 400 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 10 followed by 400 mg, po of drug coadministered with voriconazole 200 mg, p2007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID353755AUC in po dosed Beagle dog at dose molar equivalent to 5 mg/kg RTV coadministered with LPV dosed as 5% dextrose containing solution2009Journal of medicinal chemistry, May-14, Volume: 52, Issue:9
Water-soluble prodrugs of the human immunodeficiency virus protease inhibitors lopinavir and ritonavir.
AID557293Ratio of EC50 for HIV1 NL4-3 harboring L10I/G48V/I54V/L90M amino acid substitution in protease encoding region to EC50 for HIV1 NL4-32009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID1578467Drug concentration in total skeletal muscle in Wistar Han rat at 1 to 2 mg/kg, iv infused for 10 to 20 hrs by LC-MS/MS analysis2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID543384Cmax in HIV-infected patient at 100 mg, qd co-administered with 100 mg minocycline and 250 mg valporic acid, bid for 15 days2008Antimicrobial agents and chemotherapy, Sep, Volume: 52, Issue:9
Effects of minocycline and valproic acid coadministration on atazanavir plasma concentrations in human immunodeficiency virus-infected adults receiving atazanavir-ritonavir.
AID374649Decrease in steady state Cmax of voriconazole in human at 400 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 10 followed by 400 mg, po of drug c2007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID443165Inhibition of HIV1 protease expressed in Escherichia coli by fluorometric assay2010Journal of medicinal chemistry, Jan-28, Volume: 53, Issue:2
HIV-1 protease inhibitors with a transition-state mimic comprising a tertiary alcohol: improved antiviral activity in cells.
AID227925The compound was estimated for biosensor analysis2000Journal of medicinal chemistry, May-18, Volume: 43, Issue:10
Biosensor analysis of the interaction between immobilized human serum albumin and drug compounds for prediction of human serum albumin binding levels.
AID427582Inhibition of SAP6-dependent growth in Candida albicans SAP2MS4B expressing SAP6ex4A at 100 uM incubated in YCB-BSA medium2008Antimicrobial agents and chemotherapy, Jan, Volume: 52, Issue:1
Tetracycline-inducible expression of individual secreted aspartic proteases in Candida albicans allows isoenzyme-specific inhibitor screening.
AID625286Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatitis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID28091Cmax value in the period of 0-24 hr after dosing. 2000Journal of medicinal chemistry, Feb-10, Volume: 43, Issue:3
Protease inhibitors: current status and future prospects.
AID1221964Transporter substrate index ratio of permeability from basolateral to apical side in human Caco2 cells at 10 uM up to 120 mins by HPLC-MC analysis in presence of 1 uM of P-gp inhibitor LY3359792011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Attenuation of intestinal absorption by major efflux transporters: quantitative tools and strategies using a Caco-2 model.
AID322112Antiviral activity against wild type HIV1 ERS104prc X4 in phytohemagglutininin-activated PBMCs assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID369954Antiviral activity against HIV2 ROD with protease G17N/V47A mutation infected in human CEM cells assessed as inhibition of virus production after 7 days by Lenti-RT activity assay2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro selection and characterization of human immunodeficiency virus type 2 with decreased susceptibility to lopinavir.
AID572558Cmin in HIV-1 infected patient at 100 mg, po bid coadministered with 300 mg, po qd of atazanavir and NRTI2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Decrease of atazanavir and lopinavir plasma concentrations in a boosted double human immunodeficiency virus protease inhibitor salvage regimen.
AID391271Antiviral activity against multidrug-resistant HIV1/A with protease mutation in phytohemagglutininin-activated PBMC assessed as inhibition of p24G protein production2008Journal of medicinal chemistry, Oct-09, Volume: 51, Issue:19
Flexible cyclic ethers/polyethers as novel P2-ligands for HIV-1 protease inhibitors: design, synthesis, biological evaluation, and protein-ligand X-ray studies.
AID443166Antiviral activity against HIV1 infected in human MT4 cells assessed as inhibition of virus-induced cytopathogenicity after 6 days by XTT assay2010Journal of medicinal chemistry, Jan-28, Volume: 53, Issue:2
HIV-1 protease inhibitors with a transition-state mimic comprising a tertiary alcohol: improved antiviral activity in cells.
AID1079933Acute liver toxicity defined via clinical observations and clear clinical-chemistry results: serum ALT or AST activity > 6 N or serum alkaline phosphatases activity > 1.7 N. This category includes cytolytic, choleostatic and mixed liver toxicity. Value is
AID372210Cmax in human at 100 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 102007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID732420Efflux ratio of permeability from basolateral to apical side over apical to basolateral in pig LLC-PK1 cells overexpressing human MDR1 relative to efflux ratio in parental cell line2013ACS medicinal chemistry letters, Jan-10, Volume: 4, Issue:1
De novo prediction of p-glycoprotein-mediated efflux liability for druglike compounds.
AID369953Antiviral activity against HIV2 ROD with protease V47A mutation infected in human CEM cells assessed as inhibition of virus production after 7 days by Lenti-RT activity assay2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro selection and characterization of human immunodeficiency virus type 2 with decreased susceptibility to lopinavir.
AID374647Tmax in human at 400 mg, po bid from day 11 to day 20 pretreated with placebo from day 1 to day 3 followed by washout from day 4 to day 10 followed by 400 mg, po of drug coadministered with placebo from day 21 to day 302007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID625283Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for elevated liver function tests2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID322116Antiviral activity against HIV1 MDR/B X4 in phytohemagglutininin-activated PBMCs assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID540230Dose normalised AUC in rat after po administration2005Xenobiotica; the fate of foreign compounds in biological systems, Feb, Volume: 35, Issue:2
Comparative evaluation of oral systemic exposure of 56 xenobiotics in rat, dog, monkey and human.
AID104312Anti-HIV-1 activity against Wild type virus in MT-4 cells2001Journal of medicinal chemistry, Sep-13, Volume: 44, Issue:19
Design and synthesis of potent C(2)-symmetric diol-based HIV-1 protease inhibitors: effects of fluoro substitution.
AID322106Antiviral activity against atazanavir-resistant HIV1 in MT4 cells assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID374642Cmax in human at 100 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 10 followed by 100 mg, po of drug coadministered with voriconazole 200 mg, p2007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID553571Antiviral activity against HIV1 LAI infected in human MT2 cells after 7 days by MTT assay2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID1220556Fraction unbound in CD-1 mouse brain homogenates at 1 uM after 6 hrs by equilibrium dialysis method2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Species independence in brain tissue binding using brain homogenates.
AID1079942Steatosis, proven histopathologically. Value is number of references indexed. [column 'STEAT' in source]
AID1221963Transporter substrate index ratio of permeability from apical to basolateral side in human Caco2 cells at 10 uM up to 120 mins by HPLC-MC analysis in presence of 1 uM of P-gp inhibitor LY3359792011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Attenuation of intestinal absorption by major efflux transporters: quantitative tools and strategies using a Caco-2 model.
AID557294Ratio of EC50 for HIV1 NL4-3 harboring L10F/V32I/M46I/I54M//A71V/I84V amino acid substitution in protease encoding region to EC50 for HIV1 NL4-32009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID518053Toxicity in healthy human assessed as abdominal pain at 100 mg/kg, po administered as single dose2010Antimicrobial agents and chemotherapy, Jun, Volume: 54, Issue:6
Effect of vicriviroc on the QT/corrected QT interval and central nervous system in healthy subjects.
AID738316Inhibition of HIV1 protease2013Bioorganic & medicinal chemistry, Apr-01, Volume: 21, Issue:7
Synthesis and evaluation of coumarin derivatives as potential dual-action HIV-1 protease and reverse transcriptase inhibitors.
AID278963Antiviral activity against HIV1 A11 isolate with protease I84A mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID1267610Cytotoxicity against human MT4 cells assessed as cell viability after 5 days by MTT assay2015Journal of medicinal chemistry, Dec-24, Volume: 58, Issue:24
Design and Synthesis of DiselenoBisBenzamides (DISeBAs) as Nucleocapsid Protein 7 (NCp7) Inhibitors with anti-HIV Activity.
AID321694Ratio of EC50 for HIV1 mutant strain 2 to EC50 for wild type HIV12008Bioorganic & medicinal chemistry, Feb-01, Volume: 16, Issue:3
Structure-activity relationships of novel HIV-1 protease inhibitors containing the 3-amino-2-chlorobenzoyl-allophenylnorstatine structure.
AID543389Half life in HIV-infected patient at 100 mg, qd co-administered with 100 mg minocycline, bid for 15 days2008Antimicrobial agents and chemotherapy, Sep, Volume: 52, Issue:9
Effects of minocycline and valproic acid coadministration on atazanavir plasma concentrations in human immunodeficiency virus-infected adults receiving atazanavir-ritonavir.
AID1498797Cytotoxicity against human MT4 cells assessed as reduction in cell viability after 5 days by MTT assay2018Bioorganic & medicinal chemistry letters, 08-01, Volume: 28, Issue:14
Design and synthesis of selenazole-substituted ritonavir analogs.
AID572588Cmin in HIV-1 infected patient at 100 mg, po bid coadministered with 400 mg, po bid of Lopinavir and NRTI2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Decrease of atazanavir and lopinavir plasma concentrations in a boosted double human immunodeficiency virus protease inhibitor salvage regimen.
AID1474167Liver toxicity in human assessed as induction of drug-induced liver injury by measuring verified drug-induced liver injury concern status2016Drug discovery today, Apr, Volume: 21, Issue:4
DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans.
AID575060Antiviral activity against Human immunodeficiency virus 1 harboring protease inhibitor resistance-associated mutations and protease L76V mutation in viral protease assessed as fold change in drug susceptibility relative to wild type2010Antimicrobial agents and chemotherapy, Nov, Volume: 54, Issue:11
Prevalence, mutation patterns, and effects on protease inhibitor susceptibility of the L76V mutation in HIV-1 protease.
AID29722Tmax value of the compound2000Journal of medicinal chemistry, Feb-10, Volume: 43, Issue:3
Protease inhibitors: current status and future prospects.
AID219260Dissociation constant obtained by inhibition of Wild-type protease2000Journal of medicinal chemistry, Sep-07, Volume: 43, Issue:18
Identification of MK-944a: a second clinical candidate from the hydroxylaminepentanamide isostere series of HIV protease inhibitors.
AID88988Antiviral activity was assessed against K20K/R, 36M/I, 154V, A71A/V, V82T viral strains isolated from patient-129 pretreated with ritonavir(33 fold) at 16th week1998Bioorganic & medicinal chemistry letters, Dec-15, Volume: 8, Issue:24
Potent piperazine hydroxyethylamine HIV protease inhibitors containing novel P3 ligands.
AID373236Cmin in human at 100 mg, po bid for 10 days pretreated with paroxetin at 20 mg, po qd for 10 days followed by 16 days washout period then coadministered with paroxetin at 20 mg, po qd and fosamprenavir at 700 mg, po bid for 10 days2007Antimicrobial agents and chemotherapy, Nov, Volume: 51, Issue:11
Interaction study of the combined use of paroxetine and fosamprenavir-ritonavir in healthy subjects.
AID589111Mechanism based inhibition of human cytochrome P450 3A42005Current drug metabolism, Oct, Volume: 6, Issue:5
Cytochrome p450 enzymes mechanism based inhibitors: common sub-structures and reactivity.
AID322119Antiviral activity against HIV1 92UG029 X4 subtype A in phytohemagglutininin-activated PBMCs assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID257269Inhibitory activity against HIV1 protease at 0.5 uM2005Bioorganic & medicinal chemistry letters, Dec-15, Volume: 15, Issue:24
Synthesis and activity of N-acyl azacyclic urea HIV-1 protease inhibitors with high potency against multiple drug resistant viral strains.
AID408212Antiviral activity against ritonavir-resistant HIV1 NL4322008Journal of medicinal chemistry, May-22, Volume: 51, Issue:10
Combination of non-natural D-amino acid derivatives and allophenylnorstatine-dimethylthioproline scaffold in HIV protease inhibitors have high efficacy in mutant HIV.
AID374648Decrease in steady state AUC (0 to 12 hrs) of voriconazole in human at 400 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 10 followed by 400 mg,2007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID1473738Inhibition of human BSEP overexpressed in Sf9 cell membrane vesicles assessed as uptake of [3H]-taurocholate in presence of ATP measured after 15 to 20 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID235095Resistance of constructed mutant 82A virus was calculated as (IC90 for a mutant virus / IC90 of wt HXB2 virus). 1997Journal of medicinal chemistry, Jan-17, Volume: 40, Issue:2
Cyclic urea amides: HIV-1 protease inhibitors with low nanomolar potency against both wild type and protease inhibitor resistant mutants of HIV.
AID353726Aqueous solubility in water by shake flask method2009Journal of medicinal chemistry, May-14, Volume: 52, Issue:9
Water-soluble prodrugs of the human immunodeficiency virus protease inhibitors lopinavir and ritonavir.
AID1221980Transporter substrate index of efflux ratio in human Caco2 cells at 10 uM up to 120 mins by HPLC-MC analysis in presence of 10 uM of MRP2 inhibitor MK5712011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Attenuation of intestinal absorption by major efflux transporters: quantitative tools and strategies using a Caco-2 model.
AID588214FDA HLAED, liver enzyme composite activity2004Current drug discovery technologies, Dec, Volume: 1, Issue:4
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
AID588216FDA HLAED, serum glutamic oxaloacetic transaminase (SGOT) increase2004Current drug discovery technologies, Dec, Volume: 1, Issue:4
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
AID242869Association rate constant for the interaction between the compound and serum albumin2005Journal of medicinal chemistry, May-19, Volume: 48, Issue:10
Early absorption and distribution analysis of antitumor and anti-AIDS drugs: lipid membrane and plasma protein interactions.
AID20618Oral bioavailability in rats at the dose of 10 mg/kg po2000Journal of medicinal chemistry, Feb-10, Volume: 43, Issue:3
Protease inhibitors: current status and future prospects.
AID553570Cytotoxicity against human MT2 cells by MTT assay2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID738131Binding affinity to C-terminal His-tagged wild type CYP3A4 (unknown origin) expressed in Escherichia coli assessed as rate constant of fast phase of reaction by stopped flow assay2013Journal of medicinal chemistry, May-09, Volume: 56, Issue:9
Pyridine-substituted desoxyritonavir is a more potent inhibitor of cytochrome P450 3A4 than ritonavir.
AID543382Cmax in HIV-infected patient at 100 mg, qd2008Antimicrobial agents and chemotherapy, Sep, Volume: 52, Issue:9
Effects of minocycline and valproic acid coadministration on atazanavir plasma concentrations in human immunodeficiency virus-infected adults receiving atazanavir-ritonavir.
AID162686Inhibitory activity against HIV protease at a compound concentration of 0.5 nM in MT-4 cells2003Bioorganic & medicinal chemistry letters, Nov-03, Volume: 13, Issue:21
Synthesis and SAR studies of potent HIV protease inhibitors containing novel dimethylphenoxyl acetates as P2 ligands.
AID246195Protease inhibitory activity against HIV-1 r13363 mutant strain was determined2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Design of HIV-1 protease inhibitors active on multidrug-resistant virus.
AID588982Inhibitors of transporters of clinical importance in the absorption and disposition of drugs, OATP1B32010Nature reviews. Drug discovery, Mar, Volume: 9, Issue:3
Membrane transporters in drug development.
AID699540Inhibition of human liver OATP1B3 expressed in HEK293 Flp-In cells assessed as reduction in [3H]E17-betaG uptake at 20 uM incubated for 5 mins by scintillation counting2012Journal of medicinal chemistry, May-24, Volume: 55, Issue:10
Classification of inhibitors of hepatic organic anion transporting polypeptides (OATPs): influence of protein expression on drug-drug interactions.
AID624626Ratio of apparent permeability from basolateral to apical side over apical to basolateral side determined in MDR1-MDCKII cells2001The Journal of pharmacology and experimental therapeutics, Nov, Volume: 299, Issue:2
Rational use of in vitro P-glycoprotein assays in drug discovery.
AID575062Antiviral activity against Human immunodeficiency virus 1 harboring M46I, M46L, I54V, V82A and L76V mutations in viral protease assessed as fold change in drug susceptibility relative to wild type2010Antimicrobial agents and chemotherapy, Nov, Volume: 54, Issue:11
Prevalence, mutation patterns, and effects on protease inhibitor susceptibility of the L76V mutation in HIV-1 protease.
AID557230Drug level in HIV-infected Thai pregnant women serum before intrapartum initiation of 100 mg, po bid in combination with 400 mg, po bid Lopinavir2009Antimicrobial agents and chemotherapy, May, Volume: 53, Issue:5
Early postpartum pharmacokinetics of lopinavir initiated intrapartum in Thai women.
AID369957Ratio of EC50 for HIV2 ROD with protease G17N/V47A mutation to EC50 for wild type HIV2 ROD2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro selection and characterization of human immunodeficiency virus type 2 with decreased susceptibility to lopinavir.
AID575059Antiviral activity against Human immunodeficiency virus 1 harboring protease inhibitor resistance-associated mutations assessed as fold change in drug susceptibility relative to wild type2010Antimicrobial agents and chemotherapy, Nov, Volume: 54, Issue:11
Prevalence, mutation patterns, and effects on protease inhibitor susceptibility of the L76V mutation in HIV-1 protease.
AID415239Oral bioavailability in HIV1 infected patient at 600 mg administered as single dose2009Journal of medicinal chemistry, Apr-23, Volume: 52, Issue:8
Harnessing nature's insight: design of aspartyl protease inhibitors from treatment of drug-resistant HIV to Alzheimer's disease.
AID557275Antiviral activity against HIV1 C harboring L10I/I15V/K20R/L24I/M36I/M46L/I54V/I62V/L63P/K70Q/V82A/L89M in protease encoding region infected in human PHA-PBC assessed as inhibition of p24 Gag protein production by ELISA2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID321696Ratio of EC50 for HIV1 mutant strain 4 to EC50 for wild type HIV12008Bioorganic & medicinal chemistry, Feb-01, Volume: 16, Issue:3
Structure-activity relationships of novel HIV-1 protease inhibitors containing the 3-amino-2-chlorobenzoyl-allophenylnorstatine structure.
AID408216Ratio of EC50 for ritonavir-resistant HIV1 NL432 to EC50 for wild-type HIV1 NL4322008Journal of medicinal chemistry, May-22, Volume: 51, Issue:10
Combination of non-natural D-amino acid derivatives and allophenylnorstatine-dimethylthioproline scaffold in HIV protease inhibitors have high efficacy in mutant HIV.
AID278961Antiviral activity against HIV1 A9 isolate with protease I84A mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID525109Antiviral activity against HIV 1 RIN HIV 1 RIN harboring integrase gene infected in MT-4 cells assessed as inhibition of virus-induced cytopathic effect by MTT assay after 40 passages selected in presence of compound2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Mutations in human immunodeficiency virus type 1 integrase confer resistance to the naphthyridine L-870,810 and cross-resistance to the clinical trial drug GS-9137.
AID269313Antiviral activity against multi drug-resistant HIV1 G variant in human PHA-PBMC cells2006Journal of medicinal chemistry, Aug-24, Volume: 49, Issue:17
Structure-based design of novel HIV-1 protease inhibitors to combat drug resistance.
AID408215Antiviral activity against HIV1 3B in presence of 10% fetal calf serum and alpha1 acid glycoprotein2008Journal of medicinal chemistry, May-22, Volume: 51, Issue:10
Combination of non-natural D-amino acid derivatives and allophenylnorstatine-dimethylthioproline scaffold in HIV protease inhibitors have high efficacy in mutant HIV.
AID321688Antiviral activity against HIV1 mutant strain 32008Bioorganic & medicinal chemistry, Feb-01, Volume: 16, Issue:3
Structure-activity relationships of novel HIV-1 protease inhibitors containing the 3-amino-2-chlorobenzoyl-allophenylnorstatine structure.
AID557286Antiviral activity against HIV1 NL4-3 harboring L10F/V32I/M46I/I54M//A71V/I84V amino acid substitution in protease encoding region infected in human MT4 cells assessed as inhibition of p24 Gag protein production selected at 5 uM of amprenavir by ELISA2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID721747Inhibition of human MATE1-mediated [14]-metformin uptake expressed in HEK293 cells after 1.5 mins by scintillation counting analysis2013Journal of medicinal chemistry, Feb-14, Volume: 56, Issue:3
Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling.
AID1218864Inhibition of OATP1B1 (unknown origin) expressed in HEK293 cells using estradiol-17beta-glucuronide substrate2012Drug metabolism and disposition: the biological fate of chemicals, Aug, Volume: 40, Issue:8
The development, characterization, and application of an OATP1B1 inhibition assay in drug discovery.
AID679665TP_TRANSPORTER: transepithelial transport (basal to apical) in MRP2-expressing MDCKII cells2002AIDS (London, England), Nov-22, Volume: 16, Issue:17
Multidrug resistance protein 2 (MRP2) transports HIV protease inhibitors, and transport can be enhanced by other drugs.
AID369943Antiviral activity against HIV2 CDC310319 isolate infected in human PBMC assessed as inhibition of virus production after 5 days by Lenti-RT activity assay2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro selection and characterization of human immunodeficiency virus type 2 with decreased susceptibility to lopinavir.
AID721745Ratio of Cmax unbound to IC50 for human MATE1-mediated ASP+ uptake expressed in HEK293 cells2013Journal of medicinal chemistry, Feb-14, Volume: 56, Issue:3
Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling.
AID525280Antimicrobial activity against chloroquine-sensitive Plasmodium falciparum D10 after 48 hrs2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
Antimalarial asexual stage-specific and gametocytocidal activities of HIV protease inhibitors.
AID291000Antiviral activity against ritonavir and nelfinavir-resistant HIV1 NL4322007Bioorganic & medicinal chemistry letters, Aug-01, Volume: 17, Issue:15
Synthesis and antiviral property of allophenylnorstatine-based HIV protease inhibitors incorporating D-cysteine derivatives as P2/P3 moieties.
AID1217721Time dependent inhibition of CYP2C19 in human liver microsomes at 10 uM by LC/MS system2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Combination of GSH trapping and time-dependent inhibition assays as a predictive method of drugs generating highly reactive metabolites.
AID1219728Inhibition of CYP3A4-mediated lopinavir bioactivation in human liver microsomes assessed as inhibition of GSH-conjugated monohydroxylated lopinavir adduct III to XII formation at 2 uM after after 50 mins by UPLC-TOFMS analysis2012Drug metabolism and disposition: the biological fate of chemicals, Jan, Volume: 40, Issue:1
CYP3A4-mediated lopinavir bioactivation and its inhibition by ritonavir.
AID1079940Granulomatous liver disease, proven histopathologically. Value is number of references indexed. [column 'GRAN' in source]
AID1079939Cirrhosis, proven histopathologically. Value is number of references indexed. [column 'CIRRH' in source]
AID681119TP_TRANSPORTER: inhibition of Calcein-AM efflux in Mdr1a-expressing LLC-PK1 cells2003Journal of medicinal chemistry, Apr-24, Volume: 46, Issue:9
Comparison of in vitro P-glycoprotein screening assays: recommendations for their use in drug discovery.
AID243930Percent inhibition of recombinant human T-cell leukemia virus type I (HTLV-1) protease was determined at 100 uM concentration2004Bioorganic & medicinal chemistry letters, Dec-06, Volume: 14, Issue:23
Identification of peptidomimetic HTLV-I protease inhibitors containing hydroxymethylcarbonyl (HMC) isostere as the transition-state mimic.
AID557292Antiviral activity against HIV1 NL4-3 harboring L10F/L33F/M46I/I47V/Q58E/V82I/I84V/I85V amino acid substitution in protease encoding region infected in human MT4 cells assessed as inhibition of p24 Gag protein production selected at 5 uM of GRL-02031 by E2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID374651Decrease in steady state Cmax of voriconazole in human at 100 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 10 followed by 100 mg, po of drug c2007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID1069006Inhibition of HIV-1 protease2014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Structure-activity relationships of diamine inhibitors of cytochrome P450 (CYP) 3A as novel pharmacoenhancers, part I: core region.
AID519575Antiviral activity against HIV1 P13 infected in human MT4 cells derived from viral passages with A-790742 harboring protease M46I, L63P, A71V, and V82G mutation assessed as reduction in viral cytopathogenicity treated 1 hr post infection measured 5 days p2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
Characterization of a novel human immunodeficiency virus type 1 protease inhibitor, A-790742.
AID557273Antiviral activity against HIV1 A harboring L10I/I15V/E35D/N37E/K45R/I54V/L63P/A71V/V82T/L90M/I93L/C95F in protease encoding region infected in human PHA-PBC assessed as inhibition of p24 Gag protein production by ELISA2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID1079936Choleostatic liver toxicity, either proven histopathologically or where the ratio of maximal ALT or AST activity above normal to that of Alkaline Phosphatase is < 2 (see ACUTE). Value is number of references indexed. [column 'CHOLE' in source]
AID210277Inhibition of HIV RNA synthesis in T-cell line1997Journal of medicinal chemistry, Dec-05, Volume: 40, Issue:25
Nonsymmetrically substituted cyclic urea HIV protease inhibitors.
AID374594Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 NL4-32007Antimicrobial agents and chemotherapy, Nov, Volume: 51, Issue:11
In vitro antiviral activity and cross-resistance profile of PL-100, a novel protease inhibitor of human immunodeficiency virus type 1.
AID625281Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for cholelithiasis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID278966Antiviral activity against HIV1 A14 isolate with protease I84A mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID242933Dissociation rate constant for the interaction between the compound and serum albumin2005Journal of medicinal chemistry, May-19, Volume: 48, Issue:10
Early absorption and distribution analysis of antitumor and anti-AIDS drugs: lipid membrane and plasma protein interactions.
AID557224AUC (0 to 12 hrs) in HIV-infected Thai pregnant women at 100 mg, po bid initiated intrapartum in combination with 400 mg, po bid Lopinavir measured within 72 hrs postpartum2009Antimicrobial agents and chemotherapy, May, Volume: 53, Issue:5
Early postpartum pharmacokinetics of lopinavir initiated intrapartum in Thai women.
AID104972Antiviral activity was tested in absence of human serum in MT-4 cells (in vitro)2003Bioorganic & medicinal chemistry letters, Nov-03, Volume: 13, Issue:21
Synthesis and SAR studies of potent HIV protease inhibitors containing novel dimethylphenoxyl acetates as P2 ligands.
AID525110Antiviral activity against HIV 1 RIN HIV 1 RIN harboring integrase gene infected in MT-4 cells assessed as inhibition of virus-induced cytopathic effect by MTT assay after 60 passages selected in presence of compound2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Mutations in human immunodeficiency virus type 1 integrase confer resistance to the naphthyridine L-870,810 and cross-resistance to the clinical trial drug GS-9137.
AID365483Antiviral activity against HIV1 3B in MT4 cells assessed as virus p24 antigen production administered at 5 hrs post infection by ELISA2008Journal of medicinal chemistry, Sep-11, Volume: 51, Issue:17
Structure-activity relationship study on anti-HIV 6-desfluoroquinolones.
AID1079948Times to onset, minimal and maximal, observed in the indexed observations. [column 'DELAI' in source]
AID1215086Activation of human PXR expressed in human HepG2 (DPX-2) cells after 24 hrs by luciferase reporter gene based luminescent analysis2011Drug metabolism and disposition: the biological fate of chemicals, Jan, Volume: 39, Issue:1
Identification of clinically used drugs that activate pregnane X receptors.
AID322109Antiviral activity against HIV1 GRL98065p20 in MT4 cells assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID1129361Unbound fraction in HEK293 cell homogenate at 0.1 uM by equilibrium dialysis based UPLC-MS/MS analysis2014Journal of medicinal chemistry, Apr-10, Volume: 57, Issue:7
A high-throughput cell-based method to predict the unbound drug fraction in the brain.
AID369176CL/F in HIV1 infected human at 100 mg, po administered twice daily2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
Association of saquinavir plasma concentrations with side effects but not with antiretroviral outcome in patients infected with protease inhibitor-susceptible human immunodeficiency virus type 1.
AID1657045Binding affinity to human full length CYP3A4 assessed as spectral dissociation constant by spectrophotometric analysis
AID738139Binding affinity to C-terminal His-tagged wild type CYP3A4 (unknown origin)-bromoergocryptine complex expressed in Escherichia coli by spectrophotometric analysis2013Journal of medicinal chemistry, May-09, Volume: 56, Issue:9
Pyridine-substituted desoxyritonavir is a more potent inhibitor of cytochrome P450 3A4 than ritonavir.
AID572569Apparent oral clearance in HIV-1 infected patient at 100 mg, po bid coadministered with 400 mg, po bid of Lopinavir and 300 mg, po qd of atazanavir2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Decrease of atazanavir and lopinavir plasma concentrations in a boosted double human immunodeficiency virus protease inhibitor salvage regimen.
AID1578469Unbound tissue partition coefficient, ratio of drug level in brain to plasma in Wistar Han rat at 1 to 2 mg/kg, iv infused for 10 to 20 hrs by LC-MS/MS analysis2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID416857Decrease in P-glycoprotein-mediated tenofovir disoproxil fumarate efflux in MDCK2 expressing human MDR1 cells at 20 uM2007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Effects of human immunodeficiency virus protease inhibitors on the intestinal absorption of tenofovir disoproxil fumarate in vitro.
AID553578Antiviral activity against HIV1 JSL harboring L10I/L24I/L33F/E35D/M36I/N37S/M46L/I54V/R57K/I62V/L63P/A71V/G73S/82A in protease encoding region infected in human PHA-PBC assessed as inhibition of p24 Gag protein production by ELISA2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID290994Antiviral activity against HIV1 3B in MT4 cells by MTT assay2007Bioorganic & medicinal chemistry letters, Aug-01, Volume: 17, Issue:15
Synthesis and antiviral property of allophenylnorstatine-based HIV protease inhibitors incorporating D-cysteine derivatives as P2/P3 moieties.
AID150753Inhibition of P-glycoprotein, mouse L-mdr1a expressed in LLC-PK1 epithelial cells using calcein-AM polarisation assay2003Journal of medicinal chemistry, Apr-24, Volume: 46, Issue:9
Comparison of in vitro P-glycoprotein screening assays: recommendations for their use in drug discovery.
AID290998Antiviral activity against ritonavir-resistant HIV1 NL4322007Bioorganic & medicinal chemistry letters, Aug-01, Volume: 17, Issue:15
Synthesis and antiviral property of allophenylnorstatine-based HIV protease inhibitors incorporating D-cysteine derivatives as P2/P3 moieties.
AID525104Antiviral activity against HIV 1 3B harboring integrase E92Q S230N double mutant infected in MT-4 cells assessed as inhibition of virus-induced cytopathic effect by MTT assay selected after 20 passages in presence of compound2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Mutations in human immunodeficiency virus type 1 integrase confer resistance to the naphthyridine L-870,810 and cross-resistance to the clinical trial drug GS-9137.
AID278954Antiviral activity against HIV1 A2 isolate with protease I84A mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID1221972Apparent permeability from basolateral to apical side of human Caco2 cells at 10 uM up to 120 mins by HPLC-MC analysis in presence of 10 uM of MRP2 inhibitor MK5712011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Attenuation of intestinal absorption by major efflux transporters: quantitative tools and strategies using a Caco-2 model.
AID1578458Fraction unbound in Wistar Han rat brain at 2 uM incubated for 6 hrs by equilibrium dialysis method2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID1217733Time dependent inhibition of CYP2B6 (unknown origin) at 0.3 uM by LC/MS system2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Combination of GSH trapping and time-dependent inhibition assays as a predictive method of drugs generating highly reactive metabolites.
AID1220555Fraction unbound in Sprague-Dawley rat brain homogenates at 1 uM after 6 hrs by equilibrium dialysis method2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Species independence in brain tissue binding using brain homogenates.
AID470605AUC in Beagle dog at 5 mg/kg, po2009Bioorganic & medicinal chemistry letters, Sep-15, Volume: 19, Issue:18
Synthesis and evaluation of inhibitors of cytochrome P450 3A (CYP3A) for pharmacokinetic enhancement of drugs.
AID470603Intrinsic clearance in human liver microsomes2009Bioorganic & medicinal chemistry letters, Sep-15, Volume: 19, Issue:18
Synthesis and evaluation of inhibitors of cytochrome P450 3A (CYP3A) for pharmacokinetic enhancement of drugs.
AID1267626Antiviral activity against ritonavir-resistant HIV1 B12 harboring reverse transcriptase L10I/I15V/I54V/L63P/V82A/I85V mutant infected in human MT4 cells2015Journal of medicinal chemistry, Dec-24, Volume: 58, Issue:24
Design and Synthesis of DiselenoBisBenzamides (DISeBAs) as Nucleocapsid Protein 7 (NCp7) Inhibitors with anti-HIV Activity.
AID257271Antiviral activity against HIV1 in the presence of 50% human serum2005Bioorganic & medicinal chemistry letters, Dec-15, Volume: 15, Issue:24
Synthesis and activity of N-acyl azacyclic urea HIV-1 protease inhibitors with high potency against multiple drug resistant viral strains.
AID82750Fold increase in EC50 for antiviral activity against HIV Protease-Resistant strains with E protease amino acid substitutions2001Journal of medicinal chemistry, Jul-05, Volume: 44, Issue:14
4-Hydroxy-5,6-dihydropyrones as inhibitors of HIV protease: the effect of heterocyclic substituents at C-6 on antiviral potency and pharmacokinetic parameters.
AID278981Resistance to HIV1 with protease 46I, 54I and I84V mutation in HEK 293 cells relative to similar background2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID1069011Inhibition of human CYP2C192014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Structure-activity relationships of diamine inhibitors of cytochrome P450 (CYP) 3A as novel pharmacoenhancers, part I: core region.
AID278953Antiviral activity against HIV1 A1 isolate with protease I84A mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID235094Resistance of constructed mutant 48V/90M virus was calculated as (IC90 for a mutant virus / IC90 of wt HXB2 virus)1997Journal of medicinal chemistry, Jan-17, Volume: 40, Issue:2
Cyclic urea amides: HIV-1 protease inhibitors with low nanomolar potency against both wild type and protease inhibitor resistant mutants of HIV.
AID162706Equilibrium constant for the interaction between inhibitor and HIV-1 Protease2002Journal of medicinal chemistry, Dec-05, Volume: 45, Issue:25
Relationships between structure and interaction kinetics for HIV-1 protease inhibitors.
AID625285Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatic necrosis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID104304Anti-HIV-1 activity against mutant HIV-1 in MT-4 cells (mutation selected with saquinavir)2001Journal of medicinal chemistry, Sep-13, Volume: 44, Issue:19
Design and synthesis of potent C(2)-symmetric diol-based HIV-1 protease inhibitors: effects of fluoro substitution.
AID1896147Antiviral activity against hepatitis D virus infected in human Huh-7 cells overexpressing NTCP assessed as reduction in intracellular HDV RNA for 20 hrs followed by viral infection further compound washout and measured after 6 days post infection by RT-qP2022Journal of medicinal chemistry, 10-13, Volume: 65, Issue:19
Inhibiting Sodium Taurocholate Cotransporting Polypeptide in HBV-Related Diseases: From Biological Function to Therapeutic Potential.
AID160766Dissociation rate constant for the interaction between inhibitor and HIV-1 protease2002Journal of medicinal chemistry, Dec-05, Volume: 45, Issue:25
Relationships between structure and interaction kinetics for HIV-1 protease inhibitors.
AID235097Resistance of constructed mutant 84V virus was calculated as (IC90 for a mutant virus / IC90 of wt HXB2 virus). 1997Journal of medicinal chemistry, Jan-17, Volume: 40, Issue:2
Cyclic urea amides: HIV-1 protease inhibitors with low nanomolar potency against both wild type and protease inhibitor resistant mutants of HIV.
AID1069012Antiviral activity against HIV1 by cell based assay2014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Structure-activity relationships of diamine inhibitors of cytochrome P450 (CYP) 3A as novel pharmacoenhancers, part I: core region.
AID1845236Inhibition of SARS-CoV-2 MPro2021Bioorganic & medicinal chemistry, 01-01, Volume: 29Protease targeted COVID-19 drug discovery and its challenges: Insight into viral main protease (Mpro) and papain-like protease (PLpro) inhibitors.
AID408211Antiviral activity against nelfinavir-resistant HIV1 NL4322008Journal of medicinal chemistry, May-22, Volume: 51, Issue:10
Combination of non-natural D-amino acid derivatives and allophenylnorstatine-dimethylthioproline scaffold in HIV protease inhibitors have high efficacy in mutant HIV.
AID1221962Efflux ratio of permeability from apical to basolateral side over basolateral to apical side of human Caco2 cells at 10 uM up to 120 mins by HPLC-MC analysis in presence of 1 uM of P-gp inhibitor LY3359792011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Attenuation of intestinal absorption by major efflux transporters: quantitative tools and strategies using a Caco-2 model.
AID257270Antiviral activity against HIV1 in the absence of human serum2005Bioorganic & medicinal chemistry letters, Dec-15, Volume: 15, Issue:24
Synthesis and activity of N-acyl azacyclic urea HIV-1 protease inhibitors with high potency against multiple drug resistant viral strains.
AID1217723Time dependent inhibition of CYP2D6 (unknown origin) at 10 uM by LC/MS system2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Combination of GSH trapping and time-dependent inhibition assays as a predictive method of drugs generating highly reactive metabolites.
AID369837Antiviral activity against HIV1 in presence of 50% human serum2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro selection and characterization of human immunodeficiency virus type 2 with decreased susceptibility to lopinavir.
AID1221971Apparent permeability from apical to basolateral side of human Caco2 cells at 10 uM up to 120 mins by HPLC-MC analysis in presence of 10 uM of MRP2 inhibitor MK5712011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Attenuation of intestinal absorption by major efflux transporters: quantitative tools and strategies using a Caco-2 model.
AID242932Dissociation rate constant for the interaction between the compound and serum albumin2005Journal of medicinal chemistry, May-19, Volume: 48, Issue:10
Early absorption and distribution analysis of antitumor and anti-AIDS drugs: lipid membrane and plasma protein interactions.
AID1443995Hepatotoxicity in human assessed as drug-induced liver injury2014Hepatology (Baltimore, Md.), Sep, Volume: 60, Issue:3
Human drug-induced liver injury severity is highly associated with dual inhibition of liver mitochondrial function and bile salt export pump.
AID543383Cmax in HIV-infected patient at 100 mg, qd co-administered with 100 mg minocycline, bid for 15 days2008Antimicrobial agents and chemotherapy, Sep, Volume: 52, Issue:9
Effects of minocycline and valproic acid coadministration on atazanavir plasma concentrations in human immunodeficiency virus-infected adults receiving atazanavir-ritonavir.
AID408210Antiviral activity against wild type HIV1 NL4322008Journal of medicinal chemistry, May-22, Volume: 51, Issue:10
Combination of non-natural D-amino acid derivatives and allophenylnorstatine-dimethylthioproline scaffold in HIV protease inhibitors have high efficacy in mutant HIV.
AID525166Antimicrobial activity against chloroquine-resistant Plasmodium falciparum Dd2 infected in human erythrocytes assessed as potentiation of choloroquine-mediated antimalarial activity by light microscopy2008Antimicrobial agents and chemotherapy, Jul, Volume: 52, Issue:7
Synergy of human immunodeficiency virus protease inhibitors with chloroquine against Plasmodium falciparum in vitro and Plasmodium chabaudi in vivo.
AID374661Toxicity in human assessed as serious adverse events at 100 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 102007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID105372Cytotoxicity against MT-4 cells was determined by using cytopathic effect assay1998Bioorganic & medicinal chemistry letters, Dec-15, Volume: 8, Issue:24
Potent piperazine hydroxyethylamine HIV protease inhibitors containing novel P3 ligands.
AID1657049Inhibition of human full length CYP3A4 assessed using 7-benzyloxy-4 (trifluoromethyl)coumarin as substrate preincubated for 20 mins in presence of NADPH followed by substrate addition and measured for 2 mins by fluorometric analysis
AID1578470Unbound tissue partition coefficient, ratio of drug level in adipose to plasma in Wistar Han rat at 1 to 2 mg/kg, iv infused for 10 to 20 hrs by LC-MS/MS analysis2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID557235Drug level in HIV-infected Thai pregnant women serum at 100 mg, po bid initiated intrapartum administered for 30 days in combination with 400 mg, po bid Lopinavir measured 2 hrs post last dose2009Antimicrobial agents and chemotherapy, May, Volume: 53, Issue:5
Early postpartum pharmacokinetics of lopinavir initiated intrapartum in Thai women.
AID1636440Drug activation in human Hep3B cells assessed as human CYP2D6-mediated drug metabolism-induced cytotoxicity measured as decrease in cell viability at 300 uM pre-incubated with BSO for 18 hrs followed by incubation with compound for 3 hrs in presence of NA2016Bioorganic & medicinal chemistry letters, 08-15, Volume: 26, Issue:16
Development of a cell viability assay to assess drug metabolite structure-toxicity relationships.
AID246193Protease inhibitory activity against HIV-1 r13025 mutant strain was determined2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Design of HIV-1 protease inhibitors active on multidrug-resistant virus.
AID721751Inhibition of human OCT2-mediated ASP+ uptake expressed in HEK293 cells after 3 mins by fluorescence assay2013Journal of medicinal chemistry, Feb-14, Volume: 56, Issue:3
Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling.
AID1220554Fraction unbound in Wistar Han rat brain homogenates at 1 uM after 6 hrs by equilibrium dialysis method2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Species independence in brain tissue binding using brain homogenates.
AID543371AUC (0 to 24 hrs) in HIV-infected patient at 100 mg, qd co-administered with 100 mg minocycline, bid for 15 days2008Antimicrobial agents and chemotherapy, Sep, Volume: 52, Issue:9
Effects of minocycline and valproic acid coadministration on atazanavir plasma concentrations in human immunodeficiency virus-infected adults receiving atazanavir-ritonavir.
AID369175AUC in HIV1 infected human at 100 mg, po administered twice daily2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
Association of saquinavir plasma concentrations with side effects but not with antiretroviral outcome in patients infected with protease inhibitor-susceptible human immunodeficiency virus type 1.
AID1443991Induction of mitochondrial dysfunction in Sprague-Dawley rat liver mitochondria assessed as inhibition of mitochondrial respiration per mg mitochondrial protein measured for 20 mins by A65N-1 oxygen probe based fluorescence assay2014Hepatology (Baltimore, Md.), Sep, Volume: 60, Issue:3
Human drug-induced liver injury severity is highly associated with dual inhibition of liver mitochondrial function and bile salt export pump.
AID444056Fraction escaping gut-wall elimination in human2010Journal of medicinal chemistry, Feb-11, Volume: 53, Issue:3
Physicochemical space for optimum oral bioavailability: contribution of human intestinal absorption and first-pass elimination.
AID1267619Cytotoxicity against human HUT78 cells assessed as reduction in cell viability after 5 days by MTT assay2015Journal of medicinal chemistry, Dec-24, Volume: 58, Issue:24
Design and Synthesis of DiselenoBisBenzamides (DISeBAs) as Nucleocapsid Protein 7 (NCp7) Inhibitors with anti-HIV Activity.
AID238043Binding affinity for human immunodeficiency virus type 1 protease2004Journal of medicinal chemistry, Nov-18, Volume: 47, Issue:24
Improved structure-activity relationship analysis of HIV-1 protease inhibitors using interaction kinetic data.
AID525107Antiviral activity against HIV 1 RIN harboring integrase gene infected in MT-4 cells assessed as inhibition of virus-induced cytopathic effect by MTT assay2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Mutations in human immunodeficiency virus type 1 integrase confer resistance to the naphthyridine L-870,810 and cross-resistance to the clinical trial drug GS-9137.
AID444051Total clearance in human2010Journal of medicinal chemistry, Feb-11, Volume: 53, Issue:3
Physicochemical space for optimum oral bioavailability: contribution of human intestinal absorption and first-pass elimination.
AID297671Antiviral activity against HIV1 infected MT4 cells by MTT method2007Journal of medicinal chemistry, Sep-06, Volume: 50, Issue:18
Design and synthesis of HIV-1 protease inhibitors incorporating oxazolidinones as P2/P2' ligands in pseudosymmetric dipeptide isosteres.
AID238682Inhibition constant for human immunodeficiency virus type 1 protease2004Journal of medicinal chemistry, Nov-18, Volume: 47, Issue:24
Improved structure-activity relationship analysis of HIV-1 protease inhibitors using interaction kinetic data.
AID1079944Benign tumor, proven histopathologically. Value is number of references indexed. [column 'T.BEN' in source]
AID557240Ratio of drug level in 2 hrs to 30 days post last dose of HIV-infected Thai pregnant women serum at 100 mg, po bid initiated intrapartum in combination with 400 mg, po bid Lopinavir2009Antimicrobial agents and chemotherapy, May, Volume: 53, Issue:5
Early postpartum pharmacokinetics of lopinavir initiated intrapartum in Thai women.
AID1267609Antiviral activity against HIV2 ROD infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay2015Journal of medicinal chemistry, Dec-24, Volume: 58, Issue:24
Design and Synthesis of DiselenoBisBenzamides (DISeBAs) as Nucleocapsid Protein 7 (NCp7) Inhibitors with anti-HIV Activity.
AID278978Antiviral activity against HIV1 C11 isolate with protease I84C mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID705596Inhibition of CYP3A4-mediated testosterone 6 beta hydroxylation in human liver microsome by Dixon plot analysis2012Journal of medicinal chemistry, Jun-14, Volume: 55, Issue:11
Mechanism-based inactivation (MBI) of cytochrome P450 enzymes: structure-activity relationships and discovery strategies to mitigate drug-drug interaction risks.
AID1152127Antiviral activity against HIV-1 3B infected in human MT4 cells assessed as inhibition of p24 antigen production by measuring time delay in loss of compound activity at 50 to 100 times EC50 by ELISA2014Journal of medicinal chemistry, Jun-12, Volume: 57, Issue:11
Investigation of a novel series of 2-hydroxyisoquinoline-1,3(2H,4H)-diones as human immunodeficiency virus type 1 integrase inhibitors.
AID721742Ratio of Cmax unbound to IC50 for human MATE1-mediated [14]-metformin uptake expressed in polarized MDCK2 cells2013Journal of medicinal chemistry, Feb-14, Volume: 56, Issue:3
Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling.
AID372213AUC (0 to 12 hrs) in human at 400 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 10 followed by 400 mg, po of drug coadministered with voriconaz2007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID1267618Antiviral activity against HIV1 3B persistently infected in HUT78 cells assessed as reduction of p24 production after 43 hrs by ELISA2015Journal of medicinal chemistry, Dec-24, Volume: 58, Issue:24
Design and Synthesis of DiselenoBisBenzamides (DISeBAs) as Nucleocapsid Protein 7 (NCp7) Inhibitors with anti-HIV Activity.
AID1604604Half life in plasma (unknown origin)2020Journal of medicinal chemistry, 03-26, Volume: 63, Issue:6
Urea Derivatives in Modern Drug Discovery and Medicinal Chemistry.
AID588212Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in rodents2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID1221956Apparent permeability from apical to basolateral side of human Caco2 cells at 10 uM up to 120 mins by HPLC-MC analysis2011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Attenuation of intestinal absorption by major efflux transporters: quantitative tools and strategies using a Caco-2 model.
AID369173Tmax in HIV1 infected human at 100 mg, po administered twice daily2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
Association of saquinavir plasma concentrations with side effects but not with antiretroviral outcome in patients infected with protease inhibitor-susceptible human immunodeficiency virus type 1.
AID575063Antiviral activity against Human immunodeficiency virus 1 harboring M46I mutation in viral protease assessed as fold change in drug susceptibility relative to wild type2010Antimicrobial agents and chemotherapy, Nov, Volume: 54, Issue:11
Prevalence, mutation patterns, and effects on protease inhibitor susceptibility of the L76V mutation in HIV-1 protease.
AID1267608Antiviral activity against HIV1 3B infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay2015Journal of medicinal chemistry, Dec-24, Volume: 58, Issue:24
Design and Synthesis of DiselenoBisBenzamides (DISeBAs) as Nucleocapsid Protein 7 (NCp7) Inhibitors with anti-HIV Activity.
AID553576Antiviral activity against HIV1 TM harboring L10I/K14R/R41K/M46L/I54V/L63P/A71V/V82A/L90M/I93L in protease encoding region infected in human PHA-PBC assessed as inhibition of p24 Gag protein production by ELISA2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID210298The concentration required to inhibit HIV-1 RNA synthesis by 90%. Viral RNA quantified by a sandwich hybridization assay1996Journal of medicinal chemistry, Oct-11, Volume: 39, Issue:21
HIV protease inhibitory bis-benzamide cyclic ureas: a quantitative structure-activity relationship analysis.
AID1498798Therapeutic index, ratio of CC50 for human MT4 cells to EC50 for for HIV1 3B protease infected in human MT4 cells2018Bioorganic & medicinal chemistry letters, 08-01, Volume: 28, Issue:14
Design and synthesis of selenazole-substituted ritonavir analogs.
AID553575Antiviral activity against HIV1 MOKW infected in human PHA-PBC assessed as inhibition of p24 Gag protein production by ELISA2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID572589Cmax in HIV-1 infected patient at 100 mg, po bid coadministered with 400 mg, po bid of Lopinavir and NRTI2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Decrease of atazanavir and lopinavir plasma concentrations in a boosted double human immunodeficiency virus protease inhibitor salvage regimen.
AID263208Antiviral activity against HIV1 EP13 in MT4 cells2006Bioorganic & medicinal chemistry letters, Apr-01, Volume: 16, Issue:7
Ultra-potent P1 modified arylsulfonamide HIV protease inhibitors: the discovery of GW0385.
AID278973Antiviral activity against HIV1 C6 isolate with protease I84C mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID1215350Time dependent inhibition of CYP3A4 in human liver microsomes assessed as conversion of testosterone to 6beta-hydroxytestosterone at 0.01 to 100 uM preincubated for 60 mins followed by testosterone treatment measured after 10 mins by refined CYP450 IC50 s2011Drug metabolism and disposition: the biological fate of chemicals, Jun, Volume: 39, Issue:6
A refined cytochrome P540 IC₅₀ shift assay for reliably identifying CYP3A time-dependent inhibitors.
AID1578473Unbound tissue partition coefficient, ratio of drug level in brain to plasma in mouse administered as single dose2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID525171Antimicrobial activity against chloroquine-resistant Plasmodium chabaudi ASCQ infected in NIH mice (Mus musculus) assessed as potentiation of 2.5 mg/kg chloroquine-mediated antimalarial activity at 100 mg/kg, perorally administered after 72 hrs post inocu2008Antimicrobial agents and chemotherapy, Jul, Volume: 52, Issue:7
Synergy of human immunodeficiency virus protease inhibitors with chloroquine against Plasmodium falciparum in vitro and Plasmodium chabaudi in vivo.
AID1221957Apparent permeability from basolateral to apical side of human Caco2 cells at 10 uM up to 120 mins by HPLC-MC analysis2011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Attenuation of intestinal absorption by major efflux transporters: quantitative tools and strategies using a Caco-2 model.
AID427597Inhibition of SAP9-dependent growth in Candida albicans SAP2MS4B expressing SAP9deltaC493 at 100 uM incubated in YCB-BSA medium2008Antimicrobial agents and chemotherapy, Jan, Volume: 52, Issue:1
Tetracycline-inducible expression of individual secreted aspartic proteases in Candida albicans allows isoenzyme-specific inhibitor screening.
AID160316Binding affinity to inhibit the purified wild-type HIV-1 Protease1997Journal of medicinal chemistry, Jan-17, Volume: 40, Issue:2
Cyclic urea amides: HIV-1 protease inhibitors with low nanomolar potency against both wild type and protease inhibitor resistant mutants of HIV.
AID1217727Intrinsic clearance for reactive metabolites formation per mg of protein in human liver microsomes based on [3H]GSH adduct formation rate at 100 uM by [3H]GSH trapping assay2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Combination of GSH trapping and time-dependent inhibition assays as a predictive method of drugs generating highly reactive metabolites.
AID278960Antiviral activity against HIV1 A8 isolate with protease I84A mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID321687Antiviral activity against HIV1 mutant strain 22008Bioorganic & medicinal chemistry, Feb-01, Volume: 16, Issue:3
Structure-activity relationships of novel HIV-1 protease inhibitors containing the 3-amino-2-chlorobenzoyl-allophenylnorstatine structure.
AID278818Antimalarial activity against Plasmodium falciparum Dd22007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Synergistic interactions of the antiretroviral protease inhibitors saquinavir and ritonavir with chloroquine and mefloquine against Plasmodium falciparum in vitro.
AID372207Cmax in human at 400 mg, po bid from day 11 to day 20 pretreated with placebo from day 1 to day 3 followed by washout from day 4 to day 102007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID369952Antiviral activity against HIV2 ROD with protease G17N mutation infected in human CEM cells assessed as inhibition of virus production after 7 days by Lenti-RT activity assay2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro selection and characterization of human immunodeficiency virus type 2 with decreased susceptibility to lopinavir.
AID624629Inhibition of Pgp expressed in MDR1-MDCKII cells measured by calcein-AM assay2001The Journal of pharmacology and experimental therapeutics, Nov, Volume: 299, Issue:2
Rational use of in vitro P-glycoprotein assays in drug discovery.
AID557280Ratio of EC50 for HIV1 A harboring L10I/I15V/E35D/N37E/K45R/I54V/L63P/A71V/V82T/L90M/I93L/C95F in protease encoding region to EC50 for HIV1 ERS104pre2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID557278Ratio of EC50 for HIV1 MM harboring L10I/K43T/M46L/I54V/L63P/A71V/V82A/L90M/Q92K in protease encoding region infected to EC50 for HIV1 ERS104pre2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID1079941Liver damage due to vascular disease: peliosis hepatitis, hepatic veno-occlusive disease, Budd-Chiari syndrome. Value is number of references indexed. [column 'VASC' in source]
AID572571AUC in HIV-1 infected patient at 100 mg, po bid coadministered with 400 mg, po bid of Lopinavir and 300 mg, po qd of atazanavir2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Decrease of atazanavir and lopinavir plasma concentrations in a boosted double human immunodeficiency virus protease inhibitor salvage regimen.
AID33556The ability concentration.)2000Journal of medicinal chemistry, May-18, Volume: 43, Issue:10
Biosensor analysis of the interaction between immobilized human serum albumin and drug compounds for prediction of human serum albumin binding levels.
AID525281Antimicrobial activity against Plasmodium falciparum harboring HFP-tagged Pfs16 protein after 48 hrs2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
Antimalarial asexual stage-specific and gametocytocidal activities of HIV protease inhibitors.
AID557227Drug level in HIV-infected Thai pregnant women serum at 100 mg, po bid initiated intrapartum in combination with 400 mg, po bid Lopinavir measured 4 hrs post last dose2009Antimicrobial agents and chemotherapy, May, Volume: 53, Issue:5
Early postpartum pharmacokinetics of lopinavir initiated intrapartum in Thai women.
AID322111Antiviral activity against HIV1 GRL98065p40 in MT4 cells assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID104974Antiviral activity was tested in presence of 50% human serum in MT-4 cells (in vitro)2003Bioorganic & medicinal chemistry letters, Nov-03, Volume: 13, Issue:21
Synthesis and SAR studies of potent HIV protease inhibitors containing novel dimethylphenoxyl acetates as P2 ligands.
AID572585AUC in HIV-1 infected patient at 100 mg, po bid coadministered with 300 mg, po qd of atazanavir and NRTI2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Decrease of atazanavir and lopinavir plasma concentrations in a boosted double human immunodeficiency virus protease inhibitor salvage regimen.
AID573998Ratio of EC50 for Human immunodeficiency virus 1 3B to EC50 for CHI/1043-resistant Human immunodeficiency virus harboring T66I and Q146K mutations in integrase2008Antimicrobial agents and chemotherapy, Aug, Volume: 52, Issue:8
Preclinical evaluation of 1H-benzylindole derivatives as novel human immunodeficiency virus integrase strand transfer inhibitors.
AID150756Inhibition of P-gp was determined using rhodamine-assay in human CaCo-2 cells2003Journal of medicinal chemistry, Apr-24, Volume: 46, Issue:9
Comparison of in vitro P-glycoprotein screening assays: recommendations for their use in drug discovery.
AID738141Binding affinity to C-terminal His-tagged wild type CYP3A4 (unknown origin) expressed in Escherichia coli assessed as increase in melting temperature by spectrophotometric analysis (Rvb = 52.1 +/- 0.1 degC)2013Journal of medicinal chemistry, May-09, Volume: 56, Issue:9
Pyridine-substituted desoxyritonavir is a more potent inhibitor of cytochrome P450 3A4 than ritonavir.
AID374646Cmax in human at 400 mg, po bid from day 11 to day 20 pretreated with placebo from day 1 to day 3 followed by washout from day 4 to day 10 followed by 400 mg, po of drug coadministered with placebo from day 21 to day 302007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID681358TP_TRANSPORTER: inhibition of Digoxin transepithelial transport (basal to apical) (Digoxin: 5 uM) in Caco-2 cells2000Drug metabolism and disposition: the biological fate of chemicals, Jun, Volume: 28, Issue:6
Pharmacological inhibition of P-glycoprotein transport enhances the distribution of HIV-1 protease inhibitors into brain and testes.
AID573995Ratio of EC50 for Human immunodeficiency virus 1 3B to EC50 for L-708,906-resistant Human immunodeficiency virus harboring T66I, L74M and S230R mutations in integrase2008Antimicrobial agents and chemotherapy, Aug, Volume: 52, Issue:8
Preclinical evaluation of 1H-benzylindole derivatives as novel human immunodeficiency virus integrase strand transfer inhibitors.
AID540232Dose normalised AUC in monkey after po administration2005Xenobiotica; the fate of foreign compounds in biological systems, Feb, Volume: 35, Issue:2
Comparative evaluation of oral systemic exposure of 56 xenobiotics in rat, dog, monkey and human.
AID391270Antiviral activity against multidrug-resistant HIV1/C with protease mutation in phytohemagglutininin-activated PBMC assessed as inhibition of p24G protein production2008Journal of medicinal chemistry, Oct-09, Volume: 51, Issue:19
Flexible cyclic ethers/polyethers as novel P2-ligands for HIV-1 protease inhibitors: design, synthesis, biological evaluation, and protein-ligand X-ray studies.
AID322113Antiviral activity against HIV1 MDR/TM X4 in phytohemagglutininin-activated PBMCs assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID519577Antiviral activity against HIV1 clone2 infected in HEK293 cells harboring A-790742-selected protease V82L mutation assessed as reduction in viral replication by luciferase reporter gene assay relative to wild type HIV1 pNL4-32008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
Characterization of a novel human immunodeficiency virus type 1 protease inhibitor, A-790742.
AID1578459Fraction unbound in Wistar Han rat heart at 2 uM incubated for 6 hrs by equilibrium dialysis method2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID369944Ratio of EC50 for HIV2 MS to EC50 for HIV1 NL4-3 infected in human MT4 cells2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro selection and characterization of human immunodeficiency virus type 2 with decreased susceptibility to lopinavir.
AID1636357Drug activation in human Hep3B cells assessed as human CYP3A4-mediated drug metabolism-induced cytotoxicity measured as decrease in cell viability at 300 uM pre-incubated with BSO for 18 hrs followed by incubation with compound for 3 hrs in presence of NA2016Bioorganic & medicinal chemistry letters, 08-15, Volume: 26, Issue:16
Development of a cell viability assay to assess drug metabolite structure-toxicity relationships.
AID278972Antiviral activity against HIV1 C5 isolate with protease I84C mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID519578Antiviral activity against HIV1 clone3 infected in HEK293 cells harboring A-790742-selected protease L33F, K45I, V82L, and I84V mutation assessed as reduction in viral replication by luciferase reporter gene assay relative to wild type HIV1 pNL4-32008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
Characterization of a novel human immunodeficiency virus type 1 protease inhibitor, A-790742.
AID1300995Antiviral activity against HIV1 3B infected in human MT4 cells at 50 to 100 times antiviral EC50 added 1 to 25 hrs post infection assessed as time required for loss of inhibition of viral replication measured at 30 hrs post infection by p24 ELISA method2016European journal of medicinal chemistry, Jul-19, Volume: 1172-hydroxyisoquinoline-1,3(2H,4H)-diones (HIDs) as human immunodeficiency virus type 1 integrase inhibitors: Influence of the alkylcarboxamide substitution of position 4.
AID721741Selectivity ratio of IC50 for human OCT1-mediated ASP+ uptake expressed in HEK293 cells to IC50 for human MATE1-mediated ASP+ uptake expressed in HEK293 cells2013Journal of medicinal chemistry, Feb-14, Volume: 56, Issue:3
Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling.
AID1578457Fraction unbound in Wistar Han rat white adipose tissue at 2 uM incubated for 6 hrs by equilibrium dialysis method2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID572592Apparent oral clearance in HIV-1 infected patient at 100 mg, po bid coadministered with 400 mg, po bid of Lopinavir and NRTI2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Decrease of atazanavir and lopinavir plasma concentrations in a boosted double human immunodeficiency virus protease inhibitor salvage regimen.
AID557276Antiviral activity against HIV1 G harboring L10I/V11I/T12E/I15V/L19I/R41K/M46L/L63P/A71T/V82A/L90M in protease encoding region infected in human PHA-PBC assessed as inhibition of p24 Gag protein production by ELISA2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID1221961Apparent permeability from basolateral to apical side of human Caco2 cells at 10 uM up to 120 mins by HPLC-MC analysis in presence of 1 uM of P-gp inhibitor LY3359792011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Attenuation of intestinal absorption by major efflux transporters: quantitative tools and strategies using a Caco-2 model.
AID1578474Apparent permeability of the compound across dog RRCK cells by MDCK-LE assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID1666548Inhibition of HIV-1 protease at 20 uM relative to control2020Bioorganic & medicinal chemistry letters, 03-01, Volume: 30, Issue:5
Synthesis and biological evaluation of bis-N
AID239819Association rate constant for human immunodeficiency virus type 1 protease2004Journal of medicinal chemistry, Nov-18, Volume: 47, Issue:24
Improved structure-activity relationship analysis of HIV-1 protease inhibitors using interaction kinetic data.
AID1267617Inhibition of NCp7 of HIV1 3B persistently infected in HUT78 cells assessed as accumulation of unprocessed Gag polyprotein after 43 hrs by Western blot analysis2015Journal of medicinal chemistry, Dec-24, Volume: 58, Issue:24
Design and Synthesis of DiselenoBisBenzamides (DISeBAs) as Nucleocapsid Protein 7 (NCp7) Inhibitors with anti-HIV Activity.
AID408214Antiviral activity against HIV1 3B in presence of 10% fetal calf serum2008Journal of medicinal chemistry, May-22, Volume: 51, Issue:10
Combination of non-natural D-amino acid derivatives and allophenylnorstatine-dimethylthioproline scaffold in HIV protease inhibitors have high efficacy in mutant HIV.
AID269311Antiviral activity against multi drug-resistant HIV1 B variant in human PHA-PBMC cells2006Journal of medicinal chemistry, Aug-24, Volume: 49, Issue:17
Structure-based design of novel HIV-1 protease inhibitors to combat drug resistance.
AID588217FDA HLAED, serum glutamic pyruvic transaminase (SGPT) increase2004Current drug discovery technologies, Dec, Volume: 1, Issue:4
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
AID150751Inhibition of P-glycoprotein using ATPase in MDR1 membranes2003Journal of medicinal chemistry, Apr-24, Volume: 46, Issue:9
Comparison of in vitro P-glycoprotein screening assays: recommendations for their use in drug discovery.
AID519576Antiviral activity against HIV1 clone1 infected in HEK293 cells harboring A-790742-selected protease I84V mutation assessed as reduction in viral replication by luciferase reporter gene assay relative to wild type HIV1 pNL4-32008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
Characterization of a novel human immunodeficiency virus type 1 protease inhibitor, A-790742.
AID680821TP_TRANSPORTER: Northern blot in vitro, primary hepatocytes2001The Journal of biological chemistry, Sep-07, Volume: 276, Issue:36
Peptide mimetic HIV protease inhibitors are ligands for the orphan receptor SXR.
AID557228Drug level in HIV-infected Thai pregnant women serum at 100 mg, po bid initiated intrapartum in combination with 400 mg, po bid Lopinavir measured 12 hrs post last dose2009Antimicrobial agents and chemotherapy, May, Volume: 53, Issue:5
Early postpartum pharmacokinetics of lopinavir initiated intrapartum in Thai women.
AID557300Ratio of EC50 for HIV1 NL4-3 harboring L10F/L33F/M46I/I47V/Q58E/V82I/I84V/I85V amino acid substitution in protease encoding region to EC50 for HIV1 NL4-32009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID1069010Inhibition of human CYP1A22014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Structure-activity relationships of diamine inhibitors of cytochrome P450 (CYP) 3A as novel pharmacoenhancers, part I: core region.
AID557279Ratio of EC50 for HIV1 JSL harboring L10I/L24I/L33F/E35D/M36I/N37S/M46L/I54V/R57K/I62V/L63P/A71V/G73S/82A in protease encoding region to EC50 for HIV1 ERS104pre2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID1443980Inhibition of human BSEP expressed in fall armyworm sf9 cell plasma membrane vesicles assessed as reduction in vesicle-associated [3H]-taurocholate transport preincubated for 10 mins prior to ATP addition measured after 15 mins in presence of [3H]-tauroch2010Toxicological sciences : an official journal of the Society of Toxicology, Dec, Volume: 118, Issue:2
Interference with bile salt export pump function is a susceptibility factor for human liver injury in drug development.
AID1295706Aqueous solubility of the compound by shake flask method2016Journal of medicinal chemistry, Apr-14, Volume: 59, Issue:7
Anti-HIV Drug Discovery and Development: Current Innovations and Future Trends.
AID529758Antimicrobial activity against Plasmodium vivax at trophozoite stage2008Antimicrobial agents and chemotherapy, Jul, Volume: 52, Issue:7
Stronger activity of human immunodeficiency virus type 1 protease inhibitors against clinical isolates of Plasmodium vivax than against those of P. falciparum.
AID374660Toxicity in human assessed as mortality at 100 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 102007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID543376Cmin in HIV-infected patient at 100 mg, qd2008Antimicrobial agents and chemotherapy, Sep, Volume: 52, Issue:9
Effects of minocycline and valproic acid coadministration on atazanavir plasma concentrations in human immunodeficiency virus-infected adults receiving atazanavir-ritonavir.
AID247984Inhibitory concentration against wild type human immunodeficiency virus2005Bioorganic & medicinal chemistry letters, May-02, Volume: 15, Issue:9
Oximinoarylsulfonamides as potent HIV protease inhibitors.
AID269305Antiviral activity against HIV1 LAI isolate in human PHA-PBMC cells2006Journal of medicinal chemistry, Aug-24, Volume: 49, Issue:17
Structure-based design of novel HIV-1 protease inhibitors to combat drug resistance.
AID588981Inhibitors of transporters of clinical importance in the absorption and disposition of drugs, OATP1B12010Nature reviews. Drug discovery, Mar, Volume: 9, Issue:3
Membrane transporters in drug development.
AID374650Decrease in steady state AUC (0 to 12 hrs) of voriconazole in human at 100 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 10 followed by 100 mg,2007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID557285Antiviral activity against HIV1 NL4-3 harboring L10I/G48V/I54V/L90M amino acid substitution in protease encoding region infected in human MT4 cells assessed as inhibition of p24 Gag protein production selected at 5 uM of saquinavir by ELISA2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID248603Inhibitory concentration of compound against HIV LAI in MT-4 cells when tested at a range of 0.001-10 uM concentration2004Journal of medicinal chemistry, Oct-07, Volume: 47, Issue:21
A 4'-C-ethynyl-2',3'-dideoxynucleoside analogue highlights the role of the 3'-OH in anti-HIV active 4'-C-ethynyl-2'-deoxy nucleosides.
AID557225Tmax in HIV-infected Thai pregnant women at 100 mg, po bid initiated intrapartum in combination with 400 mg, po bid Lopinavir measured within 72 hrs postpartum2009Antimicrobial agents and chemotherapy, May, Volume: 53, Issue:5
Early postpartum pharmacokinetics of lopinavir initiated intrapartum in Thai women.
AID82946Tested for inhibitor binding of wild-type HIV PR2004Journal of medicinal chemistry, Apr-08, Volume: 47, Issue:8
A phenylnorstatine inhibitor binding to HIV-1 protease: geometry, protonation, and subsite-pocket interactions analyzed at atomic resolution.
AID1594777Protection against AfB1-induced toxicity in human HepG2 cells assessed as increase in live cell area at >0.5 uM measured after 48 hrs by calcein-AM/Hoechst 33342 staining based high-content imaging analysis relative to control2019Journal of natural products, 06-28, Volume: 82, Issue:6
Natural-Product-Inspired Compounds as Countermeasures against the Liver Carcinogen Aflatoxin B
AID374656Toxicity in human assessed as mortality at 400 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 102007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID321693Ratio of EC50 for HIV1 mutant strain 1 to EC50 for wild type HIV12008Bioorganic & medicinal chemistry, Feb-01, Volume: 16, Issue:3
Structure-activity relationships of novel HIV-1 protease inhibitors containing the 3-amino-2-chlorobenzoyl-allophenylnorstatine structure.
AID721752Inhibition of human MATE2K-mediated ASP+ uptake expressed in HEK293 cells after 1.5 mins by fluorescence assay2013Journal of medicinal chemistry, Feb-14, Volume: 56, Issue:3
Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling.
AID1594774Protection against AfB1-induced toxicity in human HepG2 cells assessed as live cell area at 0.5 uM measured after 48 hrs by calcein-AM/Hoechst 33342 staining based high-content imaging analysis relative to control2019Journal of natural products, 06-28, Volume: 82, Issue:6
Natural-Product-Inspired Compounds as Countermeasures against the Liver Carcinogen Aflatoxin B
AID1267628Antiviral activity against nevirapine-resistant SIV mac251 infected in human MT4 cells2015Journal of medicinal chemistry, Dec-24, Volume: 58, Issue:24
Design and Synthesis of DiselenoBisBenzamides (DISeBAs) as Nucleocapsid Protein 7 (NCp7) Inhibitors with anti-HIV Activity.
AID1578456Fraction unbound in Wistar Han rat plasma at 2 uM incubated for 6 hrs by equilibrium dialysis method2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID575061Antiviral activity against Human immunodeficiency virus 1 harboring M46I, M46L, I54V, and V82A mutations in viral protease assessed as fold change in drug susceptibility relative to wild type2010Antimicrobial agents and chemotherapy, Nov, Volume: 54, Issue:11
Prevalence, mutation patterns, and effects on protease inhibitor susceptibility of the L76V mutation in HIV-1 protease.
AID625292Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) combined score2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID679070TP_TRANSPORTER: transepithelial transport of Ritonavir at a concentration of 0.1 uM in MDR1-expressing MDCK cells2003Pharmaceutical research, Aug, Volume: 20, Issue:8
Novel experimental parameters to quantify the modulation of absorptive and secretory transport of compounds by P-glycoprotein in cell culture models of intestinal epithelium.
AID624622Apparent permeability (Papp) from apical to basolateral side determined in MDR1-MDCKII cells2001The Journal of pharmacology and experimental therapeutics, Nov, Volume: 299, Issue:2
Rational use of in vitro P-glycoprotein assays in drug discovery.
AID1473740Inhibition of human MRP3 overexpressed in Sf9 insect cell membrane vesicles assessed as uptake of [3H]-estradiol-17beta-D-glucuronide in presence of ATP and GSH measured after 10 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID248602Inhibitory concentration of compound against HIV LAI in MT-2 cells when tested at a range of 0.001-10 uM concentration2004Journal of medicinal chemistry, Oct-07, Volume: 47, Issue:21
A 4'-C-ethynyl-2',3'-dideoxynucleoside analogue highlights the role of the 3'-OH in anti-HIV active 4'-C-ethynyl-2'-deoxy nucleosides.
AID1814286Binding affinity to recombinant Cryptococcus neoformans var. grubii H99 major aspartyl peptidase 1 assessed as inhibition constant2021Journal of medicinal chemistry, 05-27, Volume: 64, Issue:10
Re-emerging Aspartic Protease Targets: Examining
AID243422log (1/Km) value for human liver microsome cytochrome P450 3A42005Bioorganic & medicinal chemistry letters, Sep-15, Volume: 15, Issue:18
Modeling K(m) values using electrotopological state: substrates for cytochrome P450 3A4-mediated metabolism.
AID322110Antiviral activity against HIV1 GRL98065p30 in MT4 cells assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID408207Antiviral activity against HIV1 3B in human MT4 cells assessed as inhibition of viral-induced cytopathic effect by MTT method2008Journal of medicinal chemistry, May-22, Volume: 51, Issue:10
Combination of non-natural D-amino acid derivatives and allophenylnorstatine-dimethylthioproline scaffold in HIV protease inhibitors have high efficacy in mutant HIV.
AID699543Inhibition of human liver OATP1B3 expressed in HEK293 Flp-In cells assessed as reduction in [3H]E17-betaG uptake incubated for 5 mins by scintillation counting2012Journal of medicinal chemistry, May-24, Volume: 55, Issue:10
Classification of inhibitors of hepatic organic anion transporting polypeptides (OATPs): influence of protein expression on drug-drug interactions.
AID160767Association rate constant for the interaction between inhibitor and HIV-1 protease2002Journal of medicinal chemistry, Dec-05, Volume: 45, Issue:25
Relationships between structure and interaction kinetics for HIV-1 protease inhibitors.
AID374643Cmin in human at 100 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 10 followed by 100 mg, po of drug coadministered with voriconazole 200 mg, p2007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID1079949Proposed mechanism(s) of liver damage. [column 'MEC' in source]
AID1498796Inhibition of HIV1 3B protease infected in human MT4 cells assessed as protection from virus induced cytopathogenicity measured after 5 days post infection by MTT assay2018Bioorganic & medicinal chemistry letters, 08-01, Volume: 28, Issue:14
Design and synthesis of selenazole-substituted ritonavir analogs.
AID682151TP_TRANSPORTER: increase in plasma concentration in mdr1a(-/-) mouse2001The Journal of pharmacology and experimental therapeutics, Mar, Volume: 296, Issue:3
In vitro substrate identification studies for p-glycoprotein-mediated transport: species difference and predictability of in vivo results.
AID1069009Inhibition of human CYP2D62014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Structure-activity relationships of diamine inhibitors of cytochrome P450 (CYP) 3A as novel pharmacoenhancers, part I: core region.
AID1896146Binding affinity to NTCP (unknown origin)2022Journal of medicinal chemistry, 10-13, Volume: 65, Issue:19
Inhibiting Sodium Taurocholate Cotransporting Polypeptide in HBV-Related Diseases: From Biological Function to Therapeutic Potential.
AID558370Drug level in HIV-infected pregnant woman maternal blood plasma at 100 mg, po BID by HPLC/UV analysis2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Genital tract, cord blood, and amniotic fluid exposures of seven antiretroviral drugs during and after pregnancy in human immunodeficiency virus type 1-infected women.
AID625290Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for liver fatty2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID520315Inhibition of human CYP3A42008Antimicrobial agents and chemotherapy, May, Volume: 52, Issue:5
Rapid clinical induction of hepatic cytochrome P4502B6 activity by ritonavir.
AID415258AUC in dog at 5 mg/kg, po2009Journal of medicinal chemistry, Apr-23, Volume: 52, Issue:8
2-Pyridyl P1'-substituted symmetry-based human immunodeficiency virus protease inhibitors (A-792611 and A-790742) with potential for convenient dosing and reduced side effects.
AID322103Antiviral activity against ritonavir-resistant HIV1 in MT4 cells assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID588210Human drug-induced liver injury (DILI) modelling dataset from Ekins et al2010Drug metabolism and disposition: the biological fate of chemicals, Dec, Volume: 38, Issue:12
A predictive ligand-based Bayesian model for human drug-induced liver injury.
AID322108Antiviral activity against amprenavir-resistant HIV1 in MT4 cells assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID680122TP_TRANSPORTER: transepithelial transport (basal to apical) in MDR1-expressing LLC-PK1 cells2001Molecular pharmacology, Apr, Volume: 59, Issue:4
P-glycoprotein limits oral availability, brain, and fetal penetration of saquinavir even with high doses of ritonavir.
AID369838Antiviral activity against HIV1 NL4-3 infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect after 5 days by MTT assay2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro selection and characterization of human immunodeficiency virus type 2 with decreased susceptibility to lopinavir.
AID82749Fold increase in EC50 for antiviral activity against HIV Protease-Resistant strains with D protease amino acid substitutions2001Journal of medicinal chemistry, Jul-05, Volume: 44, Issue:14
4-Hydroxy-5,6-dihydropyrones as inhibitors of HIV protease: the effect of heterocyclic substituents at C-6 on antiviral potency and pharmacokinetic parameters.
AID322105Antiviral activity against nelfinavir-resistant HIV1 in MT4 cells assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID625287Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatomegaly2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID269310Antiviral activity against multi drug-resistant HIV1 ET variant in human PHA-PBMC cells2006Journal of medicinal chemistry, Aug-24, Volume: 49, Issue:17
Structure-based design of novel HIV-1 protease inhibitors to combat drug resistance.
AID625279Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for bilirubinemia2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID369945Ratio of EC50 for HIV2 CBL-23 infected in human PBMC to EC50 for HIV1 NL4-3 infected in human MT4 cells2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro selection and characterization of human immunodeficiency virus type 2 with decreased susceptibility to lopinavir.
AID321699Ratio of EC50 for HIV1 mutant strain 7 to EC50 for wild type HIV12008Bioorganic & medicinal chemistry, Feb-01, Volume: 16, Issue:3
Structure-activity relationships of novel HIV-1 protease inhibitors containing the 3-amino-2-chlorobenzoyl-allophenylnorstatine structure.
AID1219729Inhibition of CYP3A4-mediated lopinavir bioactivation in human liver microsomes assessed as inhibition of LPV_hydrazone_3 adduct formation at 2 uM after after 50 mins by UPLC-TOFMS analysis2012Drug metabolism and disposition: the biological fate of chemicals, Jan, Volume: 40, Issue:1
CYP3A4-mediated lopinavir bioactivation and its inhibition by ritonavir.
AID1578472Unbound tissue partition coefficient, ratio of drug level in skeletal muscle to plasma in Wistar Han rat at 1 to 2 mg/kg, iv infused for 10 to 20 hrs by LC-MS/MS analysis2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID269304Antiviral activity against HIV1 LAI isolate in human MT2 cells2006Journal of medicinal chemistry, Aug-24, Volume: 49, Issue:17
Structure-based design of novel HIV-1 protease inhibitors to combat drug resistance.
AID1079945Animal toxicity known. [column 'TOXIC' in source]
AID369951Antiviral activity against wild type HIV2 ROD infected in human CEM cells assessed as inhibition of virus production after 7 days by Lenti-RT activity assay2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro selection and characterization of human immunodeficiency virus type 2 with decreased susceptibility to lopinavir.
AID519608Selectivity index, ratio of TC50 for hepatocytes to IC50 for Plasmodium yoelii 2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
New active drugs against liver stages of Plasmodium predicted by molecular topology.
AID353754AUC in po dosed Beagle dog at dose molar equivalent to 5 mg/kg RTV dosed as 5% dextrose containing solution2009Journal of medicinal chemistry, May-14, Volume: 52, Issue:9
Water-soluble prodrugs of the human immunodeficiency virus protease inhibitors lopinavir and ritonavir.
AID1217729Intrinsic clearance for reactive metabolites formation assessed as summation of [3H]GSH adduct formation rate-based reactive metabolites formation and cytochrome P450 (unknown origin) inactivation rate-based reactive metabolites formation2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Combination of GSH trapping and time-dependent inhibition assays as a predictive method of drugs generating highly reactive metabolites.
AID1578461Fraction unbound in Wistar Han rat skeletal muscle at 2 uM incubated for 6 hrs by equilibrium dialysis method2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID353756AUC in po dosed Beagle dog at dose molar equivalent to 5 mg/kg RTV coadministered with LPV-OMP dosed as 5% dextrose containing solution2009Journal of medicinal chemistry, May-14, Volume: 52, Issue:9
Water-soluble prodrugs of the human immunodeficiency virus protease inhibitors lopinavir and ritonavir.
AID625276FDA Liver Toxicity Knowledge Base Benchmark Dataset (LTKB-BD) drugs of most concern for DILI2011Drug discovery today, Aug, Volume: 16, Issue:15-16
FDA-approved drug labeling for the study of drug-induced liver injury.
AID278957Antiviral activity against HIV1 A5 isolate with protease I84A mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID518051Toxicity in healthy human assessed as nausea at 100 mg/kg, po administered as single dose2010Antimicrobial agents and chemotherapy, Jun, Volume: 54, Issue:6
Effect of vicriviroc on the QT/corrected QT interval and central nervous system in healthy subjects.
AID372203Cmax in human at 400 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 102007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID525488Antigametocyte activity against Plasmodium falciparum harboring GFP-tagged Pfs16 protein assessed as reduction in number of gametocytes after 40 hrs by [3H]hypoxanthine incorporation assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
Antimalarial asexual stage-specific and gametocytocidal activities of HIV protease inhibitors.
AID589164Mechanism based inhibition of human cytochrome P450 3A4 using recombinant CYP3A42005Current drug metabolism, Oct, Volume: 6, Issue:5
Cytochrome p450 enzymes mechanism based inhibitors: common sub-structures and reactivity.
AID290999Antiviral activity against nelfinavir-resistant HIV1 NL4322007Bioorganic & medicinal chemistry letters, Aug-01, Volume: 17, Issue:15
Synthesis and antiviral property of allophenylnorstatine-based HIV protease inhibitors incorporating D-cysteine derivatives as P2/P3 moieties.
AID543378Cmin in HIV-infected patient at 100 mg, qd co-administered with 100 mg minocycline and 250 mg valporic acid, bid for 15 days2008Antimicrobial agents and chemotherapy, Sep, Volume: 52, Issue:9
Effects of minocycline and valproic acid coadministration on atazanavir plasma concentrations in human immunodeficiency virus-infected adults receiving atazanavir-ritonavir.
AID470604Cmax in Beagle dog at 5 mg/kg, po2009Bioorganic & medicinal chemistry letters, Sep-15, Volume: 19, Issue:18
Synthesis and evaluation of inhibitors of cytochrome P450 3A (CYP3A) for pharmacokinetic enhancement of drugs.
AID372209AUC (0 to 12 hrs) in human at 100 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 102007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID322102Antiviral activity against saquinavir-resistant HIV1 in MT4 cells assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID1287914Prodrug conversion in PBS assessed as half life for 50% release of ritonavir in pH 7.4 at 37 degC after 1 hr by ESI-mass spectrometry2016Bioorganic & medicinal chemistry letters, Apr-01, Volume: 26, Issue:7
Novel prodrugs with a spontaneous cleavable guanidine moiety.
AID278975Antiviral activity against HIV1 C8 isolate with protease I84C mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID1267611Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 3B2015Journal of medicinal chemistry, Dec-24, Volume: 58, Issue:24
Design and Synthesis of DiselenoBisBenzamides (DISeBAs) as Nucleocapsid Protein 7 (NCp7) Inhibitors with anti-HIV Activity.
AID1069008Inhibition of human CYP2C92014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Structure-activity relationships of diamine inhibitors of cytochrome P450 (CYP) 3A as novel pharmacoenhancers, part I: core region.
AID525114Antiviral activity against HIV 1 NL4.3 integrase S230N mutant infected in human MT-4 cells assessed as inhibition of virus-induced cytopathic effect by MTT assay2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Mutations in human immunodeficiency virus type 1 integrase confer resistance to the naphthyridine L-870,810 and cross-resistance to the clinical trial drug GS-9137.
AID391267Antiviral activity against multidrug-resistant HIV1 with protease L10I, L24I, I33F, E35D, M36I, N37S, M46L, I54V, R57K, I62V, L63P, A71V, G73S, V82A mutation in phytohemagglutininin-activated PBMC assessed as inhibition of p24G protein production2008Journal of medicinal chemistry, Oct-09, Volume: 51, Issue:19
Flexible cyclic ethers/polyethers as novel P2-ligands for HIV-1 protease inhibitors: design, synthesis, biological evaluation, and protein-ligand X-ray studies.
AID1079935Cytolytic liver toxicity, either proven histopathologically or where the ratio of maximal ALT or AST activity above normal to that of Alkaline Phosphatase is > 5 (see ACUTE). Value is number of references indexed. [column 'CYTOL' in source]
AID557296Ratio of EC50 for HIV1 NL4-3 harboring L10F/D30N/K45I/A71V/T74S amino acid substitution in protease encoding region to EC50 for HIV1 NL4-32009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID721750Inhibition of human OCT1-mediated ASP+ uptake expressed in HEK293 cells after 3 mins by fluorescence assay2013Journal of medicinal chemistry, Feb-14, Volume: 56, Issue:3
Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling.
AID278976Antiviral activity against HIV1 C9 isolate with protease I84C mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID588211Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in humans2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID248098Inhibitory concentration against resistant (A17) human immunodeficiency virus2005Bioorganic & medicinal chemistry letters, May-02, Volume: 15, Issue:9
Oximinoarylsulfonamides as potent HIV protease inhibitors.
AID54923Inhibition of human cytochrome P450 3A42003Journal of medicinal chemistry, Apr-24, Volume: 46, Issue:9
Comparison of in vitro P-glycoprotein screening assays: recommendations for their use in drug discovery.
AID557291Antiviral activity against HIV1 NL4-3 harboring L23I/K43I/M46I/I50L/G51A/A71V amino acid substitution in protease encoding region infected in human MT4 cells assessed as inhibition of p24 Gag protein production selected at 1 uM of atazanavir by ELISA2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID721746Inhibition of human MATE1-mediated [14]-metformin uptake expressed in polarized MDCK2 cells after 5 mins by liquid scintillation counting analysis2013Journal of medicinal chemistry, Feb-14, Volume: 56, Issue:3
Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling.
AID1219730Inhibition of CYP3A4-mediated lopinavir bioactivation in human liver microsomes assessed as inhibition of LPV_hydrazone_2 adduct formation at 2 uM after after 50 mins by UPLC-TOFMS analysis2012Drug metabolism and disposition: the biological fate of chemicals, Jan, Volume: 40, Issue:1
CYP3A4-mediated lopinavir bioactivation and its inhibition by ritonavir.
AID104301Anti-HIV-1 activity against mutant HIV-1 in MT-4 cells (mutation selected with compound 1)2001Journal of medicinal chemistry, Sep-13, Volume: 44, Issue:19
Design and synthesis of potent C(2)-symmetric diol-based HIV-1 protease inhibitors: effects of fluoro substitution.
AID278965Antiviral activity against HIV1 A13 isolate with protease I84A mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID278979Resistance to HIV1 with protease 46M, 54I and I84V mutation in HEK 293 cells relative to similar background2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID1218863Inhibition of OATP1B1 (unknown origin) expressed in HEK293 cells using pitavastatin substrate2012Drug metabolism and disposition: the biological fate of chemicals, Aug, Volume: 40, Issue:8
The development, characterization, and application of an OATP1B1 inhibition assay in drug discovery.
AID408219Ratio of EC50 for ritonavir and nelfinavir-resistant HIV1 NL432 to EC50 for wild-type HIV1 NL4322008Journal of medicinal chemistry, May-22, Volume: 51, Issue:10
Combination of non-natural D-amino acid derivatives and allophenylnorstatine-dimethylthioproline scaffold in HIV protease inhibitors have high efficacy in mutant HIV.
AID408208Cytotoxicity against human MT4 cells by MTT assay2008Journal of medicinal chemistry, May-22, Volume: 51, Issue:10
Combination of non-natural D-amino acid derivatives and allophenylnorstatine-dimethylthioproline scaffold in HIV protease inhibitors have high efficacy in mutant HIV.
AID321661Antiviral activity against HIV1 3B2008Bioorganic & medicinal chemistry, Feb-01, Volume: 16, Issue:3
Structure-activity relationships of novel HIV-1 protease inhibitors containing the 3-amino-2-chlorobenzoyl-allophenylnorstatine structure.
AID721754Inhibition of human MATE1-mediated ASP+ uptake expressed in HEK293 cells after 1.5 mins by fluorescence assay2013Journal of medicinal chemistry, Feb-14, Volume: 56, Issue:3
Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling.
AID1674183Inhibition of human BSEP expressed in HEK293 cell membrane vesicles assessed as reduction in 3H-TCA uptake incubated for 5 mins by radiodetection method2020Journal of medicinal chemistry, 10-22, Volume: 63, Issue:20
Drug Induced Liver Injury (DILI). Mechanisms and Medicinal Chemistry Avoidance/Mitigation Strategies.
AID721749Inhibition of human OCT3-mediated ASP+ uptake expressed in HEK293 cells after 3 mins by fluorescence assay2013Journal of medicinal chemistry, Feb-14, Volume: 56, Issue:3
Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling.
AID557298Ratio of EC50 for HIV1 NL4-3 harboring L10F/M46I/I54V/V82A amino acid substitution in protease encoding region to EC50 for HIV1 NL4-32009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID150752Inhibition of P-glycoprotein, human L-MDR1 expressed in LLC-PK1 epithelial cells using calcein-AM polarisation assay2003Journal of medicinal chemistry, Apr-24, Volume: 46, Issue:9
Comparison of in vitro P-glycoprotein screening assays: recommendations for their use in drug discovery.
AID372214Cmax in human at 400 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 10 followed by 400 mg, po of drug coadministered with voriconazole 200 mg, p2007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID444052Hepatic clearance in human2010Journal of medicinal chemistry, Feb-11, Volume: 53, Issue:3
Physicochemical space for optimum oral bioavailability: contribution of human intestinal absorption and first-pass elimination.
AID1220558Fraction unbound in Beagle dog brain homogenates at 1 uM after 6 hrs by equilibrium dialysis method2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Species independence in brain tissue binding using brain homogenates.
AID625288Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for jaundice2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1267612Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV-2 ROD2015Journal of medicinal chemistry, Dec-24, Volume: 58, Issue:24
Design and Synthesis of DiselenoBisBenzamides (DISeBAs) as Nucleocapsid Protein 7 (NCp7) Inhibitors with anti-HIV Activity.
AID625284Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatic failure2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID321690Antiviral activity against HIV1 mutant strain 52008Bioorganic & medicinal chemistry, Feb-01, Volume: 16, Issue:3
Structure-activity relationships of novel HIV-1 protease inhibitors containing the 3-amino-2-chlorobenzoyl-allophenylnorstatine structure.
AID1474150Ratio of drug concentration at steady state in human at 600 mg, po BID after 12 hrs to IC50 for human MRP3 overexpressed in Sf9 insect cells2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID88992Antiviral activity was assessed against M36I, I54V, A71V, V82T viral strains isolated from patient-129 pretreated with ritonavir(14 fold) at 8th week1998Bioorganic & medicinal chemistry letters, Dec-15, Volume: 8, Issue:24
Potent piperazine hydroxyethylamine HIV protease inhibitors containing novel P3 ligands.
AID1578466Drug concentration in total liver in Wistar Han rat at 1 to 2 mg/kg, iv infused for 10 to 20 hrs by LC-MS/MS analysis2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID278968Antiviral activity against HIV1 C1 isolate with protease I84C mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID1079934Highest frequency of acute liver toxicity observed during clinical trials, expressed as a percentage. [column '% AIGUE' in source]
AID1079938Chronic liver disease either proven histopathologically, or through a chonic elevation of serum amino-transferase activity after 6 months. Value is number of references indexed. [column 'CHRON' in source]
AID278962Antiviral activity against HIV1 A10 isolate with protease I84A mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID373237Half life in human at 100 mg, po bid for 10 days pretreated with paroxetin at 20 mg, po qd for 10 days followed by 16 days washout period then coadministered with paroxetin at 20 mg, po qd and fosamprenavir at 700 mg, po bid for 10 days2007Antimicrobial agents and chemotherapy, Nov, Volume: 51, Issue:11
Interaction study of the combined use of paroxetine and fosamprenavir-ritonavir in healthy subjects.
AID372205Tmax in human at 400 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 102007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID321697Ratio of EC50 for HIV1 mutant strain 5 to EC50 for wild type HIV12008Bioorganic & medicinal chemistry, Feb-01, Volume: 16, Issue:3
Structure-activity relationships of novel HIV-1 protease inhibitors containing the 3-amino-2-chlorobenzoyl-allophenylnorstatine structure.
AID374641AUC (0 to 12 hrs) in human at 100 mg, po bid from day 11 to day 20 pretreated with voriconazole 400 mg, po bid on day 1 and 200 mg, po bid on day 2 and 3 followed by washout from day 4 to day 10 followed by 100 mg, po of drug coadministered with voriconaz2007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID1474148Drug concentration at steady state in human at 600 mg, po BID after 12 hrs2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID721739Selectivity ratio of IC50 for human OCT3-mediated ASP+ uptake expressed in HEK293 cells to IC50 for human MATE1-mediated ASP+ uptake expressed in HEK293 cells2013Journal of medicinal chemistry, Feb-14, Volume: 56, Issue:3
Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling.
AID518042Cardiotoxicity in healthy human assessed as change in corrected QT interval at 100 mg/kg, po after 10 days by ECG2010Antimicrobial agents and chemotherapy, Jun, Volume: 54, Issue:6
Effect of vicriviroc on the QT/corrected QT interval and central nervous system in healthy subjects.
AID681518TP_TRANSPORTER: Western, efflux of Carboxyfluorescein in LS-180V2001Journal of pharmaceutical sciences, Nov, Volume: 90, Issue:11
Ritonavir induces P-glycoprotein expression, multidrug resistance-associated protein (MRP1) expression, and drug transporter-mediated activity in a human intestinal cell line.
AID249499Toxicity value against wild type HIV-1 IIIB was determined2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Design of HIV-1 protease inhibitors active on multidrug-resistant virus.
AID1217716Time dependent inhibition of CYP2C8 (unknown origin) at 10 uM by LC/MS system2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Combination of GSH trapping and time-dependent inhibition assays as a predictive method of drugs generating highly reactive metabolites.
AID572572Cmax in HIV-1 infected patient at 100 mg, po bid coadministered with 400 mg, po bid of Lopinavir and 300 mg, po qd of atazanavir2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Decrease of atazanavir and lopinavir plasma concentrations in a boosted double human immunodeficiency virus protease inhibitor salvage regimen.
AID1217719Time dependent inhibition of CYP2C9 (unknown origin) at 3 uM by LC/MS system2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Combination of GSH trapping and time-dependent inhibition assays as a predictive method of drugs generating highly reactive metabolites.
AID1636356Drug activation in human Hep3B cells assessed as human CYP2C9-mediated drug metabolism-induced cytotoxicity measured as decrease in cell viability at 300 uM pre-incubated with BSO for 18 hrs followed by incubation with compound for 3 hrs in presence of NA2016Bioorganic & medicinal chemistry letters, 08-15, Volume: 26, Issue:16
Development of a cell viability assay to assess drug metabolite structure-toxicity relationships.
AID369172Cmax in HIV1 infected human at 100 mg, po administered twice daily2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
Association of saquinavir plasma concentrations with side effects but not with antiretroviral outcome in patients infected with protease inhibitor-susceptible human immunodeficiency virus type 1.
AID543388Half life in HIV-infected patient at 100 mg, qd2008Antimicrobial agents and chemotherapy, Sep, Volume: 52, Issue:9
Effects of minocycline and valproic acid coadministration on atazanavir plasma concentrations in human immunodeficiency virus-infected adults receiving atazanavir-ritonavir.
AID372208Tmax in human at 400 mg, po bid from day 11 to day 20 pretreated with placebo from day 1 to day 3 followed by washout from day 4 to day 102007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID1182731Inhibition of HTLV-1 protease2014Journal of medicinal chemistry, Jul-24, Volume: 57, Issue:14
Structural basis for HTLV-1 protease inhibition by the HIV-1 protease inhibitor indinavir.
AID588986Inhibitors of transporters of clinical importance in the absorption and disposition of drugs, OATP1A22010Nature reviews. Drug discovery, Mar, Volume: 9, Issue:3
Membrane transporters in drug development.
AID278958Antiviral activity against HIV1 A6 isolate with protease I84A mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID1221958Efflux ratio of permeability from apical to basolateral side over basolateral to apical side of human Caco2 cells at 10 uM up to 120 mins by HPLC-MC analysis2011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Attenuation of intestinal absorption by major efflux transporters: quantitative tools and strategies using a Caco-2 model.
AID321686Antiviral activity against HIV1 mutant strain 12008Bioorganic & medicinal chemistry, Feb-01, Volume: 16, Issue:3
Structure-activity relationships of novel HIV-1 protease inhibitors containing the 3-amino-2-chlorobenzoyl-allophenylnorstatine structure.
AID525106Antiviral activity against HIV 1 3B harboring integrase E92Q, S230N and L34M triple mutant infected in MT-4 cells assessed as inhibition of virus-induced cytopathic effect by MTT assay selected after 60 passages in presence of compound2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Mutations in human immunodeficiency virus type 1 integrase confer resistance to the naphthyridine L-870,810 and cross-resistance to the clinical trial drug GS-9137.
AID681128TP_TRANSPORTER: inhibition of Calcein-AM efflux in Mdr1b-expressing LLC-PK1 cells2003Journal of medicinal chemistry, Apr-24, Volume: 46, Issue:9
Comparison of in vitro P-glycoprotein screening assays: recommendations for their use in drug discovery.
AID269312Antiviral activity against multi drug-resistant HIV1 C variant in human PHA-PBMC cells2006Journal of medicinal chemistry, Aug-24, Volume: 49, Issue:17
Structure-based design of novel HIV-1 protease inhibitors to combat drug resistance.
AID680917TP_TRANSPORTER: Western, efflux of Rhodamine 123 in LS-180V2001Journal of pharmaceutical sciences, Nov, Volume: 90, Issue:11
Ritonavir induces P-glycoprotein expression, multidrug resistance-associated protein (MRP1) expression, and drug transporter-mediated activity in a human intestinal cell line.
AID404304Effect on human MRP2-mediated estradiol-17-beta-glucuronide transport in Sf9 cells inverted membrane vesicles relative to control2008Journal of medicinal chemistry, Jun-12, Volume: 51, Issue:11
Prediction and identification of drug interactions with the human ATP-binding cassette transporter multidrug-resistance associated protein 2 (MRP2; ABCC2).
AID519574Antiviral activity against HIV1 P21 infected in human MT4 cells derived from viral passages with A-790742 harboring protease L23I, L33F, K45I, A71A/V, V77I, V82L, and I84V mutation assessed as reduction in viral cytopathogenicity treated 1 hr post infecti2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
Characterization of a novel human immunodeficiency virus type 1 protease inhibitor, A-790742.
AID1578464Drug concentration in total brain in Wistar Han rat at 1 to 2 mg/kg, iv infused for 10 to 20 hrs by LC-MS/MS analysis2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID572587Apparent oral clearance in HIV-1 infected patient at 100 mg, po bid coadministered with 300 mg, po qd of atazanavir and NRTI2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Decrease of atazanavir and lopinavir plasma concentrations in a boosted double human immunodeficiency virus protease inhibitor salvage regimen.
AID681295TP_TRANSPORTER: inhibition of Calcein efflux (Calcein: 0.05 micro;M, Ritonavir: 147 uM, increase retention of calcein (34%->83% retained)) in UMCC-1/VP cells2002AIDS (London, England), Sep-06, Volume: 16, Issue:13
The protease inhibitor ritonavir inhibits the functional activity of the multidrug resistance related-protein 1 (MRP-1).
AID104302Anti-HIV-1 activity against mutant HIV-1 in MT-4 cells (mutation selected with nelfinavir)2001Journal of medicinal chemistry, Sep-13, Volume: 44, Issue:19
Design and synthesis of potent C(2)-symmetric diol-based HIV-1 protease inhibitors: effects of fluoro substitution.
AID290992Inhibition of HIV protease at 1 nM2007Bioorganic & medicinal chemistry letters, Aug-01, Volume: 17, Issue:15
Synthesis and antiviral property of allophenylnorstatine-based HIV protease inhibitors incorporating D-cysteine derivatives as P2/P3 moieties.
AID540233Dose normalised AUC in human after po administration2005Xenobiotica; the fate of foreign compounds in biological systems, Feb, Volume: 35, Issue:2
Comparative evaluation of oral systemic exposure of 56 xenobiotics in rat, dog, monkey and human.
AID417047Inhibition of esterase mediated-hydrolysis of tenofovir disoproxil fumarate in human intestinal sub cellular fraction S9 at 2 uM after 30 mins2007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Effects of human immunodeficiency virus protease inhibitors on the intestinal absorption of tenofovir disoproxil fumarate in vitro.
AID235098Resistance of constructed mutant ABT538 virus was calculated as (IC90 for a mutant virus / IC90 of wt HXB2 virus)1997Journal of medicinal chemistry, Jan-17, Volume: 40, Issue:2
Cyclic urea amides: HIV-1 protease inhibitors with low nanomolar potency against both wild type and protease inhibitor resistant mutants of HIV.
AID1578471Unbound tissue partition coefficient, ratio of drug level in liver to plasma in Wistar Han rat at 1 to 2 mg/kg, iv infused for 10 to 20 hrs by LC-MS/MS analysis2020European journal of medicinal chemistry, Jan-01, Volume: 185Structural attributes influencing unbound tissue distribution.
AID1215348Time dependent inhibition of CYP3A4 in human liver microsomes assessed as conversion of testosterone to 6beta-hydroxytestosterone at 0.01 to 100 uM preincubated for 60 mins followed by testosterone treatment measured after 10 mins by LC-MS/MS analysis2011Drug metabolism and disposition: the biological fate of chemicals, Jun, Volume: 39, Issue:6
A refined cytochrome P540 IC₅₀ shift assay for reliably identifying CYP3A time-dependent inhibitors.
AID1079937Severe hepatitis, defined as possibly life-threatening liver failure or through clinical observations. Value is number of references indexed. [column 'MASS' in source]
AID82295Binding affinity for HIV -1 Protease1999Bioorganic & medicinal chemistry letters, Nov-15, Volume: 9, Issue:22
Unsymmetrical cyclic ureas as HIV-1 protease inhibitors: novel biaryl indazoles as P2/P2' substituents.
AID278955Antiviral activity against HIV1 A3 isolate with protease I84A mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID246225Protease inhibitory activity against HIV-1 GSS004421 mutant strain was determined2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Design of HIV-1 protease inhibitors active on multidrug-resistant virus.
AID1674184Toxicity in po dosed human assessed as maximum daily dose2020Journal of medicinal chemistry, 10-22, Volume: 63, Issue:20
Drug Induced Liver Injury (DILI). Mechanisms and Medicinal Chemistry Avoidance/Mitigation Strategies.
AID269315Antiviral activity against multi drug-resistant HIV1 EV variant in human PHA-PBMC cells2006Journal of medicinal chemistry, Aug-24, Volume: 49, Issue:17
Structure-based design of novel HIV-1 protease inhibitors to combat drug resistance.
AID369955Ratio of EC50 for HIV2 ROD with protease G17N mutation to EC50 for wild type HIV2 ROD2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro selection and characterization of human immunodeficiency virus type 2 with decreased susceptibility to lopinavir.
AID518052Toxicity in healthy human assessed as diarrhea at 100 mg/kg, po administered as single dose2010Antimicrobial agents and chemotherapy, Jun, Volume: 54, Issue:6
Effect of vicriviroc on the QT/corrected QT interval and central nervous system in healthy subjects.
AID322097Antiviral activity against HIV2 EHO in MT2 cells assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID278959Antiviral activity against HIV1 A7 isolate with protease I84A mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID297668Inhibition of HIV1 Protease M1 variant by FRET based assay2007Journal of medicinal chemistry, Sep-06, Volume: 50, Issue:18
Design and synthesis of HIV-1 protease inhibitors incorporating oxazolidinones as P2/P2' ligands in pseudosymmetric dipeptide isosteres.
AID1219731Inhibition of CYP3A4-mediated lopinavir bioactivation in human liver microsomes assessed as inhibition of LPV_hydrazone_1 adduct formation at 2 uM after after 50 mins by UPLC-TOFMS analysis2012Drug metabolism and disposition: the biological fate of chemicals, Jan, Volume: 40, Issue:1
CYP3A4-mediated lopinavir bioactivation and its inhibition by ritonavir.
AID269306Antiviral activity against HIV1 BA-L isolate in human PHA-PBMC cells2006Journal of medicinal chemistry, Aug-24, Volume: 49, Issue:17
Structure-based design of novel HIV-1 protease inhibitors to combat drug resistance.
AID518054Toxicity in healthy human assessed as dizziness at 100 mg/kg, po administered as single dose2010Antimicrobial agents and chemotherapy, Jun, Volume: 54, Issue:6
Effect of vicriviroc on the QT/corrected QT interval and central nervous system in healthy subjects.
AID263209Antiviral activity against HIV D545701 in MT4 cells2006Bioorganic & medicinal chemistry letters, Apr-01, Volume: 16, Issue:7
Ultra-potent P1 modified arylsulfonamide HIV protease inhibitors: the discovery of GW0385.
AID557289Antiviral activity against HIV1 NL4-3 harboring M46I/V82F/I84V amino acid substitution in protease encoding region infected in human MT4 cells assessed as inhibition of p24 Gag protein production selected at 5 uM of ritonavir by ELISA2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID82747Fold increase in EC50 for antiviral activity against HIV Protease-Resistant strains with B protease amino acid substitutions2001Journal of medicinal chemistry, Jul-05, Volume: 44, Issue:14
4-Hydroxy-5,6-dihydropyrones as inhibitors of HIV protease: the effect of heterocyclic substituents at C-6 on antiviral potency and pharmacokinetic parameters.
AID246194Protease inhibitory activity against HIV-1 r13034 mutant strain was determined2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Design of HIV-1 protease inhibitors active on multidrug-resistant virus.
AID278980Resistance to HIV1 with protease 46M, 54I and I84C mutation in HEK 293 cells relative to similar background2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID105395Anti-HIV activity was determined in MT-4 cells in the presence of 0% human serum using cytopathic effect assay1998Bioorganic & medicinal chemistry letters, Dec-15, Volume: 8, Issue:24
Potent piperazine hydroxyethylamine HIV protease inhibitors containing novel P3 ligands.
AID1219733Inhibition of CYP3A4-mediated lopinavir bioactivation in human liver microsomes assessed as inhibition of lopinavir semicarbazide DMP adduct formation adduct formation at 2 uM after after 50 mins by UPLC-TOFMS analysis2012Drug metabolism and disposition: the biological fate of chemicals, Jan, Volume: 40, Issue:1
CYP3A4-mediated lopinavir bioactivation and its inhibition by ritonavir.
AID557236Drug level in HIV-infected Thai pregnant women serum at 100 mg, po bid initiated intrapartum administered for 30 days in combination with 400 mg, po bid Lopinavir measured 4 hrs post last dose2009Antimicrobial agents and chemotherapy, May, Volume: 53, Issue:5
Early postpartum pharmacokinetics of lopinavir initiated intrapartum in Thai women.
AID278964Antiviral activity against HIV1 A12 isolate with protease I84A mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID290995Cytotoxicity against MT4 cells by MTT assay2007Bioorganic & medicinal chemistry letters, Aug-01, Volume: 17, Issue:15
Synthesis and antiviral property of allophenylnorstatine-based HIV protease inhibitors incorporating D-cysteine derivatives as P2/P3 moieties.
AID321695Ratio of EC50 for HIV1 mutant strain 3 to EC50 for wild type HIV12008Bioorganic & medicinal chemistry, Feb-01, Volume: 16, Issue:3
Structure-activity relationships of novel HIV-1 protease inhibitors containing the 3-amino-2-chlorobenzoyl-allophenylnorstatine structure.
AID681610TP_TRANSPORTER: increase in brain concentration in mdr1a(-/-) mouse2001The Journal of pharmacology and experimental therapeutics, Mar, Volume: 296, Issue:3
In vitro substrate identification studies for p-glycoprotein-mediated transport: species difference and predictability of in vivo results.
AID225563Dissociation constant obtained by inhibition of mutant HIV-protease (K-60)2000Journal of medicinal chemistry, Sep-07, Volume: 43, Issue:18
Identification of MK-944a: a second clinical candidate from the hydroxylaminepentanamide isostere series of HIV protease inhibitors.
AID54752Antiviral activity as inhibition of human liver microsomal Cytochrome P450 3A41998Bioorganic & medicinal chemistry letters, Dec-15, Volume: 8, Issue:24
Potent piperazine hydroxyethylamine HIV protease inhibitors containing novel P3 ligands.
AID1215349Time dependent inhibition of CYP3A4 in human liver microsomes assessed as conversion of testosterone to 6beta-hydroxytestosterone at 0.01 to 100 uM preincubated for 60 mins followed by testosterone treatment measured after 10 mins by refined CYP450 IC50 s2011Drug metabolism and disposition: the biological fate of chemicals, Jun, Volume: 39, Issue:6
A refined cytochrome P540 IC₅₀ shift assay for reliably identifying CYP3A time-dependent inhibitors.
AID557234Drug level in HIV-infected Thai pregnant women serum at 100 mg, po bid initiated intrapartum administered for 30 days in combination with 400 mg, po bid Lopinavir measured before administering last dose2009Antimicrobial agents and chemotherapy, May, Volume: 53, Issue:5
Early postpartum pharmacokinetics of lopinavir initiated intrapartum in Thai women.
AID625289Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for liver disease2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID88994Antiviral activity was assessed against M36M/I, I54I/V, V82F/A/S/T viral strains isolated from patient-313 pretreated with ritonavir(14 fold) at 85th day1998Bioorganic & medicinal chemistry letters, Dec-15, Volume: 8, Issue:24
Potent piperazine hydroxyethylamine HIV protease inhibitors containing novel P3 ligands.
AID30209Plasma half life at the dose of 5 mg/kg iv.2000Journal of medicinal chemistry, Feb-10, Volume: 43, Issue:3
Protease inhibitors: current status and future prospects.
AID373235Cmax in human at 100 mg, po bid for 10 days pretreated with paroxetin at 20 mg, po qd for 10 days followed by 16 days washout period then coadministered with paroxetin at 20 mg, po qd and fosamprenavir at 700 mg, po bid for 10 days2007Antimicrobial agents and chemotherapy, Nov, Volume: 51, Issue:11
Interaction study of the combined use of paroxetine and fosamprenavir-ritonavir in healthy subjects.
AID558377Drug level in HIV-infected pregnant woman cord blood plasma at 100 mg, po BID by HPLC/UV analysis2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Genital tract, cord blood, and amniotic fluid exposures of seven antiretroviral drugs during and after pregnancy in human immunodeficiency virus type 1-infected women.
AID1443992Total Cmax in human administered as single dose2014Hepatology (Baltimore, Md.), Sep, Volume: 60, Issue:3
Human drug-induced liver injury severity is highly associated with dual inhibition of liver mitochondrial function and bile salt export pump.
AID624628Drug-stimulated Pgp ATPase activity ratio determined in MDR1-Sf9 cell membranes with test compound at a concentration of 20uM2001The Journal of pharmacology and experimental therapeutics, Nov, Volume: 299, Issue:2
Rational use of in vitro P-glycoprotein assays in drug discovery.
AID1215347Time dependent inhibition of CYP3A4 in human liver microsomes assessed as conversion of testosterone to 6beta-hydroxytestosterone at 0.01 to 100 uM preincubated for 60 mins followed by testosterone treatment measured after 10 mins by LC-MS/MS analysis in 2011Drug metabolism and disposition: the biological fate of chemicals, Jun, Volume: 39, Issue:6
A refined cytochrome P540 IC₅₀ shift assay for reliably identifying CYP3A time-dependent inhibitors.
AID163487Inhibitory activity against HIV-1 protease1996Journal of medicinal chemistry, Oct-11, Volume: 39, Issue:21
HIV protease inhibitory bis-benzamide cyclic ureas: a quantitative structure-activity relationship analysis.
AID365484Antiviral activity against HIV1 3B in MT4 cells assessed as virus p24 antigen production administered at 16 hrs post infection by ELISA2008Journal of medicinal chemistry, Sep-11, Volume: 51, Issue:17
Structure-activity relationship study on anti-HIV 6-desfluoroquinolones.
AID269317Antiviral activity against multi drug-resistant HIV1 K variant in human PHA-PBMC cells2006Journal of medicinal chemistry, Aug-24, Volume: 49, Issue:17
Structure-based design of novel HIV-1 protease inhibitors to combat drug resistance.
AID246160Protease inhibitory activity against wild type HIV-1 IIIB was determined2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Design of HIV-1 protease inhibitors active on multidrug-resistant virus.
AID543377Cmin in HIV-infected patient at 100 mg, qd co-administered with 100 mg minocycline, bid for 15 days2008Antimicrobial agents and chemotherapy, Sep, Volume: 52, Issue:9
Effects of minocycline and valproic acid coadministration on atazanavir plasma concentrations in human immunodeficiency virus-infected adults receiving atazanavir-ritonavir.
AID1474147AUC in human at 600 mg, po BID after 12 hrs2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID525105Antiviral activity against HIV 1 3B harboring integrase L34M mutant infected in MT-4 cells assessed as inhibition of virus-induced cytopathic effect by MTT assay selected after 40 passages in presence of compound2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Mutations in human immunodeficiency virus type 1 integrase confer resistance to the naphthyridine L-870,810 and cross-resistance to the clinical trial drug GS-9137.
AID162710Affinity against HIV protease1997Journal of medicinal chemistry, Dec-05, Volume: 40, Issue:25
Nonsymmetrically substituted cyclic urea HIV protease inhibitors.
AID278971Antiviral activity against HIV1 C4 isolate with protease I84C mutation in HEK 293 cells relative to wild type2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID321662Antiviral activity against HIV1 3B in presence of 50% human serum2008Bioorganic & medicinal chemistry, Feb-01, Volume: 16, Issue:3
Structure-activity relationships of novel HIV-1 protease inhibitors containing the 3-amino-2-chlorobenzoyl-allophenylnorstatine structure.
AID1221967Ratio of intestine AUC in po dosed mdr1 knock out mouse to intestine AUC in po dosed wild type mouse2011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Attenuation of intestinal absorption by major efflux transporters: quantitative tools and strategies using a Caco-2 model.
AID290997Antiviral activity against HIV1 NL4322007Bioorganic & medicinal chemistry letters, Aug-01, Volume: 17, Issue:15
Synthesis and antiviral property of allophenylnorstatine-based HIV protease inhibitors incorporating D-cysteine derivatives as P2/P3 moieties.
AID1473741Inhibition of human MRP4 overexpressed in Sf9 cell membrane vesicles assessed as uptake of [3H]-estradiol-17beta-D-glucuronide in presence of ATP and GSH measured after 20 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID557287Antiviral activity against HIV1 NL4-3 harboring L10F/L24I/M46I/L63P/A71V/G73S/V82T amino acid substitution in protease encoding region infected in human MT4 cells assessed as inhibition of p24 Gag protein production selected at 5 uM of indinavir by ELISA2009Antimicrobial agents and chemotherapy, Mar, Volume: 53, Issue:3
GRL-02031, a novel nonpeptidic protease inhibitor (PI) containing a stereochemically defined fused cyclopentanyltetrahydrofuran potent against multi-PI-resistant human immunodeficiency virus type 1 in vitro.
AID278817Antimalarial activity against Plasmodium falciparum FAC82007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Synergistic interactions of the antiretroviral protease inhibitors saquinavir and ritonavir with chloroquine and mefloquine against Plasmodium falciparum in vitro.
AID519607Cytotoxicity against Hepatocyte cells assessed as cell viability by MTT assay2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
New active drugs against liver stages of Plasmodium predicted by molecular topology.
AID415267AUC in dog at 2.5 mg/kg2009Journal of medicinal chemistry, Apr-23, Volume: 52, Issue:8
2-Pyridyl P1'-substituted symmetry-based human immunodeficiency virus protease inhibitors (A-792611 and A-790742) with potential for convenient dosing and reduced side effects.
AID322122Antiviral activity against HIV1 97ZA003 R5 subtype C in phytohemagglutininin-activated PBMCs assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID160481Inhibitory concentration against HIV-1 protease2001Journal of medicinal chemistry, Sep-13, Volume: 44, Issue:19
Design and synthesis of potent C(2)-symmetric diol-based HIV-1 protease inhibitors: effects of fluoro substitution.
AID369956Ratio of EC50 for HIV2 ROD with protease V47A mutation to EC50 for wild type HIV2 ROD2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro selection and characterization of human immunodeficiency virus type 2 with decreased susceptibility to lopinavir.
AID543370AUC (0 to 24 hrs) in HIV-infected patient at 100 mg, qd2008Antimicrobial agents and chemotherapy, Sep, Volume: 52, Issue:9
Effects of minocycline and valproic acid coadministration on atazanavir plasma concentrations in human immunodeficiency virus-infected adults receiving atazanavir-ritonavir.
AID278983Resistance to HIV1 with protease 46I/L, 54I and I84C mutation in HEK 293 cells relative to similar background2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID408209Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 3B2008Journal of medicinal chemistry, May-22, Volume: 51, Issue:10
Combination of non-natural D-amino acid derivatives and allophenylnorstatine-dimethylthioproline scaffold in HIV protease inhibitors have high efficacy in mutant HIV.
AID543390Half life in HIV-infected patient at 100 mg, qd co-administered with 100 mg minocycline and 250 mg valporic acid, bid for 15 days2008Antimicrobial agents and chemotherapy, Sep, Volume: 52, Issue:9
Effects of minocycline and valproic acid coadministration on atazanavir plasma concentrations in human immunodeficiency virus-infected adults receiving atazanavir-ritonavir.
AID1220557Fraction unbound in Hartley guinea pig brain homogenates at 1 uM after 6 hrs by equilibrium dialysis method2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Species independence in brain tissue binding using brain homogenates.
AID278816Antimalarial activity against Plasmodium falciparum D102007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Synergistic interactions of the antiretroviral protease inhibitors saquinavir and ritonavir with chloroquine and mefloquine against Plasmodium falciparum in vitro.
AID1217711Metabolic activation in human liver microsomes assessed as [3H]GSH adduct formation rate measured per mg of protein at 100 uM by [3H]GSH trapping assay2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Combination of GSH trapping and time-dependent inhibition assays as a predictive method of drugs generating highly reactive metabolites.
AID322114Antiviral activity against HIV1 MDR/MM R5 in phytohemagglutininin-activated PBMCs assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID427587Inhibition of SAP1-dependent growth in Candida albicans SAP2MS4B expressing SAP1ex4A at 100 uM incubated in YCB-BSA medium2008Antimicrobial agents and chemotherapy, Jan, Volume: 52, Issue:1
Tetracycline-inducible expression of individual secreted aspartic proteases in Candida albicans allows isoenzyme-specific inhibitor screening.
AID278985Resistance to HIV1 with protease 46I, 54M/V and I84A mutation in HEK 293 relative to similar background2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID237358Partition coefficient in octanol/water2005Bioorganic & medicinal chemistry letters, Aug-15, Volume: 15, Issue:16
Hydrophobicity in the design of P2/P2' tetrahydropyrimidinone HIV protease inhibitors.
AID297670Inhibition of HIV1 Protease M3 variant by FRET based assay2007Journal of medicinal chemistry, Sep-06, Volume: 50, Issue:18
Design and synthesis of HIV-1 protease inhibitors incorporating oxazolidinones as P2/P2' ligands in pseudosymmetric dipeptide isosteres.
AID82294Inhibitory activity against HIV -1 Protease in whole cell assay1999Bioorganic & medicinal chemistry letters, Nov-15, Volume: 9, Issue:22
Unsymmetrical cyclic ureas as HIV-1 protease inhibitors: novel biaryl indazoles as P2/P2' substituents.
AID1889294Amorphous solubility of the compound in phosphate buffer at pH 6.8 by UV spectrometer2022Journal of medicinal chemistry, 02-10, Volume: 65, Issue:3
Dose Number as a Tool to Guide Lead Optimization for Orally Bioavailable Compounds in Drug Discovery.
AID374645AUC (0 to 12 hrs) in human at 400 mg, po bid from day 11 to day 20 pretreated with placebo from day 1 to day 3 followed by washout from day 4 to day 10 followed by 400 mg, po of drug coadministered with placebo from day 21 to day 302007Antimicrobial agents and chemotherapy, Oct, Volume: 51, Issue:10
Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects.
AID699544Inhibition of human liver OATP2B1 expressed in HEK293 Flp-In cells assessed as reduction in [3H]E3S uptake incubated for 5 mins by scintillation counting2012Journal of medicinal chemistry, May-24, Volume: 55, Issue:10
Classification of inhibitors of hepatic organic anion transporting polypeptides (OATPs): influence of protein expression on drug-drug interactions.
AID427585Inhibition of SAP3-dependent growth in Candida albicans SAP2MS4B expressing SAP3ex4A at 100 uM incubated in YCB-BSA medium2008Antimicrobial agents and chemotherapy, Jan, Volume: 52, Issue:1
Tetracycline-inducible expression of individual secreted aspartic proteases in Candida albicans allows isoenzyme-specific inhibitor screening.
AID246965Effective dose of compound required to inhibit replication of human immunodeficiency virus type 1 in MT-4 cells2004Journal of medicinal chemistry, Nov-18, Volume: 47, Issue:24
Improved structure-activity relationship analysis of HIV-1 protease inhibitors using interaction kinetic data.
AID322117Antiviral activity against HIV1 MDR/C X4 in phytohemagglutininin-activated PBMCs assessed as inhibition of p24 Gag protein expression by MTT assay2007Antimicrobial agents and chemotherapy, Jun, Volume: 51, Issue:6
A novel bis-tetrahydrofuranylurethane-containing nonpeptidic protease inhibitor (PI), GRL-98065, is potent against multiple-PI-resistant human immunodeficiency virus in vitro.
AID1217704Time dependent inhibition of CYP1A2 (unknown origin) at 100 uM by LC/MS system2011Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 39, Issue:7
Combination of GSH trapping and time-dependent inhibition assays as a predictive method of drugs generating highly reactive metabolites.
AID1221979Transporter substrate index ratio of permeability from basolateral to apical side in human Caco2 cells at 10 uM up to 120 mins by HPLC-MC analysis in presence of 10 uM of MRP2 inhibitor MK5712011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Attenuation of intestinal absorption by major efflux transporters: quantitative tools and strategies using a Caco-2 model.
AID408218Ratio of EC50 for nelfinavir-resistant HIV1 NL432 to EC50 for wild-type HIV1 NL4322008Journal of medicinal chemistry, May-22, Volume: 51, Issue:10
Combination of non-natural D-amino acid derivatives and allophenylnorstatine-dimethylthioproline scaffold in HIV protease inhibitors have high efficacy in mutant HIV.
AID1221960Apparent permeability from apical to basolateral side of human Caco2 cells at 10 uM up to 120 mins by HPLC-MC analysis in presence of 1 uM of P-gp inhibitor LY3359792011Drug metabolism and disposition: the biological fate of chemicals, Feb, Volume: 39, Issue:2
Attenuation of intestinal absorption by major efflux transporters: quantitative tools and strategies using a Caco-2 model.
AID1069065Antiviral activity against HIV1 by cell based assay2014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Structure-activity relationships of diamine inhibitors of cytochrome P450 (CYP) 3A as novel pharmacoenhancers. Part II: P2/P3 region and discovery of cobicistat (GS-9350).
AID242866Dissociation rate constant for human immunodeficiency virus type 1 protease2004Journal of medicinal chemistry, Nov-18, Volume: 47, Issue:24
Improved structure-activity relationship analysis of HIV-1 protease inhibitors using interaction kinetic data.
AID391266Antiviral activity against multidrug-resistant HIV1 with protease L10I, K43T, M46L, I54V, L63P, A71V, V82A, L90M, Q92K mutation in phytohemagglutininin-activated PBMC assessed as inhibition of p24G protein production2008Journal of medicinal chemistry, Oct-09, Volume: 51, Issue:19
Flexible cyclic ethers/polyethers as novel P2-ligands for HIV-1 protease inhibitors: design, synthesis, biological evaluation, and protein-ligand X-ray studies.
AID1079946Presence of at least one case with successful reintroduction. [column 'REINT' in source]
AID82746Fold increase in EC50 for antiviral activity against HIV Protease-Resistant strains with A protease amino acid substitutions2001Journal of medicinal chemistry, Jul-05, Volume: 44, Issue:14
4-Hydroxy-5,6-dihydropyrones as inhibitors of HIV protease: the effect of heterocyclic substituents at C-6 on antiviral potency and pharmacokinetic parameters.
AID278984Resistance to HIV1 with protease 46I, 54V and I84V mutation in HEK 293 cells relative to similar background2007Antimicrobial agents and chemotherapy, Feb, Volume: 51, Issue:2
Identification and structural characterization of I84C and I84A mutations that are associated with high-level resistance to human immunodeficiency virus protease inhibitors and impair viral replication.
AID1474149Ratio of drug concentration at steady state in human at 600 mg, po BID after 12 hrs to IC50 for human BSEP overexpressed in Sf9 insect cells2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1079932Highest frequency of moderate liver toxicity observed during clinical trials, expressed as a percentage. [column '% BIOL' in source]
AID81434Compound was evaluated for Antiviral activity against HIV-12001Journal of medicinal chemistry, Oct-11, Volume: 44, Issue:21
Synthesis of potent C(2)-symmetric, diol-based hiv-1 protease inhibitors. Investigation of thioalkyl and thioaryl P1/P1' substituents.
AID573996Ratio of EC50 for Human immunodeficiency virus 1 3B to EC50 for L-870,810-resistant Human immunodeficiency virus harboring L74M, E92Q and S230N mutations in integrase2008Antimicrobial agents and chemotherapy, Aug, Volume: 52, Issue:8
Preclinical evaluation of 1H-benzylindole derivatives as novel human immunodeficiency virus integrase strand transfer inhibitors.
AID525111Antiviral activity against HIV 1 NL4.3 assessed as inhibition of virus-induced cytopathic effect by MTT assay2008Antimicrobial agents and chemotherapy, Jun, Volume: 52, Issue:6
Mutations in human immunodeficiency virus type 1 integrase confer resistance to the naphthyridine L-870,810 and cross-resistance to the clinical trial drug GS-9137.
AID651635Viability Counterscreen for Primary qHTS for Inhibitors of ATXN expression
AID504812Inverse Agonists of the Thyroid Stimulating Hormone Receptor: HTS campaign2010Endocrinology, Jul, Volume: 151, Issue:7
A small molecule inverse agonist for the human thyroid-stimulating hormone receptor.
AID504810Antagonists of the Thyroid Stimulating Hormone Receptor: HTS campaign2010Endocrinology, Jul, Volume: 151, Issue:7
A small molecule inverse agonist for the human thyroid-stimulating hormone receptor.
AID1347082qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lassa (LASV) Arenavirus: LASV Primary Screen - GLuc reporter signal2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID1347093qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for SK-N-MC cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347096qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for U-2 OS cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347108qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Rh41 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347407qHTS to identify inhibitors of the type 1 interferon - major histocompatibility complex class I in skeletal muscle: primary screen against the NCATS Pharmaceutical Collection2020ACS chemical biology, 07-17, Volume: 15, Issue:7
High-Throughput Screening to Identify Inhibitors of the Type I Interferon-Major Histocompatibility Complex Class I Pathway in Skeletal Muscle.
AID1347090qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for DAOY cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347102qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Rh18 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347098qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for SK-N-SH cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347105qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for MG 63 (6-TG R) cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347086qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lymphocytic Choriomeningitis Arenaviruses (LCMV): LCMV Primary Screen - GLuc reporter signal2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID1347095qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for NB-EBc1 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347107qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Rh30 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1745845Primary qHTS for Inhibitors of ATXN expression
AID1347099qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for NB1643 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347083qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lassa (LASV) Arenavirus: Viability assay - alamar blue signal for LASV Primary Screen2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID1347089qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for TC32 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347103qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for OHS-50 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347094qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for BT-37 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347104qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for RD cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347100qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for LAN-5 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347101qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for BT-12 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347092qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for A673 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347091qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for SJ-GBM2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347424RapidFire Mass Spectrometry qHTS Assay for Modulators of WT P53-Induced Phosphatase 1 (WIP1)2019The Journal of biological chemistry, 11-15, Volume: 294, Issue:46
Physiologically relevant orthogonal assays for the discovery of small-molecule modulators of WIP1 phosphatase in high-throughput screens.
AID1347106qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for control Hh wild type fibroblast cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347154Primary screen GU AMC qHTS for Zika virus inhibitors2020Proceedings of the National Academy of Sciences of the United States of America, 12-08, Volume: 117, Issue:49
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors.
AID1347097qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Saos-2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1508630Primary qHTS for small molecule stabilizers of the endoplasmic reticulum resident proteome: Secreted ER Calcium Modulated Protein (SERCaMP) assay2021Cell reports, 04-27, Volume: 35, Issue:4
A target-agnostic screen identifies approved drugs to stabilize the endoplasmic reticulum-resident proteome.
AID1347425Rhodamine-PBP qHTS Assay for Modulators of WT P53-Induced Phosphatase 1 (WIP1)2019The Journal of biological chemistry, 11-15, Volume: 294, Issue:46
Physiologically relevant orthogonal assays for the discovery of small-molecule modulators of WIP1 phosphatase in high-throughput screens.
AID1508612NCATS Parallel Artificial Membrane Permeability Assay (PAMPA) Profiling2017Bioorganic & medicinal chemistry, 02-01, Volume: 25, Issue:3
Highly predictive and interpretable models for PAMPA permeability.
AID1796305Protease Inhibition Assay from Article 10.1016/s0960-894x(00)00163-3: \\Novel inhibitors of HIV protease: design, synthesis and biological evaluation of picomolar inhibitors containing cyclic P1/P2 scaffolds.\\2000Bioorganic & medicinal chemistry letters, Jun-05, Volume: 10, Issue:11
Novel inhibitors of HIV protease: design, synthesis and biological evaluation of picomolar inhibitors containing cyclic P1/P2 scaffolds.
AID1797110Protease Inhibition Assay from Article 10.1021/bi051886s: \\Analysis of HIV-1 CRF_01 A/E protease inhibitor resistance: structural determinants for maintaining sensitivity and developing resistance to atazanavir.\\2006Biochemistry, May-02, Volume: 45, Issue:17
Analysis of HIV-1 CRF_01 A/E protease inhibitor resistance: structural determinants for maintaining sensitivity and developing resistance to atazanavir.
AID1797107Protease Inhibition Assay from Article 10.1021/bi049459m: \\Comparing the accumulation of active- and nonactive-site mutations in the HIV-1 protease.\\2004Biochemistry, Sep-28, Volume: 43, Issue:38
Comparing the accumulation of active- and nonactive-site mutations in the HIV-1 protease.
AID1796876Enzyme Inhibition Assay from Article 10.1016/j.bmcl.2005.03.008: \\Oximinoarylsulfonamides as potent HIV protease inhibitors.\\2005Bioorganic & medicinal chemistry letters, May-02, Volume: 15, Issue:9
Oximinoarylsulfonamides as potent HIV protease inhibitors.
AID1799570Inhibition Assay from Article 10.1111/j.1747-0285.2007.00514.x: \\Design of mutation-resistant HIV protease inhibitors with the substrate envelope hypothesis.\\2007Chemical biology & drug design, May, Volume: 69, Issue:5
Design of mutation-resistant HIV protease inhibitors with the substrate envelope hypothesis.
AID1797666Enzyme Inhibition Assay from Article 10.1021/jm070284z: \\Design and Synthesis of HIV-1 Protease Inhibitors Incorporating Oxazolidinones as P2/P2' Ligands in Pseudosymmetric Dipeptide Isosteres.\\2007Journal of medicinal chemistry, Sep-06, Volume: 50, Issue:18
Design and synthesis of HIV-1 protease inhibitors incorporating oxazolidinones as P2/P2' ligands in pseudosymmetric dipeptide isosteres.
AID1795276Protease Inhibition Assay from Article 10.1021/bi027019u: \\A major role for a set of non-active site mutations in the development of HIV-1 protease drug resistance.\\2003Biochemistry, Jan-28, Volume: 42, Issue:3
A major role for a set of non-active site mutations in the development of HIV-1 protease drug resistance.
AID1795271Protease Inhibition Assay from Article 10.1021/bi035701y: \\Secondary mutations M36I and A71V in the human immunodeficiency virus type 1 protease can provide an advantage for the emergence of the primary mutation D30N.\\2003Biochemistry, Dec-30, Volume: 42, Issue:51
Secondary mutations M36I and A71V in the human immunodeficiency virus type 1 protease can provide an advantage for the emergence of the primary mutation D30N.
AID1799449Protease Inhibtion Assay from Article 10.1016/s1074-5521(98)90117-x: \\Design and selection of DMP 850 and DMP 851: the next generation of cyclic urea HIV protease inhibitors.\\1998Chemistry & biology, Oct, Volume: 5, Issue:10
Design and selection of DMP 850 and DMP 851: the next generation of cyclic urea HIV protease inhibitors.
AID1811Experimentally measured binding affinity data derived from PDB2004Biochemistry, Sep-28, Volume: 43, Issue:38
Comparing the accumulation of active- and nonactive-site mutations in the HIV-1 protease.
AID977610Experimentally measured binding affinity data (Ki) for protein-ligand complexes derived from PDB2004Biochemistry, Sep-28, Volume: 43, Issue:38
Comparing the accumulation of active- and nonactive-site mutations in the HIV-1 protease.
AID977610Experimentally measured binding affinity data (Ki) for protein-ligand complexes derived from PDB2012Journal of enzyme inhibition and medicinal chemistry, Feb, Volume: 27, Issue:1
The crystal structure of protease Sapp1p from Candida parapsilosis in complex with the HIV protease inhibitor ritonavir.
AID977610Experimentally measured binding affinity data (Ki) for protein-ligand complexes derived from PDB2012ChemMedChem, Feb-06, Volume: 7, Issue:2
Experimental and computational active site mapping as a starting point to fragment-based lead discovery.
AID1745854NCATS anti-infectives library activity on HEK293 viability as a counter-qHTS vs the C. elegans viability qHTS2023Disease models & mechanisms, 03-01, Volume: 16, Issue:3
In vivo quantitative high-throughput screening for drug discovery and comparative toxicology.
AID1745855NCATS anti-infectives library activity on the primary C. elegans qHTS viability assay2023Disease models & mechanisms, 03-01, Volume: 16, Issue:3
In vivo quantitative high-throughput screening for drug discovery and comparative toxicology.
AID1345258Human CYP3A4 (CYP3 family)2008Antimicrobial agents and chemotherapy, May, Volume: 52, Issue:5
Rapid clinical induction of hepatic cytochrome P4502B6 activity by ritonavir.
AID1345193Human CYP3A5 (CYP3 family)2005Current drug metabolism, Oct, Volume: 6, Issue:5
Cytochrome p450 enzymes mechanism based inhibitors: common sub-structures and reactivity.
AID1811Experimentally measured binding affinity data derived from PDB1995Proceedings of the National Academy of Sciences of the United States of America, Mar-28, Volume: 92, Issue:7
ABT-538 is a potent inhibitor of human immunodeficiency virus protease and has high oral bioavailability in humans.
AID977610Experimentally measured binding affinity data (Ki) for protein-ligand complexes derived from PDB1995Proceedings of the National Academy of Sciences of the United States of America, Mar-28, Volume: 92, Issue:7
ABT-538 is a potent inhibitor of human immunodeficiency virus protease and has high oral bioavailability in humans.
AID1159607Screen for inhibitors of RMI FANCM (MM2) intereaction2016Journal of biomolecular screening, Jul, Volume: 21, Issue:6
A High-Throughput Screening Strategy to Identify Protein-Protein Interaction Inhibitors That Block the Fanconi Anemia DNA Repair Pathway.
AID977608Experimentally measured binding affinity data (IC50) for protein-ligand complexes derived from PDB2013Biochemical and biophysical research communications, Feb-08, Volume: 431, Issue:2
Insights into the mechanism of drug resistance: X-ray structure analysis of multi-drug resistant HIV-1 protease ritonavir complex.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (5,101)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's463 (9.08)18.2507
2000's1654 (32.43)29.6817
2010's1880 (36.86)24.3611
2020's1104 (21.64)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 88.44

According to the monthly volume, diversity, and competition of internet searches for this compound, as well the volume and growth of publications, there is estimated to be very strong demand-to-supply ratio for research on this compound.

MetricThis Compound (vs All)
Research Demand Index88.44 (24.57)
Research Supply Index8.81 (2.92)
Research Growth Index4.90 (4.65)
Search Engine Demand Index163.45 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (88.44)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials1,291 (23.82%)5.53%
Reviews454 (8.38%)6.00%
Case Studies536 (9.89%)4.05%
Observational125 (2.31%)0.25%
Other3,013 (55.60%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (801)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
A Multicenter, Randomized, Double-Blind, Comparative Trial Of Maraviroc + Darunavir/Ritonavir Versus Emtricitabine/Tenofovir + Darunavir/Ritonavir For The Treatment Of Antiretroviral-Naive Hiv-Infected Patients With Ccr5-Tropic Hiv-1 [NCT01345630]Phase 3813 participants (Actual)Interventional2011-09-30Terminated(stopped due to See termination reason in detailed description.)
Continued Access to DRV/Rtv in HIV-1 Infected Children and Adolescents (Rollover Patients From C212, C228, C230) [NCT01138605]Phase 246 participants (Actual)Interventional2010-10-13Completed
Open-Label, Single-Sequence Study to Evaluate the Pharmacokinetic Interaction of BMS-663068 With Darunavir/Ritonavir and/or Etravirine in Healthy Subjects [NCT02063360]Phase 142 participants (Actual)Interventional2014-02-01Completed
Intensive Pharmacokinetic Studies of Antiretroviral Drug Combinations in Children [NCT00260078]Phase 1/Phase 275 participants (Actual)Interventional2006-02-28Completed
An Adaptive, Multicenter, Randomized, Open-label, Comparative Clinical Study to Assess Efficacy and Safety of Favipiravir in Hospitalized Patients With COVID-19 [NCT04434248]Phase 2/Phase 3330 participants (Actual)Interventional2020-04-23Active, not recruiting
A 48 Week, Phase II, Open-Label Multi-Cohort, Multicenter Study to Evaluate the Safety, Tolerability, Pharmacokinetics and Antiviral Activity of GW433908/Ritonavir QD and GW433908/Ritonavir BID When Administered to HIV-1 Infected, Antiretroviral Naive and [NCT00040664]Phase 269 participants (Actual)Interventional2002-07-31Completed
Plasma and Intracellular Pharmacokinetics of Once Daily Darunavir/Ritonavir and Twice and Once Daily Raltegravir in HIV-infected Subjects [NCT01047995]Phase 126 participants (Actual)Interventional2009-06-30Completed
Pharmacokinetic Interactions Between an Herbal Medicine (African Potato) and Antiretroviral Agents (Lopinavir/Ritonavir) [NCT01227590]Phase 118 participants (Actual)Interventional2010-02-28Completed
A Phase 1, Open Label, Randomized, Multiple Dose Study To Assess The Two-Way Drug Interaction Between Sitaxsentan And Low Dose Ritonavir In Healthy Subjects [NCT01251848]Phase 10 participants (Actual)Interventional2011-01-31Withdrawn
A Two-Way Interaction Study Between RO5190591/RTV and Ketoconazole in Healthy Subjects [NCT01164488]Phase 118 participants (Actual)Interventional2010-07-31Completed
[NCT01274780]Phase 4180 participants (Actual)Interventional2011-05-31Completed
A Multiple-Dose Study To Evaluate Safety, Tolerability, Pharmacokinetics and Antiviral Activity of Ritonavir-Boosted RO5190591 in Combination With Peginterferon Alfa-2a Plus Ribavirin in Patients With Chronic Hepatitis C Genotype 1 [NCT01185860]Phase 159 participants (Actual)Interventional2009-08-31Completed
Phase II, Parallel, Randomized, Clinical Trials Comparing the Responses to Initiation of NNRTI-Based Versus PI-Based Antiretroviral Therapy in HIV Infected Infants Who Have and Have Not Previously Received Single Dose Nevirapine for Prevention of Mother-t [NCT00307151]Phase 2452 participants (Actual)Interventional2005-12-31Completed
[NCT01346800]Phase 110 participants (Actual)Interventional2011-02-28Completed
Maternal Tenofovir-containing Combination Drug Regimen During the Second and Third Trimesters of Pregnancy for Prevention of Mother-to-child Transmission of HIV and HBV in HIV-HBV Co-infected Mothers [NCT01125696]Phase 245 participants (Actual)Interventional2012-05-31Completed
A Multicenter, Non-randomized, Open-label, Parallel Controlled Phase I Clinical Study to Evaluate the Pharmacokinetics, Safety and Tolerability of SIM0417/Ritonavir After a Single Dose in Subjects With Renal or Hepatic Impairment [NCT05731804]Phase 172 participants (Anticipated)Interventional2023-03-01Not yet recruiting
Once Daily (QD) Dosing of Lonafarnib (LNF) Co-administered With Ritonavir (RTV) for Treatment of Chronic Hepatitis D Virus Infection [NCT05229991]Phase 330 participants (Anticipated)Interventional2021-05-15Active, not recruiting
A PHASE 2/3, RANDOMIZED, DOUBLE-BLIND, DOUBLE-DUMMY, PLACEBO CONTROLLED STUDY TO EVALUATE THE SAFETY AND EFFICACY OF 2 REGIMENS OF ORALLY ADMINISTERED PF 07321332/RITONAVIR IN PREVENTING SYMPTOMATIC SARS-COV-2 INFECTION IN ADULT HOUSEHOLD CONTACTS OF AN I [NCT05047601]Phase 2/Phase 32,954 participants (Actual)Interventional2021-09-09Completed
The Influence of Ritonavir, Alone and in Combination With Lopinavir, on Fenofibric Acid Pharmacokinetics in Healthy Volunteers [NCT01148004]Phase 125 participants (Actual)Interventional2010-05-13Completed
A Study to Evaluate the Potential Drug-Drug Interaction Between Efavirenz and Danoprevir With Low Dose Ritonavir When Administered Together in Healthy Adult Volunteers [NCT01588002]Phase 140 participants (Actual)Interventional2012-04-30Completed
A Phase 1, Open-label Drug Interaction Study of PBI-200 With Ritonavir or Cobicistat in Healthy Volunteers [NCT05692570]Phase 121 participants (Actual)Interventional2022-09-09Completed
Optimal Combination Therapy After Nevirapine Exposure [NCT00089505]Phase 3745 participants (Actual)Interventional2006-11-30Completed
Randomized, Open-label, Comparative Safety and Efficacy Study of Tipranavir Boosted With Low-dose Ritonavir (TPV/RTV) Verses Genotypically-defined Protease Inhibitor/Ritonavir (PI/RTV) in Multiple Antiretroviral Drug-experienced Patients. [NCT00054717]Phase 3630 participants (Actual)Interventional2003-01-31Completed
An Open-label, Randomized, Crossover Study to Evaluate the Bioavailability of Two Candidate Tablet Formulations of ABT-450 With Reference to the Hard Gelatin Capsule Formulation [NCT01091649]Phase 140 participants (Actual)Interventional2010-02-28Completed
An Open-label, Single-sequence Study to Investigate the Effects of Cytochrome P450 1A2 Induction by Ritonavir on the Pharmacokinetics of BMS-986165 in Healthy Participants [NCT04055506]Phase 116 participants (Actual)Interventional2019-08-14Completed
An Open-label, Randomised Pilot Study Comparing the Efficacy, Safety and Tolerability of Raltegravir With Protease Inhibitor-based Therapy in Treatment-naïve, HIV/Hepatitis C Co-infected Injecting Drug Users Receiving Methadone [NCT01105611]Phase 440 participants (Anticipated)Interventional2010-08-31Recruiting
A Randomised, Open Label, Prospective Study to Assess Two Different Therapeutic Strategies Following First Treatment Failure in HIV-1 Infected Subjects [NCT01118871]Phase 43 participants (Actual)Interventional2010-05-31Terminated
Microboosting of Atazanavir 300 mg With 50 mg Versus 100 mg Ritonavir Daily in HIV-infected Patients: a Pharmacokinetic Study [NCT02034838]Phase 112 participants (Actual)Interventional2014-01-31Completed
A Study to Investigate HCV Response Rates in Real World Patients Traditionally Excluded From Clinical Trials: The HEARTLAND Study [NCT03710252]Phase 4100 participants (Actual)Interventional2016-03-31Completed
Clinical Evaluation of Adjusted Doses of Darunavir/Ritonavir With Rifampicin in HIV-infected Volunteers [NCT03892161]Phase 117 participants (Actual)Interventional2018-04-12Terminated(stopped due to Risks to participation)
Randomized Single Dose Multiple Crossover Relative Bioavailability Trial of New Tablet Formulations and Suspension Formulation Compared to Current (1B) Formulation of BILR 355 BS in Healthy Male Volunteer Subjects [NCT02259868]Phase 188 participants (Actual)Interventional2005-06-30Completed
The Effect of Hepatic Impairment on the Pharmacokinetics of RO5190591/Ritonavir: A Multiple-Center, Open-Label Study Following Multiple Oral Doses of RO5190591/Ritonavir to Subjects With Mild, Moderate, or Severe Hepatic Impairment and Healthy Subjects Wi [NCT01185873]Phase 181 participants (Actual)Interventional2011-02-28Completed
A Randomized Controlled Study Compares the 48 Weeks Results of HIV-1 RNA Between Ritonavir-boosted Lopinavir Monotherapy and Ritonavir-boosted Lopinavir + Optimized Background Regimens in HIV-1 Infected Patients Who Have HIV-1 RNA <50 Copies/ml More Than [NCT01189695]Phase 463 participants (Actual)Interventional2010-12-31Completed
Comparative LUSZ Therapeutic Study of Antiviral, Antiretroviral, and Immunosuppressive Treatments in Hospitalized COVID-19 Patients With High-Risk Factors, Biomarkers, and Disease Progression. [NCT05925140]Phase 11,000 participants (Anticipated)Interventional2020-03-28Recruiting
Study of Once-Daily Versus Twice-Daily Fosamprenavir Plus Ritonavir, Administered With Abacavir/Lamivudine Once-Daily in Antiretroviral-Naive HIV-1 Infected Adult Subjects. [NCT00450580]Phase 3212 participants (Actual)Interventional2007-03-31Completed
Randomised Evaluation of COVID-19 Therapy [NCT04381936]Phase 2/Phase 350,000 participants (Anticipated)Interventional2020-03-19Recruiting
Two Randomized, Open-labeled, Parallel Designed Multiple-dose Clinical Trials to Evaluate Pharmacokinetics of Ritonavir-unboosted and Ritonavir-boosted Atazanavir Used Alone or Co-administered With Tenofovir DF in Healthy Korean and Caucasian Male Volunte [NCT01368783]Phase 132 participants (Anticipated)Interventional2011-06-30Not yet recruiting
Immune Reconstitution in Severely Immunosuppressed Antiretroviral-naive HIV-1-Infected Patients (<100 CD4+ T Cells/μL) Taking Antiretroviral Regimens Based on Dolutegravir or Ritonavir-boosted Darunavir (the Advanz-4 Trial). [NCT02337322]Phase 4104 participants (Actual)Interventional2015-03-31Active, not recruiting
Post Authorization Non-interventional Study in HIV1-Infected Patients Starting or Already in Treatment With Darunavir [NCT01375881]31 participants (Actual)Observational2009-06-30Completed
A Phase I, Open-label, Randomized Cross-over, 2-period, 2-way Interaction Trial to Investigate the Pharmacokinetic Interaction Between Lopinavir/Ritonavir and TMC125 Both at Steady-state in Healthy Subjects. [NCT00767117]Phase 116 participants (Actual)Interventional2008-09-30Completed
A Randomised Controlled Trial Comparing the Efficacy of Infant Peri-exposure Prophylaxis With Lopinavir/Ritonavir (LPV/r) Versus Lamivudine to Prevent HIV-1 Transmission by Breastfeeding [NCT00640263]Phase 31,500 participants (Anticipated)Interventional2009-12-31Completed
A PHASE 1, OPEN-LABEL, RANDOMIZED, SINGLE-DOSE, CROSSOVER STUDY TO ESTIMATE THE RELATIVE BIOAVAILABILITY OF PF-07321332/RITONAVIR ORAL POWDER IN 3 DIFFERENT DELIVERY VEHICLES RELATIVE TO THE COMMERCIAL PF-07321332/RITONAVIR TABLETS IN HEALTHY ADULT PARTIC [NCT05263921]Phase 112 participants (Actual)Interventional2022-03-10Completed
A Study Investigating Plasma Abacavir and Its Intracellular Anabolite Carbovir-triphosphate Pharmacokinetics in the Absence and in the Presence of Darunavir/Ritonavir or Raltegravir in HIV-infected Subjects. [NCT00765271]Phase 129 participants (Actual)Interventional2008-05-31Completed
A Open-label, Single Center Drug Interaction Study of Morphothiadine Mesilate/Ritonavir , Entecavir and Tenofovir Disoproxil Fumarate in Healthy Subjects [NCT03662568]Phase 156 participants (Actual)Interventional2018-06-26Completed
Dolutegravir, Darunavir/Ritonavir and Optimized NRTI Recycling as a Third-line Antiretroviral Regimen in Cambodia [NCT03602690]Phase 254 participants (Anticipated)Interventional2018-10-04Recruiting
Tipranavir Disodium: An Open-Label ExploratorySstudy of Tipranavir and Ritonavir in Combination With One Nucleoside Reverse Transcriptase Inhibitor and One Non-Nucleoside Reverse Transcriptase Inhibitor in Multiple Protease Inhibitor-Experienced HIV Patie [NCT02238314]Phase 241 participants (Actual)Interventional1999-01-31Completed
Real World Evidence of the Effectiveness of Paritaprevir/r - Ombitasvir, ± Dasabuvir, ± Ribavirin and Patient Support Program in Patients With Chronic Hepatitis C - An Observational Study in Israel (CITRINE STUDY) [NCT02803138]256 participants (Actual)Observational2016-07-07Completed
An Open-label, Parallel Group, Multiple-dose Investigation of the Pharmacokinetics of Tipranavir Soft Elastic Capsules SEDDS and Ritonavir Soft Gel Capsules and Their Effects on Cytochrome P-450 (3A4) Activity in Normal Healthy Volunteers [NCT02251132]Phase 1113 participants (Actual)Interventional2000-10-31Completed
AN INTERVENTIONAL, EFFICACY AND SAFETY, PHASE 2, RANDOMIZED, DOUBLE-BLIND, 2-ARM STUDY TO INVESTIGATE A REPEAT 5-DAY COURSE OF NIRMATRELVIR/RITONAVIR COMPARED TO PLACEBO/RITONAVIR IN PARTICIPANTS AT LEAST 12 YEARS OF AGE WITH REBOUND OF COVID-19 SYMPTOMS [NCT05567952]Phase 2436 participants (Actual)Interventional2022-10-19Active, not recruiting
AN INTERVENTIONAL EFFICACY AND SAFETY, PHASE 2, RANDOMIZED, DOUBLE-BLIND, 3-ARM STUDY TO INVESTIGATE NIRMATRELVIR/RITONAVIR IN NONHOSPITALIZED PARTICIPANTS AT LEAST 12 YEARS OF AGE WITH SYMPTOMATIC COVID-19 WHO ARE IMMUNOCOMPROMISED [NCT05438602]Phase 2152 participants (Actual)Interventional2022-08-03Active, not recruiting
AGILE: Seamless Phase I/IIa Platform for the Rapid Evaluation of Candidates for COVID-19 Treatment [NCT04746183]Phase 1/Phase 2600 participants (Anticipated)Interventional2020-07-03Active, not recruiting
Sofosbuvir /Simeprevir/ Daclatasvir/Ribavirin Versus Sofosbuvir /Ombitasvir/ Paritaprevir /Ritonavir/Ribavirin in the Management of Hepatitis C Patients Fauilre to Prior Sofosbuvir/ Daclatasvir (An Open-labeled Randomized Trial) [NCT03549832]40 participants (Actual)Interventional2018-01-01Completed
An Open-label, Randomized, Crossover Relative BA Study of Pharmacokinetics and Safety of New SDS-containing Tablet and Capsule Formulations of BILR 355 Compared to the Current Formulation (50 mg Tablet), After Single Dose Oral Administration of BILR 355 P [NCT02253940]Phase 140 participants (Actual)Interventional2006-10-31Completed
Assessment of Single-dose Oral Tadalafil Pharmacokinetic Characteristics When Simultaneously Co-administered With Single-dose and Steady-state Tipranavir/Ritonavir 500 mg/200 mg to Healthy Male Volunteers [NCT02253862]Phase 117 participants (Actual)Interventional2005-12-31Completed
The Effect of Kaletra on CD4 Immune Reconstitution in HIV-infected Patients With Long-term Virologic Suppression on a Non-Kaletra Containing ART Regimen, But With a Blunted Immune Response [NCT00344487]3 participants (Actual)Interventional2005-12-31Terminated(stopped due to Inability to enroll subjects.)
Pilot Study of the Effect of a Non-tenofovir, Non-efavirenz-based HIV Regimen on Bone Density and Vitamin D Levels in African-American Patients With HIV Infection [NCT01343225]Phase 440 participants (Anticipated)Interventional2011-05-31Not yet recruiting
A Phase 1, Double-blind, Placebo-controlled, Randomized, Single Ascending Dose Study to Investigate the Safety, Tolerability, and Pharmacokinetics of TMC647055 in Combination With a Pharmacokinetic Enhancer (Part 1) Followed by an Open-label, Randomized, [NCT02064842]Phase 124 participants (Actual)Interventional2014-02-28Completed
Pharmacokinetic Interaction Study to Evaluate the Pharmacokinetic Effect of Rifabutin on BMS-626529, the Active Moiety of BMS-663068, With and Without Ritonavir in Healthy Subjects [NCT02138084]Phase 1102 participants (Actual)Interventional2014-05-14Completed
An Investigator Blinded, Randomized, Placebo-controlled Multiple Dose Escalation Study of the Safety, Tolerability and Pharmacokinetics of BILR 355 BS Plus Low Dose Ritonavir in HIV-uninfected Male Volunteers [NCT02253914]Phase 1100 participants (Actual)Interventional2004-02-29Completed
A Single-Centre Open-Label Study in Healthy Adult Volunteers to Assess the Pharmacokinetic Interactions Between Steady-State TPV (500 mg) and Single-Dose and Steady-State Atazanavir (300 mg QD) in the Presence of Ritonavir (100 mg) [NCT02253836]Phase 121 participants (Actual)Interventional2005-01-31Completed
Body Composition Sub-study of the D2EFT Trial [NCT03675815]Phase 4155 participants (Actual)Interventional2019-12-05Active, not recruiting
Randomized Trial of a Switch to a Kaletra + Current Dual Nucleoside Reverse Transcriptase Inhibitor (NRTI) Backbone Versus Continuation of the Current Regimen in Patients With Poor Immune Responses to Highly Active Antiretroviral Therapy (HAART) in Patien [NCT00145795]Phase 420 participants (Actual)Interventional2004-04-30Completed
"Safeguard the Household - A Study of HIV Antiretroviral Therapy Treatment Strategies Appropriate for a Resource Poor Country" [NCT00255840]812 participants (Actual)Interventional2006-07-31Completed
An Open Label, Parallel Design Study to Assess the Pharmacokinetics of Narlaprevir 200 mg as a Single Oral Dose and in Combination With Ritonavir 100 mg in Patients With Hepatic Impairment and Healthy Matched Volunteers [NCT03832426]Phase 132 participants (Actual)Interventional2013-11-08Completed
A Study Comparing Efficacy and Tolerance of Two Maintenance Strategies : a Monotherapy With Lopinavir/Ritonavir or a Single-tablet Triple Therapy by Efavirenz/Emtricitabin/Tenofovir in HIV-1 Infected Patients With HIV RNA Below 50 cp/mL [NCT00946595]Phase 2/Phase 3420 participants (Anticipated)Interventional2009-11-30Completed
Randomized, Open-label, Comparative Safety and Efficacy Study of Tipranavir Boosted With Low Dose Ritonavir (TPV/RTV) Versus Genotypically-defined Protease Inhibitor/Ritonavir (PI/RTV) in Multiple Antiretroviral Drug-experienced Patients (RESIST 2: Random [NCT00144170]Phase 3882 participants (Actual)Interventional2003-02-28Completed
Efficacy of Simplification to Atazanavir/Ritonavir + Lamivudine as Maintenance Therapy in Patients With Viral Suppression. Randomized, Open-label 96 Weeks Non-inferiority Trial [NCT01307488]Phase 4286 participants (Actual)Interventional2011-09-30Completed
Metabolic Effects of Switching Kaletra to Boosted Reyataz [NCT00413153]15 participants (Actual)Interventional2006-05-31Completed
A Phase I, Open-label, Randomized, 2-panel, 2-way Crossover Pivotal Bioequivalence Trial Between the Commercially Available 400-mg Tablet Formulation (F030) and the 800-mg Tablet Formulation of Darunavir (G002), in the Presence of Low-dose Ritonavir Under [NCT01308658]Phase 1128 participants (Actual)Interventional2011-01-31Completed
Assessment of the Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of SCH 900518 in Naive or Treatment-Experienced Subjects Infected With Hepatitis C Virus Genotype 1 (Protocol No. P04695) [NCT01081158]Phase 141 participants (Actual)Interventional2007-07-31Completed
A Phase IV, Open-Label, Randomized, Multicenter Trial Assessing a Reyataz-Based Substitution Approach in the Management of Lipodystrophy Syndrome. Research Into Atazanavir in Lipodystrophy (The REAL Study) [NCT00135356]Phase 4219 participants (Actual)Interventional2005-07-31Completed
A Phase IIIB, Randomized Trial of Open-Label Efavirenz or Atazanavir With Ritonavir in Combination With Double-Blind Comparison of Emtricitabine/Tenofovir or Abacavir/Lamivudine in Antiretroviral-Naive Subjects [NCT00118898]Phase 31,864 participants (Actual)Interventional2005-09-30Completed
Maintaining Options for Mothers Study (MOMS): A Phase II Randomized Comparison of Three Antiretroviral Strategies Administered for 7 or 21 Days to Reduce the Emergence of Nevirapine Resistant HIV-1 Following a Single Intrapartum Dose of Nevirapine [NCT00099632]Phase 2484 participants (Actual)Interventional2006-03-31Completed
Use of an Aluvia Based Highly Active Antiretroviral Therapy (HAART) Regimen in the Prevention of Mother to Child HIV Transmission (PMTCT) Antepartum, Intrapartum and Postpartum in Africa [NCT01088516]Phase 4280 participants (Actual)Interventional2008-12-31Completed
An Evaluation of the Uptake and Safety of, and Adherence to Antiretroviral Treatment Among Individuals With CD4 ≥ 250 Cells/mm3 and HIV Virus Load ≥ 50,000 cp/mL [NCT01583439]11 participants (Actual)Interventional2012-09-30Terminated(stopped due to Low Accrual.)
Efficacy and Safety in Clinical Practice of Ombitasvir/Paritaprevir/ Ritonavir and Dasabuvir Administered for 8 Weeks (3D8) in Treatment-naïve Genotype 1b Hepatitis C Virus Infected Patients: Analysis of Data From Hepa-C Registry. [NCT03122132]200 participants (Actual)Observational2017-02-20Completed
A Pilot Phase II Study of a Nucleoside Sparing Regimen of Dolutegravir + Atazanavir/r in HIV-1 Infected Patients With Detectable Viremia (DOLATAV Study) [NCT02542852]Phase 210 participants (Actual)Interventional2015-09-30Completed
Non-comparative Phase II Open Study Evaluating the Efficacy of a Reduced Dose Atazanavir / Ritonavir 200/100 mg + 2 NRTI in HIV-1-infected Patients With Virological Success With Atazanavir / Ritonavir 300/100 mg + 2 NRTI [NCT02473328]Phase 290 participants (Actual)Interventional2015-06-30Completed
Changes in Insulin Resistance in Healthy Volunteers on STRIBILD® Medication - A Controlled, Mono Center, Three-arm, Randomized Phase I Study. [NCT02203461]Phase 130 participants (Actual)Interventional2014-07-31Completed
A Retrospective Observational Non-Interventional Study (NIS) to Assess Patient Characteristics and Healthcare Resource Use (HCRU) Among COVID-19 Patients Receiving Treatment With Nirmatrelvir; Ritonavir (PAXLOVID TM) in the Kingdom of Saudi Arabia (KSA). [NCT06016556]500 participants (Anticipated)Observational2023-10-17Recruiting
A Phase I Study to Evaluate the Effect of Darunavir/Ritonavir and Lopinavir/Ritonavir on GSK2248761 Pharmacokinetics and to Assess the Effect of GSK2248761 on CYP450 Probe Drugs in Healthy Adult Subjects [NCT00920088]Phase 124 participants (Actual)Interventional2009-06-30Completed
PI or NNRTI as First-line Treatment of HIV in a West African Population With Low Adherence - the PIONA Trial [NCT01192035]Phase 4400 participants (Actual)Interventional2011-05-31Completed
Treatment of Chronic Delta Hepatitis With Lonafarnib, Ritonavir and Lambda Interferon [NCT03600714]Phase 226 participants (Actual)Interventional2018-08-01Completed
Effect Of Interferon-Free Direct Acting Antiviral Agents For Treatment Of Hepatitis C Virus Patients On The Normal Kidney [NCT03296930]Phase 4100 participants (Anticipated)Interventional2017-10-01Not yet recruiting
Multicenter Study of Options for Second-Line Effective Combination Therapy (SELECT) [NCT01352715]Phase 3515 participants (Actual)Interventional2012-03-13Completed
A Phase IIIB, Open-label, Randomized, Multi-center Study Evaluating the Effect on Serum Lipids Following a Switch to ATV/r in HIV-1 Infected Subjects Who Have Achieved Virologic Suppression on a LPV/r Based Regimen. [NCT00120393]Phase 3192 participants (Anticipated)Interventional2004-01-31Completed
Combination Therapies to Reduce the Nasopharyngeal Carriage of SARS-CoV-2 and Improve the Outcome of COVID-19 Infection in Ivory Coast (INTENSE-COV): a Phase IIb Randomized Clinical Trial [NCT04466241]Phase 2/Phase 3294 participants (Anticipated)Interventional2020-11-27Recruiting
A Randomized, Open-Label, Multicenter Study to Evaluate the Efficacy and Safety of ABT-493/ABT-530 in Japanese Adults With Chronic Hepatitis C Virus Infection (CERTAIN-1) [NCT02707952]Phase 3295 participants (Actual)Interventional2016-02-22Completed
A Double Blind, Placebo Controlled, Single and Repeat Dose Escalation Study of GSK2838232 With and Without Ritonavir for 8-11 Days in Healthy Subjects [NCT02289495]Phase 124 participants (Actual)Interventional2014-11-18Terminated
An Open Labe Study to Evaluate the Drug-Drug Interaction of Itraconazole, Rifampicin and Midazolam With SIM0417/Ritonavir in Healthy Adult Chinese Participants [NCT05665647]Phase 136 participants (Actual)Interventional2022-12-29Completed
A Randomized, Open Label Trial to Investigate the Efficacy and Safety of Nitazoxanide Plus Atazanavir/Ritonavir for the Treatment of COVID-19: a Pilot Study [NCT04459286]Phase 257 participants (Actual)Interventional2020-10-09Terminated(stopped due to IDSMB recommendation)
A Study to Evaluate Safety, Tolerability, Pharmacokinetics and Antiviral Activity of Ritonavir-Boosted DANOPREVIR and RO5024048 in Different Combinations in Null Responder or Treatment Naïve Patients With Chronic Hepatitis C and Compensated Cirrhosis [NCT01483742]Phase 243 participants (Actual)Interventional2012-04-30Completed
A Phase 1/2a, Multicenter, Open-Label, First in Human Study to Assess the Safety, Tolerability, Pharmacokinetics, and Preliminary Antitumor Activity of DB-1303 in Patients With Advanced/Metastatic Solid Tumors [NCT05150691]Phase 1/Phase 2631 participants (Anticipated)Interventional2022-01-31Recruiting
Safety and Clinical and Virologic Outcomes in COVID-19 Patients With Chronic Kidney Disease Treated With Nirmatrelvir-ritonavir [NCT05624840]100 participants (Anticipated)Observational2022-11-07Recruiting
Relative Bioavailability of 500/200 mg of Tipranavir/Ritonavir Paediatric Solution Compared to 500/200 mg of Tipranavir/Ritonavir Capsules Following Oral Administration and Bioavailability of 500/200 mg Tipranavir/Ritonavir Paediatric Solution Under the I [NCT02251158]Phase 130 participants (Actual)Interventional2003-10-31Completed
Evaluation of SSD8432 in Combination With Ritonavir in Asymptomatic Infections or Mild/Common Randomized, Double-blind, Safety Study of Efficacy and Safety in Adult Subjects With COVID-19 Placebo-Controlled, Phase II Clinical Study [NCT05373446]Phase 272 participants (Anticipated)Interventional2022-05-20Not yet recruiting
A Double-Blind, Randomized, Placebo-Controlled, Continuation Single Dose Escalation Study to Investigate the Safety, Tolerability and Pharmacokinetics of GSK2838232 With and Without Ritonavir, and to Evaluate Different Formulations of GSK2838232, in Healt [NCT02289482]Phase 120 participants (Actual)Interventional2014-11-17Terminated(stopped due to Study prematurely terminated due to safety concerns leading to clinical hold)
An Open-label, Randomized, Single-dose, Two-way Crossover Bioequivalence Study Assessing Subjects Receiving Tipranavir 500 mg Stored at Controlled Temperature (Testproduct 30°C/70% RH) Compared With Tipranavir 500 mg Stored at Controlled Refrigerated Cond [NCT02253888]Phase 130 participants (Actual)Interventional2005-05-31Completed
Phase II Study of the Pharmacokinetics of Nevirapine and the Incidence of Nevirapine Resistance Mutations in HIV-Infected Women Receiving a Single Intrapartum Dose of Nevirapine With the Concomitant Administration of Zidovudine/Didanosine or Zidovudine/Di [NCT00109590]Phase 2175 participants (Actual)Interventional2006-06-30Completed
A Single-Center, Randomized, Double-Blind, Single and Multiple Ascending Dose, Placebo-Controlled Study to Investigate the Safety, Tolerability and Pharmacokinetics of RO6889678 and the Combination of RO6889678 With Ritonavir Following Oral Administration [NCT02321384]Phase 1100 participants (Actual)Interventional2014-12-15Terminated
Multicenter, Open-label, Phase II Safety and Efficacy Study of All-oral Combination Narlaprevir/Ritonavir and Daclatasvir Administered for 12 Weeks in Patients With Genotype 1b Chronic Hepatitis C [NCT03485846]Phase 2105 participants (Actual)Interventional2017-11-27Completed
A Phase III, Randomized, Open-Label Trial to Evaluate Strategies for Providing Antiretroviral Therapy to Infants Shortly After Primary Infection in a Resource Poor Setting [NCT00102960]Phase 3377 participants (Actual)Interventional2005-07-31Completed
A PHASE 1, OPEN-LABEL, NON-RANDOMIZED STUDY TO INVESTIGATE THE SAFETY AND PK FOLLOWING MULTIPLE ORAL DOSES OF PF-07321332 (NIRMATRELVIR)/RITONAVIR IN ADULT PARTICIPANTS WITH COVID-19 AND SEVERE RENAL IMPAIRMENT EITHER ON HEMODIALYSIS OR NOT ON HEMODIALYSI [NCT05487040]Phase 115 participants (Actual)Interventional2022-09-07Terminated(stopped due to Decision to terminate study was due to slow enrolment and FDA input that available data may be enough for dosing recommendations for severe renal disease (subject to review by FDA).)
Safety and Antiviral Activity of TPV in Hepatitis C or Hepatitis B HIV Coinfected Patients - TDM Randomised Pilot Evaluation [NCT00447902]Phase 311 participants (Actual)Interventional2007-03-31Terminated
Relative Bioavailability of a Single Oral Dose of BIBW 2992 (20 mg) After Coadministration With Multiple Oral Doses of Ritonavir (200 mg Bid for 3 Days) Compared to the Bioavailability of a Single Oral Dose of BIBW 2992 (20 mg) Alone in Healthy Male Volun [NCT02171754]Phase 122 participants (Actual)Interventional2009-09-30Completed
Weekly ModraDoc/r in Combination With Hormonal Treatment and High-dose Intensity-modulated Radiation Therapy in Patients With High-risk Early Stage Prostate Cancer [NCT03066154]Phase 124 participants (Anticipated)Interventional2016-09-30Recruiting
Multiple-Dose Pharmacokinetics, Safety and Tolerability of the Co-administration of ABT-450/Ritonavir/ABT-267 (ABT-450/r/ABT-267) and ABT-333 in Healthy Chinese Subjects [NCT02534870]Phase 118 participants (Actual)Interventional2015-09-30Completed
Open Label, Study to Determine the Pharmacokinetic Interactions of Steady State Tipranavir/Ritonavir (500/200 mg) and Steady State Ribavirin and Pegylated Interferon Alfa 2a in HIV Negative, HCV Infected Subjects With Mild Hepatic Impairment and the Pharm [NCT02259855]Phase 136 participants (Actual)Interventional2006-01-31Completed
Study of Pharmacokinetic Interaction Between Combivir® (ZDV+3TC) and BILR 355 BS Plus Ritonavir [NCT02256774]Phase 151 participants (Actual)Interventional2004-10-31Completed
An Open Study to Investigate the Effect of Different Boosting Agents on Pharmacokinetics of Single Doses of BILR 355 BS (Dose Steps: 5 and 12.5 mg) Dissolved in 5 mL PEG 400 After Oral Administration in Healthy Male Volunteers [NCT02257008]Phase 144 participants (Actual)Interventional2003-03-31Completed
A Single Centre, Open-label Study, in Healthy Adult Volunteers, to Determine the Effects of Single-dose and Steady-state TPV/RTV 500/200 mg on the Steady-state Pharmacokinetics of Fluconazole 100 mg qd (200 mg Loading Dose) [NCT02195466]Phase 120 participants (Actual)Interventional2003-06-30Completed
Efficacy and Safety of Darunavir/Cobicistat vs. Lopinavir/Ritonavir in the Management of Patients With COVID-19 Pneumonia in Qatar [NCT04425382]400 participants (Actual)Observational2020-03-01Completed
An Open-label One-sequence Cross-over Pharmacokinetic Interaction Study of Steady-state Tipranavir/Ritonavir 500/200 mg With Single-dose Valaciclovir (500 mg) in Healthy Volunteers [NCT02226978]Phase 129 participants (Actual)Interventional2007-02-28Completed
Multi-centre, Adaptive, Randomized Trial of the Safety and Efficacy of Treatments of COVID-19 in Hospitalized Adults [NCT04315948]Phase 31,552 participants (Actual)Interventional2020-03-22Completed
A Prosp., Multic., Randomized, Open-label Trial to Assess the Safety, Tolerability and Efficacy of Dual Therapy With Boosted Darunavir + Dolutegravir When Switching From SOC ART in HIV-patients With Sustained Virological Suppr. [NCT02486133]Phase 3269 participants (Actual)Interventional2015-07-31Completed
A Randomized Open-label Study to Evaluate the Sustained Virologic Response of Danoprevir/Ritonavir and Copegus in Combination With RO5024048 and/or Pegasys in Chronic Hepatitis C Genotype 1 Patients Who Failed Previous Standard Therapy [NCT01331850]Phase 2381 participants (Actual)Interventional2011-05-31Completed
A Phase IIIB/IV Randomised Open-label Trial Comparing Dolutegravir With Pharmaco-enhanced Darunavir Versus Dolutegravir With Predetermined Nucleosides Versus Recommended Standard of Care ART Regimens in Patients With HIV-1 Infection Failing First Line The [NCT03017872]Phase 4831 participants (Actual)Interventional2017-11-23Active, not recruiting
HIV Infection and Gut Mucosal Immune Function: Longitudinal Analyses of Intestinal CD4+ and Th17 T Cells in HIV-infected Individuals on Short-term Antiretroviral Therapy [NCT02097381]10 participants (Actual)Interventional2010-04-30Active, not recruiting
Implementation and Evaluation of an HIV-2 Viral Load and ARV Resistance Informed Algorithm for 2nd-line ART in HIV-2 Infected Patients in the Initiative Sénégalaise d'Accès Aux Antirétroviraux (ISAARV) Program [NCT03394196]152 participants (Actual)Interventional2018-07-04Terminated(stopped due to COVID-19 and Funding)
The Pharmacodynamic/Pharmacokinetic Interaction of Tipranavir and Ritonavir With Loperamide in Healthy Volunteers [NCT02251119]Phase 124 participants (Actual)Interventional2002-07-31Completed
Safety and Antiviral Efficacy of a Novel HIV-1 Protease Inhibitor, BMS-232632, in Combination Regimen(s) as Compared to a Reference Combination Regimen(s) in Antiretroviral-Experienced HIV-Infected Subjects [NCT00004584]Phase 20 participants Interventional1999-12-31Completed
Phase I Sequential Dose Escalation Study of Pharmacokinetics, Safety and Tolerability After Single Dose (225 Mg-450 mg) Oral Administration of BILR 355 (SDS) Plus Low-dose Ritonavir in Healthy Volunteers [NCT02253927]Phase 148 participants (Actual)Interventional2006-05-31Completed
A Single Centre, Open-label Study With Healthy Adult Volunteers to Determine the Effects of Single-dose and Steady-state TPV/r 500/200 mg on the Steady-state Pharmacokinetics of Carbamazepine (200 mg Twice Daily) [NCT02253849]Phase 128 participants (Actual)Interventional2005-11-30Completed
An Open-Label, Single Arm Study to Evaluate the Safety and Efficacy of Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir in Treatment-Naïve Adults With Genotype 1b Hepatitis C Virus (HCV) Without Cirrhosis (GARNET) [NCT02582632]Phase 3166 participants (Actual)Interventional2015-11-24Completed
Open-Label, Fixed-Sequence, Crossover Study To Estimate The Pharmacokinetic Interaction Between Multiple Dose Maraviroc And Fosamprenavir/Ritonavir In Healthy Subjects [NCT01140412]Phase 114 participants (Actual)Interventional2010-07-31Terminated(stopped due to The trial prematurely terminated on November 1, 2010, due to healthy volunteer participants experiencing non-serious fosamprenavir-related skin rash.)
A Phase 3, Randomized, Open Label, Controlled Study of Lopinavir/Ritonavir and Lamivudine Versus Standard Therapy in Naïve HIV-1 Infected Subjects. [NCT01237444]Phase 3417 participants (Actual)Interventional2010-12-31Completed
Study of Pharmacokinetic Interaction Between Kaletra® (LPV/r) and BILR 355 BS Plus Ritonavir [NCT02256826]Phase 134 participants (Actual)Interventional2005-04-30Completed
A Double-Blind (Sponsor Unblinded), Randomized, Placebo-Controlled, Continuation Single and Repeated Dose Escalation Study to Investigate the Safety, Tolerability and Pharmacokinetics of GSK2838232 With Ritonavir, in Healthy Subjects [NCT02795754]Phase 162 participants (Actual)Interventional2016-03-31Completed
The SETPOINT Study - A Randomized Study of the Effect of Immediate Treatment With Potent Antiretroviral Therapy Versus Observation With Treatment as Indicated in Newly Infected HIV-1 Infected Subjects: Does Early Therapy After the Virologic Setpoint? [NCT00090779]Phase 2130 participants (Actual)Interventional2005-01-31Terminated(stopped due to The DSMB concluded that the findings regarding the primary analysis would persist and that no additional study goals would be achieved by continuing the study.)
Effect of Steady State TPV/r 500 mg/200 mg on Intracellular Concentrations of Zidovudine Triphosphate and Carbovir Triphosphate [NCT02229760]Phase 1/Phase 23 participants (Actual)Interventional2006-08-31Terminated
Pharmacokinetic Properties of Antiretroviral and Related Drugs During Pregnancy and Postpartum [NCT00042289]1,578 participants (Actual)Observational2003-06-09Completed
Drug Interaction Study Between Inhaled Beclomethasone and Protease Inhibitors in Healthy Volunteers [NCT00936793]Phase 153 participants (Actual)Interventional2009-07-06Completed
Open-Label, Multiple-Dose, Drug Interaction Study to Assess the Effect of Famotidine With or Without Tenofovir on the Pharmacokinetics of Atazanavir When Given With Ritonavir in HIV-Infected Subjects [NCT00384904]Phase 440 participants (Actual)Interventional2006-12-31Completed
An Open-label Phase II Pilot Study of Prevention of Perinatal Transmission of HIV-1 Without Nucleoside Reverse Transcriptase Inhibitors [NCT02738502]Phase 291 participants (Actual)Interventional2016-07-06Completed
Exploratory, Cross-sectional Study to Compare the Virologic Efficacy in Cerebrospinal Fluid (CSF) and Neurocognitive State in Patients Infected by HIV-1 Long-term Treatment (> 3 Years) With Lopinavir / Ritonavir Monotherapy [NCT01116817]Phase 435 participants (Actual)Interventional2010-08-31Completed
An Open, Prospective Study to Compare the Safety and Efficacy of Raltegravir vs. Atazanavir / Ritonavir, Both in Combination With Tenofovir DF and Emtricitabine, in the Treatment of HIV-infection in ART Naive Subjects With HCV Co-infection. [NCT01225705]Phase 40 participants (Actual)Interventional2010-10-31Withdrawn(stopped due to no pts recruited)
Pharmacokinetics of Low- Dose Lopinavir/Ritonavir Tablet Formulation HIV-1 Infected Children [NCT01139905]Phase 224 participants (Actual)Interventional2010-04-30Completed
A PHASE 1, OPEN-LABEL, 3-TREATMENT, 6-SEQUENCE, 3-PERIOD CROSSOVER STUDY TO ESTIMATE THE EFFECT OF PF-07321332/RITONAVIR AND RITONAVIR ON THE PHARMACOKINETICS OF DABIGATRAN IN HEALTHY PARTICIPANTS [NCT05064800]Phase 124 participants (Actual)Interventional2021-09-21Completed
A Phase IIa Randomized, Controlled Study of Combination Therapies to Treat COVID-19 Infection [NCT04459702]Phase 20 participants (Actual)Interventional2020-07-31Withdrawn(stopped due to Was never started)
Tenofovir, Emtricitabine, Efavirenz and Atazanavir Pharmacokinetics in the Aging HIV-Infected Population [NCT01180075]85 participants (Actual)Observational2010-05-31Completed
Open-Label, Fixed-Sequence Study To Estimate The Pharmacokinetic Interaction Between Multiple Dose Maraviroc And Fosamprenavir/Ritonavir In Healthy Subjects [NCT01290211]Phase 128 participants (Actual)Interventional2011-04-30Completed
Phase I, Open Label, Two Period Study to Evaluate the Effects of Fosamprenavir/Ritonavir on GSK1349572 Pharmacokinetics and a Phase I, Randomized, Three-Way Crossover Study to Evaluate the Relative Bioavailability of Three Tablet Variants Made Using Micro [NCT01209065]Phase 127 participants (Actual)Interventional2010-09-30Completed
OPtimisation of Antiviral Therapy in Immunocompromised COVID-19 Patients: a Randomized Factorial Controlled Strategy Trial: the OPTICOV Study [NCT05587894]Phase 2256 participants (Anticipated)Interventional2023-04-27Recruiting
COVID-19 Ring-based Prevention Trial With Lopinavir/Ritonavir [NCT04321174]Phase 3123 participants (Actual)Interventional2020-04-17Active, not recruiting
Investigation of the Pharmacokinetics of Atazanavir in Pregnant Women, Individuals at Extremes of BMI, Children, and Adolescents: An Observational Study Nested Within the VirTUAL Consortium [NCT03923231]32 participants (Actual)Observational2019-09-02Completed
A Randomized Prospective Open Label Study of Switching to Raltegravir Based ART Compared to Maintaining Ritonavir Boosted PI-based ART on Liver Fibrosis Progression in HIV-HCV Coinfected Patients [NCT01231685]Phase 29 participants (Actual)Interventional2011-12-31Completed
Pharmacokinetics of Maraviroc and Boosted Atazanavir Dual Regimen in Stable HIV-infected Patients [NCT03708861]Phase 30 participants (Actual)Interventional2016-01-31Withdrawn
A Double-Blind, Randomized, Placebo-Controlled Phase II Study of Lopinavir/Ritonavir Versus Placebo in COVID-19 Positive Patients With Cancer and Immune Suppression in the Last Year [NCT04455958]Phase 20 participants (Actual)Interventional2021-05-01Withdrawn(stopped due to limited resources)
Un-randomized, Open, Multiple-Dose Study to Evaluate the Safety, Antiviral Activity, and Pharmacokinetics of Morphothiadine Mesilate Capsules (GLS4) /Ritonavir Tablets(RTV) in Patients With Chronic Hepatitis B [NCT03638076]Phase 220 participants (Actual)Interventional2017-07-26Completed
Post-prandial Lipid Effects of Raltegravir (RAL) vs Ritonavir-boosted Darunavir (DRV-r) in Anti-retroviral Therapy (ART)-Naive Adults or Adults Recommencing ART. [NCT01258439]Phase 425 participants (Actual)Interventional2010-11-30Completed
Ensayo clínico, Abierto, Aleatorizado Para Comparar la Calidad de Vida de Los Pacientes VIH+ Que Inician Monoterapia Con Comprimidos de LPV/r vs Triple Terapia Que Contenga un IP Potenciado [NCT01166477]Phase 4228 participants (Actual)Interventional2010-01-31Completed
the Investigation Into Beneficial Effects of High-dose Interferon Beta 1-a, Compared to Low-dose Interferon Beta 1-a in Moderate to Severe Covid-19 [NCT04521400]Phase 2100 participants (Anticipated)Interventional2020-08-20Not yet recruiting
A Two Part Study to Assess i) the Relative Bioavailability and Food Effect of a Novel Tablet Formulation of Boosted-GSK2838232 Compared to Capsule and ii) the Safety and Pharmacokinetics of Repeated Once-Daily Doses of Non-boosted GSK2838232 [NCT03234036]Phase 126 participants (Actual)Interventional2017-08-02Completed
Effect of Cobicistat Versus Ritonavir Boosting on the Brain Permeation of Darunavir in HIV-infected Individuals [NCT02503462]Phase 47 participants (Actual)Interventional2015-07-31Terminated(stopped due to No additional patients fulfilling the inclusion criteria)
A Randomized, Double Blind, Dose Escalation, Fusion, First Time in Human Study to Assess the Safety, Tolerability, Pharmacokinetics, and Antiviral Activity of Single and Repeat Doses of GSK2485852 in Chronically Infected Hepatitis C Subjects [NCT01332552]Phase 127 participants (Actual)Interventional2011-01-14Terminated(stopped due to Terminated due to unfavorable human pharmacokinetic properties)
An Open-label, Randomized Bioequivalence Study of Ritonavir Versus NORVIR in Healthy Chinese Subjects [NCT03302182]Phase 196 participants (Actual)Interventional2017-09-23Completed
A Prospective, Randomized, Open-labelled, Multi-centre Trial Comparing the Safety and Efficacy of Ritonavir-boosted Aptivus (Tipranavir, TPV/r) to That of Prezista® (Darunavir, DRV/r) in Three-class (NRTI, NNRTI, and PI) Treatment-experienced Patients Wit [NCT00517192]Phase 340 participants (Actual)Interventional2007-09-30Terminated
An Open-Label Pilot Study to Evaluate the Antiviral Activity, Safety and Pharmacokinetics of ABT-450 With Ritonavir (ABT-450/r) Dosed in Combination With ABT-333 and Ribavirin (RBV) in Treatment-Naive and Non-responder Subjects With Genotype 1 Chronic Hep [NCT01306617]Phase 250 participants (Actual)Interventional2011-02-28Completed
Phase II Trial of Ritonavir/Lopinavir in Patients With Progressive of Recurrent High-Grade Gliomas [NCT01095094]Phase 219 participants (Actual)Interventional2009-01-31Terminated(stopped due to Study did not meet its primary objective)
Multicenter Clinical Study on the Efficacy and Safety of Xiyanping Injection in the Treatment of New Coronavirus Infection Pneumonia (General and Severe) [NCT04295551]80 participants (Anticipated)Interventional2020-03-14Not yet recruiting
A Phase 1, Open-Label, Randomized, Single Dose, Crossover Study to Estimate the Relative Bioavailability of Nirmatrelvir and Ritonavir Following Oral Administration of 4 Different Fixed Dose Combination Tablet Formulations Relative to The Commercial Table [NCT05525910]Phase 115 participants (Actual)Interventional2022-08-31Completed
Open-Label, Multiple-Dose, Drug Interaction Study to Assess the Effect of Famotidine on the Pharmacokinetics of Atazanavir in HIV-Infected Subjects Receiving Atazanavir With Ritonavir and Tenofovir [NCT01232127]Phase 425 participants (Actual)Interventional2011-02-28Completed
Protease Inhibitors to Reduce Malaria Morbidity in HIV-Infected Pregnant Women [NCT00993031]Phase 3389 participants (Actual)Interventional2009-12-15Completed
An Open Clinical Trial to Evaluate Ganovo(Danoprevir ) Combined With Ritonavir in the Treatment of SARS-CoV-2 Infection [NCT04291729]Phase 411 participants (Actual)Interventional2020-02-17Completed
Atazanavir (BMS-232632) for HIV Infected Individuals Completing Atazanavir Clinical Trials: An Extended Access Study [NCT01003990]Phase 3710 participants (Actual)Interventional2002-10-31Completed
A Phase 1, 2-Panel, Fixed-Sequence, Open-Label Single-Center Study to Assess the Effect of Single and Multiple Doses of Darunavir in Combination With Cobicistat or Ritonavir on the Pharmacokinetics of Single Dose Dabigatran Etexilate in Healthy Subjects [NCT04208061]Phase 128 participants (Actual)Interventional2019-12-18Completed
A Randomized Open Label Trial of HIV Protease Inhibitors for the Prevention of Malaria in HIV-Infected Children [NCT00978068]Phase 3176 participants (Actual)Interventional2009-09-30Completed
Pharmacokinetics of Rifabutin Combined With Antiretroviral Therapy in the Treatment of Tuberculosis Patient With HIV Infection in South Africa: A Phase II Trial [NCT00640887]Phase 248 participants (Anticipated)Interventional2009-02-28Completed
Phase IV, Non Randomized Study in ARV Experienced Patients Under Switch Therapy With Kaletra [NCT00648999]Phase 4207 participants (Actual)Interventional2003-11-30Completed
A PHASE 1, NON-RANDOMIZED, OPEN-LABEL STUDY TO ASSESS THE PHARMACOKINETICS, SAFETY AND TOLERABILITY OF PF-07321332 BOOSTED WITH RITONAVIR IN ADULT PARTICIPANTS WITH RENAL IMPAIRMENT AND IN HEALTHY PARTICIPANTS WITH NORMAL RENAL FUNCTION. [NCT04909853]Phase 135 participants (Actual)Interventional2021-06-15Completed
Clinical Trial to Assess the Security of the Dose Reduction of Ritonavir in HIV-Infected Patients in Treatment With Tipranavir/Ritonavir 500/200 mg Every 12 Hours [NCT00607958]Phase 411 participants (Actual)Interventional2007-12-31Completed
Radiation Therapy in Combination With Indinavir / Ritonavir (Crixivan / Norvir) for the Treatment of Brain Metastases: a Randomized Phase II Study [NCT00637637]Phase 260 participants (Anticipated)Interventional2007-09-30Recruiting
Pharmacokinetic Study of Posaconazole Boosted Fosamprenavir [NCT00817765]Phase 124 participants (Anticipated)Interventional2009-01-31Completed
A Pilot Study Of the Effects of Highly Active Antiretroviral Therapy on Kaposi's Sarcoma in Zimbabwe [NCT00834457]Phase 2/Phase 349 participants (Actual)Interventional2007-06-30Completed
An Open-Label Study to Evaluate the Safety and Efficacy of Ombitasvir/Paritaprevir/Ritonavir in Japanese Adults With Genotype 1b Chronic Hepatitis C Virus (HCV) Infection With End Stage Renal Disease on Hemodialysis (GIFT-R) [NCT02806362]Phase 30 participants (Actual)Interventional2016-09-30Withdrawn(stopped due to Study halted prematurely, prior to enrollment of first participant)
Interaction of Buprenorphine With HIV Medications and Tuberculosis Medications [NCT00877591]Phase 163 participants (Actual)Interventional2008-04-30Completed
A Randomized, Open-label Trial to Compare the Efficacy and Safety of Early Initiation of cART With or Without Autologous HIV-1 Specific Cytotoxic T Lymphocyte (CTL) Infusion in Treatment-Naïve Acute HIV-1 Infected Adults [NCT02231281]Phase 365 participants (Anticipated)Interventional2014-08-31Active, not recruiting
A Single-centre, Open-label Study to Assess the Effects of Steady-state Efavirenz 600 mg QD (Sustiva®) on Tipranavir Concentration When Tipranavir/Ritonavir Are Administered at Doses 500 mg/200 mg BID to Steady-state in Healthy Adult Volunteers [NCT02226991]Phase 134 participants (Actual)Interventional2006-04-30Completed
A Phase I, Single and Multiple Dose, to Evaluate the Safety, Tolerability, and Pharmacokinetics of SSD8432 and SSD8432 Co-administrated in Healthy Adult Subjects [NCT05339646]Phase 1108 participants (Anticipated)Interventional2022-04-07Recruiting
A Randomized, Non-comparative, Phase IIb, Unblinded Trial, Evaluating the Efficacy and Safety of Tenofovir-emtricitabine or Lamivudine Plus Zidovudine, Lopinavir/Ritonavir, or Raltegravir, Among ARV-naïve HIV-2 Infected Adult Patients, in West Africa [NCT02150993]Phase 2/Phase 3210 participants (Actual)Interventional2016-01-26Completed
Trial of Early Therapies During Non-hospitalized Outpatient Window (TREAT NOW) for COVID-19 [NCT04372628]Phase 2452 participants (Actual)Interventional2020-06-01Completed
[NCT01220947]Phase 2421 participants (Actual)Interventional2010-11-30Completed
Randomized, Open Label, Multiple-Dose Study to Evaluate the Pharmacodynamics, Safety and Pharmacokinetics of BMS-663068 in HIV-1 Infected Subjects [NCT01009814]Phase 250 participants (Actual)Interventional2009-11-23Completed
Relative Bioavailability of a Single Oral Dose of BI 1356 (5 mg) After Co-administration With Multiple Oral Doses of Ritonavir (200 mg Bid for 3 Days) Compared to the Bioavailability of a Single Oral Dose of BI 1356 (5 mg) Alone in Healthy Male Volunteers [NCT02183441]Phase 112 participants (Actual)Interventional2008-04-30Completed
Lopinavir/Ritonavir as an Immunomodulator to Enhance Vaccine Responsiveness [NCT01165645]0 participants (Actual)Interventional2010-11-30Withdrawn(stopped due to no patients enrolled)
Phase I, Drug-drug Interaction Study in Healthy Volunteers to Investigate the Safety, Tolerability and Pharmacokinetics of VM-1500 When Administered Orally, in Combination With Raltegravir or Darunavir [NCT02489487]Phase 124 participants (Actual)Interventional2014-09-30Completed
An Interventional Decentralized Phase 2, Randomized, Double-Blind, 2-Arm Study to Investigate the Efficacy and Safety of Orally Administered Nirmatrelvir/Ritonavir Compared With Placebo/Ritonavir in Participants With Long COVID [NCT05668091]Phase 2100 participants (Anticipated)Interventional2023-04-14Recruiting
A Single-centre Open-label Study in Healthy Adult Volunteers to Determine the Effects of Steady-state TPV/RTV (500 mg/200 mg) on the Single-dose Pharmacokinetics of Rifabutin (MYCOBUTIN®) 150 mg, and the Effects of Single-dose Rifabutin (150 mg) on the St [NCT02251171]Phase 124 participants (Actual)Interventional2003-08-31Completed
Effects of Steady State Tipranavir/Ritonavir or Darunavir/Ritonavir or Ritonavir on Platelet Function, Coagulation and Fibrinolysis Biomarkers in Healthy Subjects [NCT02251795]Phase 152 participants (Actual)Interventional2007-08-31Completed
Immunologic and Virologic Consequences of Long-Term Highly Active Antiretroviral Therapy (HAART) in Subjects With Moderately Advanced HIV-1 Disease: A Follow-Up Study to ACTG 315 [NCT00000891]Phase 234 participants InterventionalCompleted
Tipranavir: An Open-label, Randomized Study Comparing Combination Therapy (Tipranavir and Ritonavir vs. Saquinavir and Ritonavir) Used With Two Nucleoside Reverse Transcriptase Inhibitors in Single Protease Inhibitor-experienced HIV-1 Patients [NCT02239835]Phase 279 participants (Actual)Interventional1999-12-31Terminated
A Study of the Effects of Combination Antiretroviral Therapy in Acute HIV-1 Infection With an Emphasis on Immunological Responses [NCT00001119]288 participants Interventional1999-10-31Completed
Study of Pharmacokinetic Interaction Between TRUVADA™ and BILR 355 BS Plus Ritonavir [NCT02253901]Phase 133 participants (Actual)Interventional2005-03-31Completed
A 48-week, Randomized, Open-label, 2-arm Study to Compare the Efficacy of Saquinavir/Ritonavir Twice Daily (BID) Plus Emtricitabine/Tenofovir Once Daily (QD) Versus Lopinavir/Ritonavir BID Plus Emtricitabine/Tenofovir QD in Treatment-naïve Human Immunodef [NCT00105079]Phase 3337 participants (Actual)Interventional2005-04-30Completed
An Open Label Randomized Clinical Trial, to Evaluate the Treatment With Darunavir/Ritonavir + Lamivudine Once Daily Versus Continuing With Darunavir/Ritonavir Once Daily + Tenofovir/Emtricitabine or Abacavir/Lamivudine in HIV Infected Subject With Suppres [NCT02159599]Phase 4249 participants (Actual)Interventional2014-07-31Completed
A Single Centre, Open-Label, Randomised, Parallel, Multiple Dose Comparison of the Effects of Tipranavir 500 mg and Ritonavir 100 mg or Tipranavir 750 mg and Ritonavir 200 mg Twice a Day for 11.5 Days on the Pharmacokinetic Characteristics of Tenofovir Di [NCT02251145]Phase 149 participants (Actual)Interventional2002-05-31Completed
An Open Label Multinational Study of the Effects of Three Dose Pairs of Tipranavir/Ritonavir (b.i.d.) on the Pharmacokinetic Characteristics of Protocol -Defined, Baseline, Triple Drug Nucleoside and Non-nucleoside Reverse Transcriptase Inhibitor Therapy [NCT02251223]Phase 1/Phase 2208 participants (Actual)Interventional2001-02-28Completed
A Single-centre Open-label Study in Healthy Adult Volunteers to Determine the Effects of Single-dose and Steady-state TPV/r (500 mg/200 mg) on the Steady-state Pharmacokinetics of Clarithromycin (BIAXIN®) 500 mg Bid and a Preliminary Assessment of the Eff [NCT02251769]Phase 124 participants (Actual)Interventional2003-08-31Completed
A PHASE 2/3, INTERVENTIONAL SAFETY, PHARMACOKINETICS, AND EFFICACY, OPEN-LABEL, MULTI-CENTER, SINGLE-ARM STUDY TO INVESTIGATE ORALLY ADMINISTERED PF-07321332 (NIRMATRELVIR)/RITONAVIR IN NONHOSPITALIZED SYMPTOMATIC PEDIATRIC PARTICIPANTS WITH COVID-19 WHO [NCT05261139]Phase 2/Phase 3160 participants (Anticipated)Interventional2022-03-07Recruiting
An Open Study to Investigate the Effect of Two Times Oral 100 mg Ritonavir Capsules on Pharmacokinetics of Single Doses of BILR 355 BS (Dose Steps: 5 and 12.5 mg) Dissolved in 5 mL PEG 400 After Oral Administration in Healthy Male Volunteers, and a Double [NCT02253953]Phase 162 participants (Actual)Interventional2002-11-30Completed
ProSpective, MultI-Center, Observational PrograM to Assess the Effectiveness of Dual TheraPy (Lopinavir/Ritonavir + LamivudinE) in Treatment-Experienced HIV Infected Patients in the Routine Clinical Settings of the Russian Federation (SIMPLE) [NCT02581202]216 participants (Actual)Observational2015-12-21Completed
A Phase I Multiple Oral Dose Trial of Tipranavir 500 mg/Ritonavir 200 mg Dosed to Steady State Followed by Single-dose 14C-radiolabeled Tipranavir Co-administered With Tipranavir 500 mg/Ritonavir 200 mg to Characterize the Excretion Balance and Metabolite [NCT02253797]Phase 10 participants Interventional2003-07-31Completed
A Single-centre, Open-label Study in Healthy Adult Volunteers to Determine the Effects of Multiple-dose Omeprazole (ANTRA® 40 mg qd) on the Single-dose Pharmacokinetics of Tipranavir 500 mg Coadministered With Ritonavir 200 mg [NCT02253875]Phase 115 participants (Actual)Interventional2005-11-30Completed
Study of Pharmacokinetic Interaction Between Tipranavir and BILR 355 BS Plus Ritonavir [NCT02257021]Phase 134 participants (Actual)Interventional2005-02-28Completed
Prophylaxis for HIV-1: Tenofovir/Emtricitabine (Truvada ®) + Lopinavir/Ritonavir (Kaletra ®) vs Tenofovir/Emtricitabine/Cobicistat/Elvitegravir (Stribild ®). Prospective, Randomized, Open. [NCT02198443]Phase 4160 participants (Actual)Interventional2015-06-06Completed
PAxlovid loNg cOvid-19 pRevention triAl With recruitMent In the Community in Norway [NCT05852873]Phase 32,000 participants (Anticipated)Interventional2023-05-12Recruiting
A Phase I/II Trial of Short Course Pre-Operative Ritonavir To Determine Akt Inhibition in Breast Cancer [NCT01009437]Phase 128 participants (Actual)Interventional2010-05-26Completed
A Pilot Study to Assess the Safety, Efficacy, and PK Profile of a Switch in Antiretroviral Therapy to a RTI Sparing Combination of LPV/r and RAL in Virologically Suppressed HIV-infected Patients [NCT00700115]Phase 460 participants (Actual)Interventional2008-06-30Completed
Pilot Assessment of Lopinavir/Ritonavir and Maraviroc in Experienced Patients [NCT00981318]Phase 43 participants (Actual)Interventional2009-12-31Terminated(stopped due to unable to enroll expected number of subjects)
IMPAACT P1058A: Intensive Pharmacokinetic Studies of New Classes of Antiretroviral Drug Combinations in Children, Adolescents and Young Adults [NCT00977756]168 participants (Actual)Observational2002-08-31Completed
A Randomized Trial to Evaluate the Effectiveness of Antiretroviral Therapy Plus HIV Primary Care Versus HIV Primary Care Alone to Prevent the Sexual Transmission of HIV-1 in Serodiscordant Couples [NCT00074581]Phase 33,526 participants (Actual)Interventional2005-02-28Completed
Continued Access to Darunavir/Ritonavir (DRV/Rtv) in HIV-1 Infected Adults, Adolescents and Children Aged 3 Years and Above [NCT01281813]Phase 3145 participants (Actual)Interventional2011-08-08Completed
A Multicenter, Double-Blind, Randomized, Active-Controlled Study to Evaluate the Safety and Antiretroviral Activity of MK0518 Versus KALETRA in HIV-Infected Patients Switched From a Stable KALETRA-Based Regimen - Study A [NCT00443703]Phase 3352 participants (Actual)Interventional2007-05-31Terminated(stopped due to primary efficacy analysis at Week 24 did not demonstrate non-inferiority of raltegravir versus lopinavir (+) ritonavir)
See Detailed Description. [NCT00363142]Phase 3211 participants (Actual)Interventional2006-05-31Completed
Randomised and Prospective Clinical Study to Evaluate the Efficacy and Safety of Lopinavir/Ritonavir Monotherapy Versus Darunavir/Ritonavir Monotherapies as Simplification Switching Strategies of PI/NNRTI-Triple Therapy Based-Regimens [NCT00994344]Phase 473 participants (Actual)Interventional2009-10-31Completed
Randomized, Embedded, Multifactorial Adaptive Platform Trial for Community- Acquired Pneumonia [NCT02735707]Phase 310,000 participants (Anticipated)Interventional2016-04-11Recruiting
"PIQD: The Once a Day Protease Inhibitor Regimens. Ritonavir Boosted Atazanavir vs. Ritonavir Boosted Fosamprenavir Used in Combination With Tenofovir and Emtricitabine in HIV-1 Infected Antiretroviral Treatment-Naïve Patients." [NCT00242216]Phase 476 participants (Actual)Interventional2004-05-31Completed
Phase III Open Label Atazanavir (BMS-232632) in Combination With Ritonavir or Saquinavir, and Lopinavir/Ritonavir, Each With Tenofovir and a Nucleoside in Subjects With HIV [NCT00035932]Phase 3571 participants (Actual)Interventional2001-11-30Completed
A Phase I, 2-panel, Open-label, Randomized, Crossover Trial in Healthy Subjects to Investigate the Pharmacokinetic Interaction Between TMC435 and CYP3A Inhibitors, Erythromycin and Darunavir/Ritonavir (DRV/r) [NCT01323257]Phase 149 participants (Actual)Interventional2011-03-31Completed
A Multicenter, Double-Blind, Randomized, Active-Controlled Study to Evaluate the Safety and Antiretroviral Activity of MK0518 Versus KALETRA in HIV-Infected Patients Switched From a Stable KALETRA-Based Regimen - Study B [NCT00443729]Phase 3355 participants (Actual)Interventional2007-05-31Terminated(stopped due to Primary efficacy analysis at Week 24 did not demonstrate non-inferiority of raltegravir versus lopinavir (+) ritonavir)
Phase I/II, Open-Label, Pharmacokinetic and Safety Study of a Novel Protease Inhibitor (BMS 232632, Atazanavir, ATV, Reyataz) in Combination Regimens in Antiretroviral Therapy (ART)-Naive and -Experienced HIV-Infected Infants, Children, and Adolescents [NCT00006604]Phase 1/Phase 2195 participants (Actual)Interventional2000-11-30Completed
A Randomized, Double-blind, Placebo-controlled, Phase II/III Clinical Study Evaluating the Efficacy and Safety of SSD8432 in Combination With Ritonavir in Adult Subjects With Mild/Common COVID-19 [NCT05373433]Phase 2/Phase 3670 participants (Anticipated)Interventional2022-05-26Not yet recruiting
International Trial of Modified Directly Observed Therapy Versus Self-Administered Therapy for Participants With First Virologic Failure on a Non-Nucleoside Reverse Transcriptase Inhibitor-Containing Antiretroviral Regimen [NCT00608569]529 participants (Actual)Interventional2009-03-31Completed
A Randomized, Open-label, Multi-centre Clinical Trial Evaluating and Comparing the Safety and Efficiency of ASC09/Ritonavir and Lopinavir/Ritonavir for Confirmed Cases of Pneumonia Caused by Novel Coronavirus Infection [NCT04261907]6 participants (Actual)Interventional2020-02-11Terminated(stopped due to There were no more subjects enrolled.)
Pilot Study to Evaluate the Potential Interactions Between Paclitaxel and Protease Inhibitors in Patients With AIDS-Related Kaposi's Sarcoma [NCT00003008]Phase 233 participants (Anticipated)Interventional1997-12-15Completed
A Randomised Open-label Study Comparing the Safety and Efficacy of Ritonavir Boosted Lopinavir and 2-3N(t)RTI Backbone Versus Ritonavir Boosted Lopinavir and Raltegravir in Participants Virologically Failing First-line NNRTI/2N(t)RTI Therapy [NCT00931463]Phase 4558 participants (Actual)Interventional2009-09-30Completed
Clinical Pilot, Open, Comparative and Randomized Trial to Evaluate the Efficacy and Security of Darunavir/Ritonavir 900/100 mg Once a Day as an Antiretroviral Treatment Simplification Strategy [NCT00611039]Phase 430 participants (Actual)Interventional2008-02-29Completed
Multicentric, Non-inferiority, Randomized, Non-blinded Phase 3 Trial Comparing Virological Response at 48 Weeks of 3 Antiretroviral Treatment Regimens in HIV-1-infected Patients With Treatment Failure After 1st Line Antiretroviral Therapy (Cameroon, Burki [NCT00928187]Phase 3454 participants (Actual)Interventional2009-11-30Completed
A Phase 4, Randomized, Open Label, Controlled Study of Boosted Darunavir and Lamivudine Versus Boosted Darunavir and Emtricitabine/Tenofovir or Lamivudine/Tenofovir in Naïve HIV-1 Infected Subjects [NCT02770508]Phase 4145 participants (Actual)Interventional2015-11-30Completed
The Effects of Darunavir Plus Ritonavir on the Pharmacokinetics and Pharmacodynamics of Rosuvastatin [NCT00885495]Phase 1/Phase 217 participants (Actual)Interventional2009-01-31Completed
An International Randomized Trial of Additional Treatments for COVID-19 in Hospitalized Patients Who Are All Receiving the Local Standard of Care Philippines [NCT05024006]1,314 participants (Actual)Interventional2020-04-23Completed
A Prospective Longitudinal Pilot Study to Measure the Effect of Intensification With Raltegravir +/- a Protease Inhibitor (PI) or Non-Nucleoside Reverse Transcriptase Inhibitor (NNRTI) on HIV-1 Levels in the Gut [NCT00884793]8 participants (Actual)Interventional2008-09-30Completed
Lumefantrine Pharmacokinetics When Administered as a Fixed Dose Combination With Artemether in HIV Positive Patients on Lopinavir/Ritonavir [NCT00619944]Phase 432 participants (Anticipated)Interventional2008-02-29Completed
Pharmacokinetics of Low Dose Ritonavir in Thai Patients on a Saquinavir 1500 mg Based HAART Regimen [NCT00622206]Phase 1/Phase 220 participants (Actual)Interventional2008-01-31Completed
TMC125-TiDP2-C238: A Randomized, Exploratory, Open-label 48-week Trial to Investigate the Pharmacokinetics, Safety, Tolerability and Antiviral Activity of Etravirine (ETR) in Combination With Ritonavir-boosted Atazanavir (ATV/Rtv) and 1 NRTI in Treatment- [NCT00896051]Phase 250 participants (Actual)Interventional2009-08-31Completed
Pharmacokinetics of Rifabutin Combined With Antiretroviral Therapy in the Treatment of Tuberculosis Patient With HIV Infection in Vietnam : A Phase II Trial [NCT00651066]Phase 247 participants (Actual)Interventional2010-06-30Completed
A PHASE 1, NON-RANDOMIZED, OPEN-LABEL STUDY TO ASSESS THE PHARMACOKINETICS, SAFETY AND TOLERABILITY OF PF-07321332 BOOSTED WITH RITONAVIR IN ADULT PARTICIPANTS WITH MODERATE HEPATIC IMPAIRMENT AND HEALTHY PARTICIPANTS WITH NORMAL HEPATIC FUNCTION [NCT05005312]Phase 117 participants (Actual)Interventional2021-08-31Completed
A Prospective Randomized Open-Label Clinical Trial to Evaluate the Comparative Efficacy and Safety of a Potent Antiretroviral Treatment Regimen With or Without Hydroxyurea for Subjects With Acute HIV-1 Infection or Recent HIV-1 Seroconversion [NCT00006339]Phase 20 participants (Actual)InterventionalWithdrawn
P1060 Substudy Comparing Differences in Malaria Parasitemia by Real Time Quantitative PCR in HIV-Infected Infants and Children on PI-Based HAART Versus NNRTI-Based HAART [NCT00719602]Early Phase 1105 participants (Actual)Interventional2009-08-31Completed
A Phase I/II Study of Invirase® Boosted With Ritonavir in HIV Infected Infants and Children 4 Months to Less Than 6 Years Old [NCT00623597]Phase 218 participants (Actual)Interventional2008-06-30Completed
A Multicenter, Randomized, Open Label, Pilot Study to Assess the Possibility of Concomitant Treatment of HCV/HIV co Infection With Peg-interferon + Ribavirin, and Lopinavir/r as a Single Antiretroviral Agent. [NCT00866021]Phase 468 participants (Actual)Interventional2008-02-29Completed
A Phase IV-III Comparative, Randomized, Open-label Study to Evaluate the Efficacy for the Recovery of Peripheral Fat (or of the Extremities) of Lopinavir/Ritonavir in Monotherapy Versus Abacavir/Lamivudine and Lopinavir/Ritonavir [NCT00865007]Phase 488 participants (Actual)Interventional2008-12-31Completed
Observational Non-interventional Study About Antiretroviral Combination Treatment With Aptivus in Combination With Low-dose Ritonavir in HIV Type 1 Infected Patients [NCT00531206]65 participants (Actual)Observational2006-08-31Completed
Randomized, Double-blind, Phase Ib Clinical Trial to Evaluate the Safety, Pharmacodynamics, and Pharmacokinetic of SSD8432/ Ritonavir Multiple Doses in Treatment of Adults With Asymptomatic Infection, Mild, and Common Type of COVID-19 [NCT05369676]Phase 1/Phase 232 participants (Actual)Interventional2022-05-12Completed
A Phase II Rolling Arm Master Protocol (PRAM) of Novel Antiretroviral Therapy in Stable Experienced HIV- Infected Children; PRAM-1: ZDV+3TC vs. d4T+Ritonavir vs. ZDV+3TC+Ritonavir; PRAM-1, Step 2: d4T+Nevirapine+Ritonavir; PRAM-1, Step 3: d4T+Indinavir vs [NCT00001083]Phase 2240 participants InterventionalCompleted
A Randomized, Open-Label, Two Arm Trial to Compare the Safety and Antiviral Efficacy of GW433908/Ritonavir QD to Nelfinavir BID When Used in Combination With Abacavir and Lamivudine for 48 Weeks in Antiretroviral Therapy Naive HIV-1 Infected Subjects [NCT00009061]Phase 3624 participants Interventional2000-11-30Active, not recruiting
Multicentric Randomised Controlled Trial Assessing the Efficacy of Two Strategies of Structured Treatment Interruption of Highly Active Antiretroviral Therapy (HAART) Compared With a Continuous HAART in HIV- Infected Adults in Abidjan [NCT00158405]Phase 3840 participants (Actual)Interventional2002-12-31Completed
Pharmacokinetics, Pharmacodynamics, and Safety Profile of Understudied Drugs [NCT04278404]5,000 participants (Anticipated)Observational2020-03-05Recruiting
SPRING: Safety, Efficacy, Pharmacokinetics of tipRanavi/r IN Race/Gender HIV+ Patients Randomized to Therapeutic Drug Monitoring or Standard of Care [NCT00440271]Phase 333 participants (Actual)Interventional2007-02-28Terminated
Randomized, Open-label, 2-period Cross-over Study in Healthy Adults to Evaluate the Effect of Food on Pharmacokinetics, Safety and Tolerability of ABT-450 With Ritonavir [NCT00909311]Phase 18 participants (Actual)Interventional2009-05-31Completed
Pharmacokinetic Interactions Between Buprenorphine/Naloxone and Tipranavir/Ritonavir in HIV-Negative Subjects Chronically Receiving Buprenorphine/Naloxone [NCT00486330]12 participants (Actual)Interventional2006-05-31Completed
Open Label Phase 3b, 48 wk Pilot Study of the Antiviral Efficacy and Tolerability of Combination of PREZISTA/r and TMC125 When Substituted for Enfuvirtide, Current Protease Inhibitor(s) and NNRTI(s) in Antiretroviral Resistant Patients With Viral Suppress [NCT00460746]Phase 310 participants (Actual)Interventional2007-05-31Completed
A Study of the Pharmacokinetics of Atazanavir (ATV)/Ritonavir(RTV) Administered as Part of Highly Active Antiretroviral Therapy (HAART) in HIV-1 Infected Pregnant Women [NCT00326716]Phase 169 participants (Actual)Interventional2006-06-30Completed
An Open-label, Dose-ranging, Proof-of-Concept Study to Evaluate the Safety and Efficacy of Lonafarnib With and Without Ritonavir Boosting in Patients Chronically Infected With Delta Hepatitis (HDV) (LOWR-1) [NCT02430181]Phase 221 participants (Actual)Interventional2014-11-30Completed
GRACE: An Open-label, Multicenter Trial to Compare the Efficacy, Safety, and Tolerability of PREZISTA (Darunavir)/Ritonavir by Gender and Race, When Administered in Combination With an Individually Optimized Background Regimen Over a 48-week Treatment Per [NCT00381303]Phase 3429 participants (Actual)Interventional2006-11-30Completed
A Phase 3, Randomized, Open-Label Study of Lopinavir/Ritonavir (LPV/r) Tablets 800/200 Milligram (mg) Once-Daily (QD) Versus 400/100 mg Twice-Daily (BID) When Coadministered With Nucleoside/Nucleotide Reverse Transcriptase Inhibitors (NRTIs) in Antiretrov [NCT00358917]Phase 3599 participants (Actual)Interventional2006-08-31Completed
Phase IIIb Multicenter, Single Arm, Open-Label Pilot Study to Evaluate the Effectiveness and Safety of Maintenance With Atazanavir/Ritonavir as Single Enhanced Protease Inhibitor Therapy in HIV-Infected Patients Evidencing Virologic Suppression OREY (Only [NCT00337467]Phase 361 participants (Actual)Interventional2006-06-30Completed
A Comparison of the Bioavailability of Rifabutin With and Without Lopinavir/Ritonavir in Healthy Adult Subjects [NCT00743470]Phase 115 participants (Actual)Interventional2008-08-31Terminated
Impact of Menstrual Cycle on Antiretroviral Pharmacokinetics in Healthy Women [NCT00869960]Phase 424 participants (Actual)Interventional2009-03-31Completed
Pharmacokinetic Interactions Between Antiretroviral Agents, Lopinavir/Ritonavir and Efavirenz and Antimalarial Drug Combinations, Artesunate/Amodiaquine and Artemether/Lumefantrine. [NCT00697892]Phase 138 participants (Actual)Interventional2005-07-31Completed
Nucleoside-Sparing Combination Therapy With Lopinavir/Ritonavir (LPV/r) + Raltegravir (RAL) vs. Efavirenz (EFV) + Tenofovir Disoproxil Fumarate + Emtricitabine (TDF/FTC) in Antiretroviral-Naïve Patients [NCT00752856]Phase 251 participants (Actual)Interventional2008-08-26Completed
Pilot Study Of Novel Combination Of Maraviroc + Atazanavir/Ritonavir vs. Atazanavir/Ritonavir + Emtricitabine/Tenofovir For The Treatment Of Naïve HIV-Infected Patients With R5 HIV-1 [NCT00827112]Phase 2129 participants (Actual)Interventional2009-03-31Completed
Prospective, Open Label and Randomized Clinical Trial About Hepatic Security of Antiretroviral Treatment Based on Kaletra Versus Nevirapine in Co-infected HIV/HCV Patients [NCT00661349]Phase 49 participants (Actual)Interventional2008-02-29Terminated(stopped due to It has been impossible to achieve the number of patients defined by protocol)
The Pharmacokinetics of Two Generic Co-formulations of Lopinavir/Ritonavir for HIV Infected Children: a Pilot Study of Lopimune vs. the Branded Product (SURF Study). [NCT00665951]Phase 112 participants (Actual)Interventional2008-09-30Completed
Pharmacokinetics of Efavirenz and Lopinavir Nano-formulations in HIV Negative Healthy Volunteers: an Adaptive Design Study [NCT02631473]Phase 150 participants (Anticipated)Interventional2015-11-30Suspended(stopped due to Study is on hold whilst a grant application for further funding is put together)
Effects of 2 Initial Standard Antiretroviral Combinations Therapies on Lipid Metabolism in ARV-naive HIV-infected Subjects [NCT00759070]Phase 450 participants (Anticipated)Interventional2008-09-30Active, not recruiting
Phase IV, Two-arm, Open-label, Single-centre Randomised Pilot Study to Assess the Feasibility of Immediate or Deferred Switching of HIV-infected Individuals Intolerant of Efavirenz, Ritonavir-boosted Lopinavir or Ritonavir-boosted Darunavir [NCT00765154]Phase 412 participants (Actual)Interventional2008-10-31Terminated(stopped due to Difficulties in recruitment due to a change in the nature of practice.)
A Phase IV, Randomized, Open-label Study of the Tolerability of Once Daily Lopinavir/Ritonavir (LPV/r) Liquid Versus Capsules [NCT00281606]Phase 465 participants (Actual)Interventional2006-02-14Completed
A Phase IIb, 96 Week, Randomized, Open-label Multicenter, Parallel Group, Repeat Dose Study to Evaluate the Safety, Tolerability, PK and Antiviral Effect of Different Doses and Regimens of GW873140 in Combination With Kaletra (Lopinavir and Ritonavir) in [NCT00102778]Phase 2175 participants Interventional2004-12-31Terminated
Pharmacokinetics of Lopinavir Crushed Versus Whole Tablets in Pediatric Patients [NCT00810108]Phase 412 participants (Actual)Interventional2006-06-30Completed
Baricitinib Combined With Antiviral Therapy in Symptomatic Patients Infected by COVID-19: an Open-label, Pilot Study [NCT04320277]Phase 2/Phase 3200 participants (Anticipated)Interventional2020-05-16Not yet recruiting
Bioequivalence Study of Generic GPO Ritonavir Versus Norvir® in Thai Healthy Volunteers [NCT00477126]Phase 116 participants (Actual)Interventional2007-01-31Completed
A Randomized, Open-Label, Six-Period, Drug Interaction Study to Assess Steady-State Plasma Amprenavir (APV) and Raltegravir (RTG) Pharmacokinetics Following Administration of RTG 400 mg BID for 14 Days Alone and in Combination With 14 Days of Either Fosam [NCT00802074]45 participants (Actual)Interventional2008-12-31Completed
A Pilot Efficacy and Safety Trial of Raltegravir Plus Darunavir/Ritonavir for Treatment-Naive HIV-1-Infected Subjects [NCT00830804]Phase 2113 participants (Actual)Interventional2009-04-30Completed
A Single Arm, Open Label Study to Assess the Pharmacokinetics of Darunavir and Ritonavir, Darunavir and Cobicistat, Etravirine, and Rilpivirine in HIV-1 Infected Pregnant Women [NCT00855335]Phase 377 participants (Actual)Interventional2009-04-09Completed
A Randomized, Open-label Study of Lopinavir/Ritonavir 400/100 mg Tablet Twice Daily + Co-formulated Emtricitabine/Tenofovir Disoproxil Fumarate 200/300 mg Once Daily Versus Lopinavir/Ritonavir 400/100 mg Tablet Twice Daily + Raltegravir 400 mg Twice Daily [NCT00711009]Phase 3206 participants (Actual)Interventional2008-07-31Completed
Safety and Efficacy of Lopinavir/Ritonavir in Combination With Raltegravir in HIV-infected Patients [NCT00752037]Phase 430 participants (Actual)Interventional2008-09-30Completed
A Multicenter, Single Arm, Open-Label Study of the Once Daily Combination of Etravirine and Darunavir/Ritonavir As Dual Therapy in Early Treatment-Experienced Patients [NCT01199939]Phase 254 participants (Actual)Interventional2010-05-31Completed
A Phase I, Open-label, Randomized, 2-way Crossover Trial in 40 Healthy Subjects to Investigate the Potential Pharmacokinetic Interactions Between Telaprevir and Darunavir/Ritonavir and Between Telaprevir and Fosamprenavir/Ritonavir at Steady-state. [NCT00775125]Phase 140 participants (Actual)Interventional2008-06-30Completed
See Detailed Description [NCT00440947]Phase 3515 participants (Actual)Interventional2007-03-31Completed
Direct Antiviral Agents for the Treatment of Chronic HCV/HBV Co-infection Patients [NCT02555943]Phase 2/Phase 323 participants (Actual)Interventional2015-02-28Completed
An Open-label, Dose-ranging, Proof-of-Concept Study to Evaluate the Safety and Efficacy of Lonafarnib With Ritonavir-Boosting +/- Peginterferon Alfa-2a in Patients Chronically Infected With Delta Hepatitis (HDV) (LOWR-2) [NCT02430194]Phase 255 participants (Actual)Interventional2014-12-31Completed
Steady State Pharmacokinetics (PK) of Fosamprenavir (FPV) Alone or in Combination With Low Dose Ritonavir (/r) (1400mg BID, 1400mg/100mg QD or 700/100mg BID) and the Chemokine Receptor 5 (CCR5) Entry Inhibitor Maraviroc (MVC) 300mg BID in Healthy Voluntee [NCT00764465]Phase 245 participants (Actual)Interventional2008-10-31Completed
A Study of SCH 900518 in Previously Untreated Subjects With Genotype 1 Chronic Hepatitis C (Protocol No. P05104) [NCT00797745]Phase 2111 participants (Actual)Interventional2008-11-30Completed
Evaluate the Safety, Tolerability, and Antiviral Activity of GLS4 With Ritonavir in Combination With Entecavir in Comparison With Entecavir Alone in Patients With Chronic HBV Who Are HBeAg Positive [NCT04147208]Phase 2250 participants (Anticipated)Interventional2019-02-28Recruiting
Effect of HIV-1 Protease Inhibitors on Endothelial Function and Glucose Metabolism in Normal, HIV-Uninfected Subjects: Atazanavir or Lopinavir/Ritonavir or Placebo [NCT00720590]30 participants (Actual)Interventional2003-11-30Completed
CID 0708 - Sex, Aging and Antiretroviral Pharmacokinetics [NCT00666055]11 participants (Actual)Observational2008-03-31Completed
A Phase I/II Study of Lopinavir/Ritonavir in HIV-1 Infected Infants Less Than 6 Months of Age [NCT00038480]Phase 131 participants (Actual)InterventionalCompleted
Comparing the Efficacy and Safety of Single Versus Double Ritonavir-boosted Protease Inhibitor (PI)-Based Antiretroviral Therapy (ART) Regimens for Children Failing Non Nucleoside Reverse Transcriptase Inhibitor (NNRTI)-Based Treatment [NCT00886990]240 participants (Actual)Observational2007-10-31Completed
A Drug-drug Interaction Study Between the Novel Anti-HCV Agent Daclatasvir and the Antiretroviral Agents Atazanavir/Ritonavir or Atazanavir/Cobicistat in Healthy Volunteers [NCT02565888]Phase 116 participants (Actual)Interventional2015-11-30Completed
A Phase 3, Randomized, Double-Blind Study to Evaluate the Safety and Efficacy of Elvitegravir/Emtricitabine/Tenofovir Disoproxil Fumarate/GS-9350 Versus Ritonavir-Boosted Atazanavir Plus Emtricitabine/Tenofovir Disoproxil Fumarate in HIV-1 Infected, Antir [NCT01106586]Phase 3708 participants (Actual)Interventional2010-04-30Completed
Randomized Controlled Clinical Trials of Lopinavir/Ritonavir or Hydroxychloroquine in Patients With Mild Coronavirus Disease (COVID-19) [NCT04307693]Phase 265 participants (Actual)Interventional2020-03-11Terminated(stopped due to Terminated early because no patients were further enrolled since mid-Apr 2020.)
An Open, Prospective/Retrospective, Randomized Controlled Cohort Study to Compare the Efficacy of Three Antiviral Drugs(Abidol Hydrochloride, Oseltamivir and Lopinavir/Ritonavir) in the Treatment of 2019-nCoV Pneumonia. [NCT04255017]Phase 4400 participants (Anticipated)Interventional2020-02-01Recruiting
Randomized, Non-inferiority Trial Comparing a Dual Maintenance Therapy Strategy With Dolutegravir + Lamivudine (DTG/3TC) or Atazanavir/Ritonavir + Lamivudine (ATV/r+3TC) Versus the Standard WHO First Line Triple Therapy Tenofovir + Lamivudine + Efavirenz [NCT04022967]Phase 3480 participants (Actual)Interventional2020-09-21Active, not recruiting
Investigation of the Effect of Multiple Doses of Danoprevir/Ritonavir on Methadone in Subjects on Stable Methadone Maintenance Therapy (MMT) [NCT01389544]Phase 118 participants (Actual)Interventional2011-06-30Completed
Steady-State Plasma Amprenavir (APV) and Raltegravir (RTG) Pharmacokinetics After Fosamprenavir (FPV) and Raltegravir (RTG) Are Each Administered Alone Versus in Combination With or Without Ritonavir (RTV) Boosting in Healthy Adult Subjects [NCT00614991]44 participants (Actual)Interventional2008-01-31Completed
Phase I, Open Label, Randomized, Drug-Drug Interaction Study in Healthy Subjects to Investigate the Effects of Co-Administered Atazanavir/Ritonavir (300mg/100mg) or Atazanavir 400mg Administered Once Daily on the Steady-State Plasma Pharmacokinetics of GS [NCT00883935]Phase 124 participants (Actual)Interventional2009-04-30Completed
A Single-center, Single-dose, Randomized, Double-blind, Double-dummy, Placebo-controlled, Positive-controlled, Four-way Crossover Study to Investigate the Effect of Danoprevir With Low Dose Ritonavir (DNV/r) on the QT/QTc Interval in Healthy Subjects [NCT01398293]Phase 152 participants (Actual)Interventional2011-08-31Completed
Phase 3, Single Center, Controlled, Investigator-blinded, Randomized Matched Pair Design Study of CD4 Cell Recovery in HIV-1 Patients With Sustained Virologic Response Comparing Protease Inhibitor and Non-nucleoside Reverse Transcriptase Inhibitor Based T [NCT00966160]Phase 3215 participants (Actual)Interventional1999-01-31Completed
A Phase I, Open Label, Randomized, Three Period, One-way, Two Cohort, Adaptive Crossover Study to Evaluate the Effect of Darunavir/Ritonavir Plus Etravirine and Lopinavir/Ritonavir Plus Etravirine on GSK1349572 Pharmacokinetics in Healthy Adult Subjects ( [NCT00867152]Phase 117 participants (Actual)Interventional2009-04-30Completed
Pharmacokinetics and Efficacy of Low- or Standard-dose of Lopinavir/Ritonavir (Kaletra®) in PI-naïve HIV-1 Infected Children [NCT00887120]Phase 224 participants (Actual)Interventional2007-04-30Completed
Low Doses of Lung Radiation Therapy in Cases of COVID-19 Pneumonia: Prospective Multicentric Study in Radiation Oncology Centers [NCT04394182]15 participants (Anticipated)Interventional2020-04-21Suspended(stopped due to lack of recruitment)
A Blinded, Randomized, Placebo-controlled, Dose Ranging Study to Evaluate the Safety, Tolerability, Pharmacokinetics, and Antiviral Activity of Multiple Doses of ABT-450 With Ritonavir (ABT-450/r), ABT-333 or ABT-072 Each Administered Alone and in Combina [NCT01074008]Phase 274 participants (Actual)Interventional2010-03-31Completed
An Open Labe Study to Evaluate the Drug-Drug Interaction of Itraconazole With GST-HG171/Ritonavir in Healthy Adult Chinese Participants [NCT06087055]Phase 112 participants (Actual)Interventional2023-05-12Completed
A Phase II, Open Label Trial, to Evaluate Pharmacokinetics, Safety, Tolerability and Antiviral Activity of DRV in Combination With Low-Dose Ritonavir (DRV/Rtv) in Treatment-Experienced HIV-1 Infected Children From 3 Years to Below 6 Years of Age [NCT00919854]Phase 227 participants (Actual)Interventional2009-09-30Completed
A Blinded, Randomized, Nonfasting, Placebo-Controlled Study in Healthy Adults to Evaluate Safety, Tolerability, and Pharmacokinetic Profiles of Multiple Ascending Doses of ABT-450 With Ritonavir [NCT00931281]Phase 138 participants (Actual)Interventional2009-06-30Completed
Study to Assess the Pharmacokinetic Drug - Drug Interactions Between Atazanavir Plus Ritonavir Coadministered With Voriconazole in Healthy Subjects [NCT00833482]Phase 1185 participants (Actual)Interventional2009-09-30Completed
A Multicenter, Open-label, Randomized Study to Assess the Metabolics, Efficacy, and Safety of Once-daily Darunavir Versus Atazanavir in HIV-infected Treatment-naive Adult Patients [NCT00757783]Phase 468 participants (Actual)Interventional2008-10-31Completed
A Phase I Study of Intra-anally Administered Lopinavir/Ritonavir in People Living With HIV (PLWH) With High-Grade Anal Intraepithelial Neoplasia (AIN 2/3) [NCT05334004]Phase 121 participants (Anticipated)Interventional2024-01-31Recruiting
A Phase I, Single-Centre, Open-Label, Randomized, One-sequence Crossover, Three-Group Study to Evaluate the Effect of Ketoconazole, Ritonavir and Erythromicin on the Safety and Pharmacokinetics of Avanafil (TA-1790) in Healthy Male Subjects [NCT00770042]Phase 144 participants (Actual)Interventional2008-10-31Completed
A Multicenter, Randomized, Open-Label, Active-Controlled Pilot Study to Evaluate the Safety and Antiretroviral Activity of Unboosted Atazanavir BID Plus Raltegravir BID and Boosted Atazanavir QD in Combination With Tenofovir/Emtricitabine QD in Treatment [NCT00768989]Phase 2167 participants (Actual)Interventional2008-11-30Terminated(stopped due to Efficacy endpoint met, but overall experimental dosing regimen not considered optimal to support further clinical development in this population.)
Evolution of Estimated Glomerular Filtration Rate in Chronic Hepatitis C Patients Receiving Sofosbuvir-based or Sofosbuvir-free Direct Acting Antivirals [NCT04047680]441 participants (Actual)Observational2015-02-28Completed
The ARDENT Study: Atazanavir, Raltegravir, or Darunavir With Emtricitabine/Tenofovir for Naive Treatment [NCT00811954]Phase 31,814 participants (Actual)Interventional2009-05-31Completed
Pilot Evaluation of the Influence of ABT450r, Ombitasvir, Dasabuvir +/- Ribavirin HCV Therapy on Insulin Resistance and Lipid Profile [NCT02734173]Phase 424 participants (Actual)Interventional2015-07-31Completed
A Multicenter Study to Evaluate the Pharmacokinetic Profile and Safety of TMC125 Plus Tenofovir DF/Emtricitabine All Dosed Once Daily With and Without Darunavir (PREZISTA™)/ Ritonavir Once Daily in Antiretroviral naïve HIV-1 Infected Subjects [NCT00534352]Phase 223 participants (Actual)Interventional2008-01-31Completed
Treatment Outcomes and Plasma Level of Ritonavir-boosted Lopinavir Monotherapy Among HIV-infected Patients Who Had Non-nucleoside Reverse Transcriptase Inhibitor (NRTI) and NNRTI Failure: A Pilot Study [NCT01002898]Phase 340 participants (Actual)Interventional2007-04-30Completed
A Multicenter, Randomized, Open Label, Clinical Trial to Evaluate Three Doses of Tipranavir Boosted With Ritonavir (500 mg/200 mg qd, 250 mg/100 mg Bid and 500 mg/100 mg Bid) by Assessing the Steady-state Pharmacokinetics and Short-term Efficacy and Safet [NCT00530920]Phase 285 participants (Actual)Interventional2007-10-31Completed
An Open-Label, Parallel Group, Single And Multiple Dose Study To Evaluate The Pharmacokinetics, Safety And Toleration Of Maraviroc Administered To Subjects With Various Degrees Of Renal Impaired And Normal Renal Function [NCT00717067]Phase 430 participants (Actual)Interventional2008-07-31Completed
A Phase 4 Study of the Effect on Immune Reconstitution of a Lopinavir/Ritonavir-Based Versus an Efavirenz-based HAART (Highly Active Antiretroviral Therapy) Regimen in Antiretroviral-Naïve Subjects With Advanced HIV Disease [NCT00775606]Phase 415 participants (Actual)Interventional2008-10-31Terminated(stopped due to Study stopped 12/2010 due to poor enrollment. Only 15 of 60 needed enrolled.)
A Randomized, Open-label Trial to Compare the Efficacy, Safety and Tolerability of DRV/Rtv (800mg/100mg) q.d Versus DRV/Rtv (600mg/100mg) b.i.d in Early Treatment-experienced HIV-1 Infected Subjects [NCT00524368]Phase 3590 participants (Actual)Interventional2007-10-31Completed
A Randomized Study to Evaluate the Safety and Efficacy of IDX719 in Combinations With Simeprevir and/or TMC647055/Ritonavir With or Without Ribavirin for 12 Weeks in Subjects With Chronic Hepatitis C Infection [NCT01852604]Phase 2143 participants (Actual)Interventional2013-03-31Completed
Efavirenz and Ritonavir Influence on Human Brain Levo-acetylmethadol (LAAM) Disposition Assessed Using PET Imaging [NCT01935830]Early Phase 110 participants (Actual)Interventional2013-08-31Completed
A Double-Blind, Randomized, Placebo-Controlled, Nonfasting Study in Healthy Adults to Evaluate the Safety, Tolerability, and Pharmacokinetic Profiles of Single Doses of ABT-450 With and Without Ritonavir [NCT00850044]Phase 187 participants (Actual)Interventional2009-02-28Completed
A Phase II Exploratory Study Examining Immunologic and Virologic Indices in Two Age-Differentiated Cohorts of HIV-Infected Subjects to Explore the Basis of Accelerated HIV-Disease Progression Associated With Aging [NCT00006144]Phase 290 participants Interventional2000-10-31Completed
A Study of the Combination of Indinavir, Ritonavir, Enteric-Coated ddI and d4T In Nucleoside and Non-Nucleoside Reverse Transcriptase Inhibitor Experienced Patients: An Open-Label Study Investigating Differences Between Women and Men [NCT00006397]Phase 3200 participants Interventional2000-08-31Active, not recruiting
A Randomized Trial of the Efficacy and Safety of a Strategy of Starting With Nelfinavir Versus Ritonavir Added to Background Antiretroviral (AR) Nucleoside Therapy in HIV-Infected Individuals With CD4+ Cell Counts Less Than or Equal to 200/mm3 [NCT00000859]1,300 participants InterventionalCompleted
Activity of the Soft Gelatin Capsule of Saquinavir (SQVsgc) in Combination With Ritonavir or Nelfinavir and Combinations of Delavirdine and/or Adefovir Dipivoxil in HIV-Infected Subjects With Prior Indinavir Use and Viral Loads From 2,000 to 200,000 Copie [NCT00000892]300 participants InterventionalCompleted
A Prototype Study to Test the Effect of Staggered Dosing on the Pharmacokinetic Interactions Between Paired Combinations of Nelfinavir (NFV), Ritonavir (RTV), and Soft Gelatin Capsule of Saquinavir (SQVsgc) [NCT00000898]18 participants InterventionalCompleted
Study of Protease Inhibitor and/or Non-Nucleoside Reverse Transcriptase Inhibitor With Dual Nucleosides in Initial Therapy of HIV Infection [NCT00000919]900 participants InterventionalCompleted
A Phase I/II Study of Ritonavir Therapy in HIV-1 Infected Infants and Children [NCT00000952]Phase 160 participants InterventionalCompleted
A Pilot Study to Evaluate the Immunologic Consequences of a Highly Active Antiretroviral Therapy Regimen (HAART) Consisting of Ritonavir (ABT-538), Zidovudine (AZT), and Lamivudine (3TC) in Moderately Advanced HIV-1 Disease [NCT00001075]55 participants InterventionalCompleted
A Phase I/II Double-Blind Controlled Trial to Determine the Safety and Immunogenicity of HIV-1 MN rgp160 Immuno AG Vaccine Therapy in HIV-Infected Individuals With Greater Than or Equal to 500/mm3 CD4+ T Cells and 200-400/mm3 CD4+ T Cells [NCT00000822]Phase 146 participants InterventionalCompleted
A Phase I Trial of the Safety, Tolerance, and Pharmacokinetics of Oral Ritonavir Co-Administered With Lamivudine (3TC) and Zidovudine (ZDV) in HIV-1-Infected Pregnant Women and Their Infants [NCT00000888]Phase 114 participants InterventionalCompleted
The Effect of HIV Protease Inhibitors on the Stereospecific Metabolism of Methadone in HIV-Infected Subjects [NCT00000906]Phase 112 participants InterventionalCompleted
An Open-Label Study of a Once Daily Dose of Emtricitabine in Combination With Other Antiretroviral Agents in HIV-Infected Pediatric Patients [NCT00017992]Phase 2100 participants InterventionalRecruiting
The Effect of FOsamprenavir/Ritonavir on the Pharmacokinetics of a Single-dose of the Antipsychotic Agent olanZApine (FORZA) [NCT00977301]Phase 124 participants (Anticipated)Interventional2009-11-30Completed
A Phase III, Randomized, Controlled, Open-Label, Multicentre, Three Arm Study to Compare the Efficacy and Safety of a Dual-Boosted HIV-1 Protease Inhibitor Regimen of Fosamprenavir/Lopinavir/Ritonavir 1400mg/533mg/133mg Twice Daily and an Increased Dosage [NCT00144833]Phase 3150 participants (Anticipated)Interventional2005-03-31Terminated(stopped due to Incomplete data)
Pilot Simplification Study to Lopinavir/Ritonavir 800/200 mg Monotherapy Regimen Once Daily [NCT01581853]Phase 421 participants (Actual)Interventional2012-05-31Completed
Once-daily Antiretroviral Therapy in HIV-1 Infected Patients With CD4+ Cell Counts Below 100 Cells/Mcl. A Prospective, Randomized, Multicentre, Open Clinical Study. [NCT00532168]Phase 4108 participants (Actual)Interventional2007-09-30Completed
Pharmacokinetics of Darunavir/Ritonavir Once Daily and Atazanavir/Ritonavir Once Daily Over 72 Hours Following Drug Intake Cessation [NCT01073761]Phase 125 participants (Anticipated)Interventional2010-04-30Completed
Prevention of HIV1 Mother to Child Transmission Without Nucleoside Analogue Reverse Transcriptase Inhibitors in the Pre-partum Phase. A Multicenter Randomised Phase II/III Open Label Study With a Group of 100 Pregnant Women Receiving Lopinavir/Ritonavir a [NCT00424814]Phase 2/Phase 3105 participants (Actual)Interventional2007-03-31Completed
Optimisation of Primary HIV1 Infection Treatment (ANRS 147 OPTIPRIM) [NCT01033760]Phase 390 participants (Actual)Interventional2010-04-30Completed
A Phase 3, Randomized, Open-label, Study of Lopinavir/Ritonavir Tablets Versus Soft Gel Capsules and Once Daily Versus Twice Daily Administration, When Coadministered With NRTIs in Antiretroviral Naive HIV-1 Infected Subjects [NCT00262522]Phase 3664 participants (Actual)Interventional2005-11-30Completed
Comprehensive in Vitro Proarrhythmia Assay (CiPA) Clinical Phase 1 ECG Biomarker Validation Study (CiPA Phase 1 ECG Biomarker Study) [NCT03070470]Phase 160 participants (Actual)Interventional2017-03-14Completed
A Phase IV, Open-Label, Randomized, Multicenter Trial Assessing the Efficacy of a Treatment Maintenance Phase With Unboosted vs. Boosted Reyataz After an Induction Phase With Reyataz and Ritonavir in Treatment Naive HIV Patients (the INDUMA Study) [NCT00207142]Phase 4252 participants (Actual)Interventional2005-11-30Completed
A Pilot Study of Lopinavir/Ritonavir in Participants Experiencing Virologic Relapse on NNRTI-Containing Regimens [NCT00357552]123 participants (Actual)Interventional2008-01-31Completed
Nucleosides And Darunavir/Dolutegravir In Africa (NADIA): a Randomised Controlled Trial of Darunavir Versus Dolutegravir and Tenofovir Versus Zidovudine in Second-line Antiretroviral Therapy Regimens for the Public Health Approach in Sub-Saharan Africa [NCT03988452]Phase 3465 participants (Actual)Interventional2019-07-30Active, not recruiting
A Randomised, Open-label, 96-week Study Comparing the Safety and Efficacy of Three Different Combination Antiretroviral Regimens as Initial Therapy for HIV Infection. [NCT00335322]Phase 4329 participants (Actual)Interventional2007-02-28Completed
A Phase 4, Single Arm, Open Label, Pilot Study of Maraviroc (Celsentri) in Combination With Raltegravir and Darunavir/Ritonavir for the Treatment of Triple Class Failure in Adult HIV-1 Infected Patients. [NCT01013987]Phase 460 participants (Anticipated)Interventional2010-02-28Recruiting
An Open-label Randomised Two-year Trial Comparing Two First-line Regimens in HIV-infected Antiretroviral naïve Subjects: Darunavir/r + Tenofovir/Emtricitabine vs. Darunavir/r + Raltegravir (ANRS 143/NEAT 001) [NCT01066962]Phase 3800 participants (Actual)Interventional2010-08-31Completed
An Open-Label Phase III Study to Assess the Long Term Safety Profile of GW433908 Containing Regimens in HIV-1 Infected Subjects [NCT00296504]Phase 3753 participants (Actual)Interventional2001-11-30Completed
A Pilot Study of Atazanavir/Ritonavir/Efavirenz as a Nucleoside Sparing Regimen [NCT00135343]Phase 360 participants Interventional2004-04-30Completed
A Phase I, Partially Randomized, Open Label, Two-way, Two Period Cross-over Study to Investigate the Pharmacokinetic Interaction Between Etravirine or Darunavir/Rtv and Artemether/Lumefantrine at Steady-state in Healthy HIV-negative Subjects [NCT01876966]Phase 133 participants (Actual)Interventional2011-03-31Completed
An Open Label, Randomized Study to Compare Antiretroviral Therapy (ART) Initiation When CD4 is Between 15% to 24% to ART Initiation When CD4 Falls Below 15% in Children With HIV Infection and Moderate Immune Suppression [NCT00234091]Phase 3300 participants (Actual)Interventional2006-04-30Completed
A Two-way Pharmacokinetic Interaction Study of Single-Dose Atorvastatin (LIPITOR®) With Tipranavir/Ritonavir (500mg/200mg) at Steady-State and the Effect of Antacid (MAALOX®) on the Pharmacokinetics of Single-Dose Tipranavir/Ritonavir (500mg/200mg) in Hea [NCT02245269]Phase 123 participants (Actual)Interventional2003-07-31Completed
The Influence of Cobicistat or Ritonavir on Dabigatran Pharmacokinetics and Pharmacodynamics in Healthy Volunteers [NCT01896622]Phase 140 participants (Actual)Interventional2013-06-18Completed
A Prospective, Open-label Trial of Two Abacavir/Lamivudine Based Regimen (ABC/3TC + Darunavir/Ritonavir or ABC/3TC + Raltegravir) in Late Presenter naïve Patients (With CD4 Count <200 Cells/µL - Advanced HIV Disease) [NCT01900106]Phase 347 participants (Actual)Interventional2013-11-30Completed
Switching HIV-positive Women on Tenofovir/Emtricitabine Plus Boosted Atazanavir to RALtegravir Plus Boosted ATazanavir: A Pilot Randomized Clinical Trial Investigating 48-weeks Changes in Bone Mineral Density [NCT01902186]Phase 44 participants (Actual)Interventional2014-09-30Terminated(stopped due to Low enrollment)
Rosuvastatin Versus Protease Inhibitor Switching for Hypercholesterolaemia in HIV-infected Adults [NCT01935674]Phase 421 participants (Actual)Interventional2013-09-30Completed
A Phase Ib Study of Extending Relugolix Dosing Intervals Through Addition of the CYP3A4 and Pg-P Inhibitor Itraconazole or Ritonavir in Prostate Cancer Patients [NCT05679388]Phase 1100 participants (Anticipated)Interventional2023-02-13Recruiting
Boosted Atazanavir and Truvada Given Once-Daily (BATON Study): A Phase 4 Study of Safety, Efficacy & Adherence in HIV Infected, Antiretroviral Naïve Subjects Treated With a Simple Once-Daily Regimen [NCT00224445]Phase 4100 participants (Actual)Interventional2005-09-30Completed
Optimizing Antiretroviral Therapy in HIV-Infected Children and Adolescents [NCT00207948]4 participants (Actual)Observational2004-11-30Terminated(stopped due to no measurable response was detected at the 50% increase threshold.)
Study on Safety and Efficacy of Salvage Therapy With Amprenavir, Lopinavir and Ritonavir 200 Mg/d or 400 Mg/d in HIV-Infected Patients in Virological Failure.ANRS 104 PUZZLE 1 [NCT00196625]Phase 2100 participants Interventional2000-11-30Completed
A Pharmacokinetic Study to Assess the Inter-patient Variability of Indinavir Drug Levels When Boosted With Ritonavir in Thai Patients on Highly Active Antiretroviral Therapy [NCT00197639]19 participants (Actual)Observational2006-09-30Completed
A Randomized, Phase II, Open Label Study to Compare Twice Daily and Once Daily Potent Antiretroviral Therapy and to Compare Self-Administered Therapy and Therapy Administered Under Direct Observation [NCT00036452]Phase 2402 participants (Actual)InterventionalCompleted
A Phase I, Randomized, Open-label Study to Evaluate the Effect of Ritonavir or Omeprazole on the Pharmacokinetics of IDX719 in Healthy Subjects [NCT01813552]Phase 124 participants (Actual)Interventional2013-02-28Completed
MULTICENTRE STUDY TO ASSESS CHANGES IN BONE MINERAL DENSITY OF THE SWITCH FROM PROTEASE INHIBITORS TO DOLUTEGRAVIR IN HIV-1-INFECTED SUBJECTS WITH LOW BONE MINERAL DENSITY [NCT01966822]Phase 375 participants (Actual)Interventional2014-01-31Completed
Effect of HIV Protease Inhibitor Drugs on Glucose and Insulin Metabolism [NCT00135434]Phase 125 participants Interventional2004-09-30Completed
An Open-Label, Sequential, 3-Period Study to Evaluate Pharmacokinetics of Coadministered Raltegravir (Isentress) and Lopinavir-Ritonavir (Kaletra) in Healthy Adults [NCT00564772]Phase 415 participants (Actual)Interventional2007-11-30Completed
Phase 3 Randomized Trial Evaluating the Virological Efficacy and the Tolerance of 4 New Simplified Antiretroviral Treatments in Naive HIV-1 Infected Patients in Dakar and Yaounde [NCT00573001]Phase 3120 participants (Actual)Interventional2008-07-31Completed
Evaluation of Clinical Response and Safety in HIV Positive Subjects Co-infected With Hepatitis C Treated With a Kaletra Containing HAART Regimen [NCT00234975]Phase 486 participants (Actual)Interventional2002-10-31Completed
A Randomized, Open-Label, Phase III Study of ABT-378/Ritonavir in Combination With Nevirapine and Two Nucleoside Reverse Transcriptase Inhibitors (NRTIs) vs. Investigator Selected Protease Inhibitor(s) in Combination With Nevirapine and Two NRTIs in Antir [NCT00004581]Phase 3300 participants InterventionalCompleted
A Phase II/III Trial of Lopinavir/Ritonavir Dosed According to the WHO Pediatric Weight Band Dosing Guidelines [NCT01172535]Phase 2/Phase 397 participants (Actual)Interventional2010-11-30Completed
Long-term Effectiveness and Safety in Hepatitis-co-infected Patients [NCT01153269]33 participants (Actual)Observational2001-05-31Completed
Treatment Options for Protease Inhibitor-exposed Children [NCT01146873]Phase 3300 participants (Actual)Interventional2010-07-31Completed
[NCT02023450]Early Phase 120 participants (Anticipated)Interventional2013-12-31Recruiting
Efficiency and Safety of Paxlovid for the Treatment of COVID-19 Patients With Severe Chronic Kidney Disease [NCT05938140]Phase 430 participants (Anticipated)Interventional2023-06-22Recruiting
A Randomized, Pilot Study on the Antiviral Activity and Immunological Effects of Lopinavir/Ritonavir vs. Efavirenz in Treatment-naïve HIV-Infected Patients With CD4 Cell Counts Below 100 Cells/mm3 [NCT00386659]Phase 460 participants InterventionalTerminated
The Safety and Efficacy of a Ritonavir-Enhanced Agenerase Regimen as Salvage Therapy in HIV-Infected Individuals [NCT00006591]0 participants InterventionalActive, not recruiting
Randomized, Controlled, Open Label, Multi-Center Phase III Trial Comparing the Safety and Antiviral Activity of a Protease-Containing Regimen (d4T/ddI/IDV/RTV) Versus a Protease-Sparing Regimen (d4T/ddI/EFV) and the Ability of Interleukin-2 to Purge HIV F [NCT00006154]Phase 3165 participants (Anticipated)InterventionalCompleted
A Phase II Trial to Evaluate the Safety and Efficacy of Induction Treatment With Lamivudine Plus Stavudine Plus Abacavir Plus Amprenavir/Ritonavir Followed by Supervised Treatment Interruption in Subjects With Acute HIV Infection or Recent Seroconversion [NCT00000940]Phase 2121 participants (Actual)Interventional1999-05-31Completed
Comparison of the Virologic Efficacy of Nelfinavir and/or DMP 266 (Efavirenz, EFV) in Combination With One or Two New Nucleoside Analogs in Nucleoside Experienced Subjects: A Roll-Over Study to ACTG 302/303 [NCT00001087]Phase 2300 participants InterventionalCompleted
Safeguard the Household: A Study of HIV Antiretroviral Therapy Treatment Strategies Appropriate for a Resource Poor Country [NCT00080522]813 participants Interventional2005-02-28Completed
Evaluation of Kaletra Therapy Over the Long-term [NCT01083810]284 participants (Actual)Observational2001-06-30Completed
Treatment Outcome of Children With HIV Infection [NCT00476606]1,000 participants (Anticipated)Observational2003-03-31Active, not recruiting
Randomized, Double-blinded, Controlled Trial of Intensive HAART Including Raltegravir, and Maraviroc, on HIV-1 Pro-viral DNA and Reservoir Decay in HIV-1-infected Individuals During the Acute/Early Infection [NCT01154673]Phase 2/Phase 332 participants (Actual)Interventional2011-11-30Completed
A Multi-centre, Double-blinded, Randomized, Placebo-controlled Trial on the Efficacy and Safety of Lopinavir / Ritonavir Plus Ribavirin in the Treatment of Severe Acute Respiratory Syndrome [NCT00578825]340 participants (Anticipated)InterventionalNot yet recruiting
An Open-Label, Multicenter Study to Evaluate Long-Term Outcomes With ABT-450/Ritonavir/ ABT-267 (ABT-450/r/ABT-267) and ABT-333 With or Without Ribavirin (RBV) in Adults With Genotype 1 Chronic Hepatitis C Virus (HCV) Infection (TOPAZ-I) [NCT02219490]Phase 31,596 participants (Actual)Interventional2014-10-30Completed
Study on the Impact of Triptolide Woldifiion on HIV-1 Reservoir of Chinese HIV/AIDS Patients In Acute HIV-1 Infection [NCT02219672]Phase 318 participants (Anticipated)Interventional2014-07-31Recruiting
Effect of Atazanavir-ritonavir on the Pharmacokinetics and Toxicity of Lumefantrine in People Living With HIV Attending Lagos University Teaching Hospital [NCT04531072]Phase 420 participants (Actual)Interventional2018-09-18Completed
A Multicenter, Randomized, Double-blind, Placebo-controlled, Phase Ⅱ/Ⅲ Clinical Study to Evaluate the Efficacy and Safety of GST-HG171/Ritonavir in Patients With Mild to Moderate COVID-19 [NCT05656443]Phase 2/Phase 31,246 participants (Actual)Interventional2022-12-19Completed
A PHASE 1, RANDOMIZED, FIXED SEQUENCE, MULTIPLE-DOSE, OPEN-LABEL STUDY TO ESTIMATE THE EFFECT OF NIRMATRELVIR (PF-07321332)/RITONAVIR ON ROSUVASTATIN PHARMACOKINETICS IN HEALTHY ADULT PARTICIPANTS [NCT05898672]Phase 112 participants (Actual)Interventional2023-06-09Completed
General Investigation for PAXLOVID PAC [NCT05263908]3,346 participants (Actual)Observational2022-03-31Completed
A Phase IV Open-label Evaluation of Safety, Tolerability and Patient Acceptance of Atazanavir Boosted With Ritonavir Combined With a Fixed-dose Formulation of Tenofovir DF and Emtricitabine for Non-occupational Prophylaxis Following Potential Exposure to [NCT01602822]Phase 411 participants (Actual)Interventional2012-02-29Terminated(stopped due to Grade 3 elevation in liver function tests)
A Prospective Single Arm, Open-label, International, Multicenter Study to Evaluate the Safety, Efficacy and Pharmacokinetics of Atazanavir (ATV) Powder Boosted With Ritonavir (RTV) With an Optimized NRTI Background Therapy, in Human Immunodeficiency Virus [NCT01335698]Phase 3160 participants (Actual)Interventional2011-05-27Completed
An Open-Label, Randomized Study Evaluating a Switch From a Regimen of Two Nucleoside Reverse Transcriptase Inhibitors Regimen Plus Any Third Agent to Either a Regimen of Atazanavir/Ritonavir Once Daily and Raltegravir Twice Daily or to a Regimen of Atazan [NCT01332227]Phase 4132 participants (Actual)Interventional2011-10-31Completed
A Prospective Single Arm, Open-label, International, Multicenter Study to Evaluate the Safety, Efficacy and Pharmacokinetics of Atazanavir (ATV) Powder Boosted With Ritonavir (RTV) With an Optimized NRTI Background Therapy, in HIV Infected Pediatric Patie [NCT01099579]Phase 382 participants (Actual)Interventional2010-10-13Completed
Clinical, Virological and Safety Outcomes of a Lopinavir/Ritonavir-Based Regimen in HIV-1 Infected Patients in Routine Clinical Use in China: A Multicenter Post-Marketing Observational Study [NCT01074931]98 participants (Actual)Observational2008-04-30Completed
A Randomized, Controlled Trial of Two Potent, Simplified Regimens Utilizing A Protease Inhibitor-Sparing Regimen Versus A Nucleoside-Sparing Regimen for HIV-Infected Subjects Who Participated in ACTG 388 or Who Responded to A First Potent Combination Regi [NCT00014937]240 participants InterventionalCompleted
A Phase IV, Open Label Study in Healthy Male Subjects to Investigate the Extent of Darunavir/Ritonavir and Etravirine Exposure in Blood, Seminal Fluid, and Rectal Mucosal Tissue Following Single and Multiple Dosing of Darunavir/Ritonavir and Etravirine [NCT00855088]Phase 113 participants (Actual)Interventional2009-07-31Completed
Safety and Efficacy of the Universal Use of EFV-TDF-FTC and AZT-3TC-LPV/r Combinations in Pregnant and Breastfeeding Women to Prevent mother-to Child Transmission of HIV-1 o, Resource-limited Settings: A Multicentre Randomized Phase 3 Clinical Trial [NCT00936195]Phase 30 participants (Actual)Interventional2010-01-31Withdrawn(stopped due to faillure to obtain insurance because of refusal from insurance companies)
Study to Evaluate the Exposure of Rifabutin Administered in an Alternate Regimen in Combination With Atazanavir and Ritonavir Healthy Subjects [NCT00646776]Phase 185 participants (Actual)Interventional2008-04-30Completed
Therapeutic Drug Monitoring of the Generic Lopinavir/Ritonavir Tablets 200/50 mg in the Thai HIV-infected Patient [NCT00802334]Phase 270 participants (Actual)Interventional2008-01-31Completed
A Phase I, Open-label, Single Sequence, Crossover Study Evaluating the Safety and the Pharmacokinetics of Lopinavir/Ritonavir and Eltrombopag Given Alone and When Co-administered in Healthy Adult Subjects. [NCT00833378]Phase 140 participants (Actual)Interventional2009-01-19Completed
Open-label, Randomised, Drug Interaction Study of Pyramax (Pyronaridine Artesunate) and the Protease Inhibitor Ritonavir in Healthy Volunteers [NCT01156389]Phase 134 participants (Actual)Interventional2010-07-31Completed
Pilot Study for the Evaluation of the Safety and the Feasibility of Treatment Simplification to Atazanavir/Ritonavir + Lamivudine in Patients Stably Treated With Two NRTIs + Atazanavir/Ritonavir With Optimal Virologic Response. [NCT00885482]Phase 440 participants (Actual)Interventional2009-05-31Completed
Cardiovascular, Anthropometric, and Skeletal Effects of Antiretroviral Therapy (ART) Initiation With Emtricitabine/Tenofovir Disoproxil Fumarate (FTC/TDF) Plus Atazanavir/Ritonavir (ATV/r), Darunavir/Ritonavir (DRV/r), or Raltegravir (RAL): Metabolic Subs [NCT00851799]334 participants (Actual)Observational2009-06-30Completed
Evaluation of Safety and Efficacy of Raltegravir/Darunavir Combination in Antiretroviral-Naive Patients [NCT00677300]Phase 485 participants (Actual)Interventional2009-01-31Completed
Pharmacokinetics of the Tablet Formulation of Lopinavir/r as Standard and Increased Dosage During Pregnancy in HIV-infected Women [NCT00605098]Phase 460 participants (Actual)Interventional2008-02-29Completed
A Randomized,Open,Controlled Small Sample Clinical Study to Evaluate the Efficacy and Safety of ASC09/Ritonavir Compound Tablets and Ritonavir for 2019-nCoV Pneumonia [NCT04261270]Phase 360 participants (Anticipated)Interventional2020-02-01Recruiting
A Proof-of-concept Clinical Trial Assessing the Safety of the Coordinated Undermining of Survival Paths by 9 Repurposed Drugs Combined With Metronomic Temozolomide (CUSP9v3 Treatment Protocol) for Recurrent Glioblastoma [NCT02770378]Phase 1/Phase 210 participants (Actual)Interventional2016-11-30Completed
The Pharmacokinetics and Safety of Generic Lopinavir/Ritonavir (200/50 mg Tablets) 400/100 mg q12h in Thai HIV-infected Pregnant Women [NCT00621166]Phase 220 participants (Actual)Interventional2008-06-30Completed
The Impact of Co-infections on Inflammation in Patients Commencing Second-line Antiretroviral Therapy. A Sub-study of D²EFT (Dolutegravir and Darunavir Evaluation in Adults Failing Therapy) [NCT04183738]Phase 40 participants (Actual)Interventional2021-02-01Withdrawn(stopped due to In the context of COVID-19 pandemic.)
A Post Marketing Surveillance Study Assessing the Long-term Efficacy and Safety of Tipranavir (Aptivus®) Co-administered With Low-dose Ritonavir in Treatment Experienced Patients With HIV-1 Infection in the Daily Clinical Practice. [NCT00976950]42 participants (Actual)Observational2009-09-30Completed
Maraviroc Plus Darunavir/Ritonavir Study for Treatment-Naïve Patients Infected With R5-tropic HIV-1 Based on Enhanced Sensitivity Trofile [NCT00993148]Phase 225 participants (Actual)Interventional2010-05-31Completed
Prospective Trial to Evaluate How Therapeutic Drug Monitoring of Protease Inhibitors Increases Virologic Success and Tolerance of HAART (ANRS 111 COPHAR2) [NCT00122590]115 participants Interventional2002-07-31Terminated
Breastfeeding Version of the PROMISE Study (Promoting Maternal and Infant Survival Everywhere) [NCT01061151]Phase 33,747 participants (Actual)Interventional2011-03-01Completed
A Randomised Prospective Study Assessing Changes in Neurocognitive Function, Using a Computerised Test Battery, in Treatment Naïve HIV-1 Positive Subjects Commencing Two Different Antiretroviral Regimens [NCT00540137]Phase 421 participants (Actual)Interventional2007-07-31Completed
A Phase I/II Trial of Ritonavir and Indinavir in Children Failing Other Antiretroviral Therapy [NCT00012519]Phase 1/Phase 229 participants (Actual)InterventionalCompleted
A Phase IIIb/IV, Randomized, Open Label, Multicenter, Pilot Trial to Explore the Safety and Tolerability of GW433908 +/- Ritonavir (1400mg Twice Daily or 700mg/100mg Twice Daily) When Used in Combination With a Zidovudine-containing Regimen (TRIZIVIR or C [NCT00043888]Phase 360 participants Interventional2002-01-31Completed
A Study to Evaluate the Effects of Two Different Meal Types, Omeprazole and Ranitidine on Danoprevir Pharmacokinetics When Coadministered With Ritonavir in Healthy Subjects [NCT01392755]Phase 132 participants (Actual)Interventional2011-07-31Completed
Impact of Menstrual Cycle on Antiretroviral Pharmacokinetics in HIV-Infected Women [NCT01394133]0 participants (Actual)Observational2011-07-31Withdrawn(stopped due to Lack of enrollment)
A Study to Investigate the Effect of Multiple Doses of Danoprevir/Ritonavir (DNV/RTV) on the Pharmacokinetics (PK) of Escitalopram (ESC) and S-Demethylcitalopram (S-DCT) in Healthy Subjects [NCT01418274]Phase 120 participants (Actual)Interventional2011-08-31Completed
A Randomized, Prospective Study of the Efficacy, Safety and Tolerability of Two Doses of GW433908Ritonavir Given With Abacavir/Lamivudine Fixed Dose Combination [NCT00335270]Phase 4100 participants Interventional2006-03-31Completed
Open-Label, Multiple-Dose, Drug Interaction Study to Assess the Effect of Nevirapine on the Pharmacokinetics of Atazanavir in HIV-Infected Individuals [NCT00162149]Phase 146 participants Interventional2005-10-31Completed
Analysis of Lipodystrophy in HIV-Infected Individuals A Prospective, Non-randomised, 48 Week Study of the Effect of PI Containing and Non-PI Containing Antiretroviral Regimens on the Expression of Adipocyte Specific Genes, Protein Levels and Cellular Stru [NCT00192660]Phase 480 participants (Actual)Interventional2003-02-28Completed
Bioequivalence Study of Atazanavir Single 300 mg Capsule Relative to Two Atazanavir 150 mg Capsules in Healthy Subjects [NCT00393328]Phase 146 participants Interventional2006-11-30Completed
Finding Treatments for COVID-19: A Phase 2 Multi-centre Adaptive Platform Trial to Assess Antiviral Pharmacodynamics in Early Symptomatic COVID-19 (PLATCOV) [NCT05041907]Phase 23,000 participants (Anticipated)Interventional2021-09-30Recruiting
A Prospective, Open-Label, Pilot Trial of Regimen Simplification to Atazanavir/Ritonavir Alone as Maintenance Antiretroviral Therapy After Sustained Virologic Suppression [NCT00084019]36 participants (Actual)Interventional2004-07-31Completed
A Phase I/II Safety, Tolerability, and Pharmacokinetic Study of High Dose Lopinavir/Ritonavir With or Without Saquinavir in HIV-Infected Pediatric Subjects Previously Treated With Protease Inhibitors [NCT00084058]Phase 1/Phase 226 participants (Actual)InterventionalCompleted
A Phase III Randomized Trial of the Safety and Antiretroviral Effects of Zidovudine/Lamivudine/Abacavir Versus Zidovudine/Lamivudine/Lopinavir/Ritonavir in the Prevention of Perinatal Transmission of HIV [NCT00086359]Phase 319 participants (Actual)Interventional2004-07-31Completed
A Phase II, Restrictively Randomized, Open-Label, Pilot Study of Treatment Intensification of Early Virologic Failure [NCT00006152]Phase 242 participants InterventionalCompleted
A Study to Evaluate the Potential Drug-Drug Interaction Between Danoprevir/Low-Dose Ritonavir When Given With Raltegravir in Healthy Adult Volunteers [NCT01531647]Phase 120 participants (Actual)Interventional2012-01-31Completed
Safety, Tolerability, and Pharmacokinetic Interactions of Atazanavir and Rifampin in Healthy Volunteers [NCT00096850]18 participants (Actual)InterventionalCompleted
A Randomized, Open-Label Study to Evaluate 3 Salvage Regimens in HIV-Infected Subjects Experiencing Virologic Failure on an Initial HAART Regimen Containing Nelfinavir [NCT00006326]60 participants Interventional2000-08-31Terminated
Disulfiram Interactions With HIV Medications: Clinical Implications [NCT00878306]Phase 140 participants (Actual)Interventional2008-11-30Completed
Efficacy and Safety of Interferon Based Therapy and Interferon-free Direct-acting Antivirals in Cirrhotic Patients With Hepatitis C. Impact of Antiviral Therapy on Gastroesophageal Varices. [NCT02758509]237 participants (Actual)Observational2010-01-01Completed
Relation Between Darunavir Levels and Virological Efficacy, Integrated Proviral ADN and Resistance Mutations in HIV-infected Patients on Treatment With Darunavir/Ritonavir Monotherapy [NCT01606722]150 participants (Actual)Observational2010-01-31Completed
An Open-Label Study to Evaluate Long-Term Outcomes With Ombitasvir-Paritaprevir-Ritonavir and Dasabuvir With or Without Ribavirin (RBV) in Fertile Women With Genotype 1 and 4 Chronic Hepatitis C Virus (HCV) Infection [NCT02950870]Phase 475 participants (Anticipated)Interventional2016-12-31Not yet recruiting
A PHASE 1, OPEN-LABEL STUDY TO EVALUATE THE PHARMACOKINETICS, SAFETY, AND TOLERABILITY OF ORALLY ADMINISTERED NIRMATRELVIR/RITONAVIR IN PREGNANT WOMEN WITH MILD-TO-MODERATE COVID-19 [NCT05386472]Phase 149 participants (Anticipated)Interventional2022-06-22Recruiting
Evaluation of the Use of Antiretroviral Regimens Containing Raltegravir for Prophylaxis of Mother-to-child-transmission of HIV Infection in Pregnant Women Presenting With Detectable Viral Load After 32 Weeks of Gestation: a Pilot Study [NCT01854762]Phase 2/Phase 340 participants (Anticipated)Interventional2015-03-31Recruiting
The Pharmacokinetics and Safety of Ritonavir-boosted Indinavir 600/100mg Bid Combined With NRTIs in ARV naïve HIV/TB Co-infected Patients Receiving Rifampicin Containing Anti-tuberculosis Therapy [NCT00411996]Phase 1/Phase 220 participants (Actual)Interventional2006-12-31Completed
A PHASE 1, OPEN-LABEL, RANDOMIZED, SINGLE DOSE, 2 SEQUENCE, 2 PERIOD CROSSOVER STUDY TO EVALUATE THE EFFECT OF HIGH-FAT MEAL ON THE RELATIVE BIOAVAILABILITY OF PF-07321332 BOOSTED WITH RITONAVIR IN HEALTHY ADULT PARTICIPANTS [NCT05129475]Phase 112 participants (Actual)Interventional2021-11-12Completed
A Single and Multiple Ascending Dose Study to Evaluate the Safety, Tolerability and Pharmacokinetics of PBI-0451 in Healthy Subjects. [NCT05011812]Phase 1130 participants (Actual)Interventional2021-08-14Completed
Phase IV Study of Boosted Atazanavir (ATV) Versus Non-boosted ATV in Naive Patients [NCT00084253]Phase 4200 participants (Anticipated)Interventional2004-06-30Completed
Kaletra and Maraviroc in Antiretroviral Therapy-Naïve Patients - KALMAR Study -Version 1.0 Amendment 2 [NCT01068873]Phase 41 participants (Actual)Interventional2010-04-30Terminated(stopped due to Poor enrollment)
A Phase 2,Multicenter,Open-Label Study to Investigate the Efficacy, Safety and Pharmacokinetics of Ritonavir-boosted Danoprevir in Combination With Peg-IFN and RBV in Treatment-Naive Non-Cirrhotic Patients Who Have Chronic Hepatitis GT1 [NCT03020004]Phase 270 participants (Actual)Interventional2016-01-31Completed
A Multi-centered, Open Label, Phase III Study on Efficacy, Safety of Ritonavir-boosted ASC08 (Danoprevir) in Combination With Peg-IFN and RBV in Treatment-Naive Non-Cirrhotic Patients Who Have Chronic Hepatitis Genotype 1 [NCT03020082]Phase 3141 participants (Actual)Interventional2016-06-30Completed
A Phase II Randomized, Multicenter Protocol Evaluating Two Antiretroviral Regimens Containing Combinations of Protease Inhibitors, NRTIs, and an NNRTI [NCT00000924]Phase 2120 participants InterventionalCompleted
An Open-label Study to Determine the Pharmacokinetics of Single-dose and/or Steady-state TPV/r 500/200 mg in Subjects With Mild and Moderate Hepatic Insufficiency [NCT02249442]Phase 124 participants (Actual)Interventional2003-10-31Completed
[NCT01483729]Phase 136 participants (Actual)Interventional2011-12-31Completed
An Open-Label, Multicenter Study to Evaluate the Efficacy and Safety of a Fixed-Dose Combination of Abacavir 600 mg/Lamivudine 300 mg Once-Daily in Combination With Atazanavir 300 mg + Ritonavir 100 mg Once-Daily in Antiretroviral-Naïve HIV-1 Infected Sub [NCT00426296]Phase 40 participants Interventional2006-08-31Recruiting
Drug-Drug Interaction Study to Assess the Effects of Steady-State Lopinavir/Ritonavir on Pitavastatin in Healthy Adult Volunteers [NCT01057433]Phase 424 participants (Actual)Interventional2010-01-31Completed
A Multicenter, Open-Label, 24-Week Pilot Study to Evaluate the Safety and Activity of Indinavir Sulfate 1200 Mg q.d. and Ritonavir 200 Mg q.d. in Combination With Stavudine and Lamivudine in Treatment Naive HIV-1 Infected Patients [NCT00002451]Phase 28 participants InterventionalActive, not recruiting
A 24-week, Randomized, Open-label, 2-arm Study to Compare the Safety, Efficacy and Tolerability of Invirase® Tablets With Ritonavir Versus Kaletra® Tablets in HIV 1 Infected Adults on a Kaletra® Based Regimen With 2 Nucleosides/Nucleotides [NCT00438152]Phase 453 participants (Actual)Interventional2006-09-30Completed
Evaluation of Potential Pharmacokinetic Interactions Between Protease Inhibitors and Lipid Lowering Agents [NCT00000941]Phase 156 participants InterventionalCompleted
A Phase I, Open-Label, 2-Period, Single-Sequence, Drug Interaction Study to Assess Steady-State Plasma Methadone Enantiomer Pharmacokinetics Following Co-Administration of Methadone QD With Fosamprenavir 700mg BID + RTV 100mg BID in Opiate-Dependent, HIV [NCT00481182]Phase 120 participants Interventional2005-02-28Completed
A Phase II, Open Label Trial in Treatment na�ve, HIV 1 Infected Subjects Who Will Receive TMC114/Rtv as a Monotherapy [NCT00513513]Phase 27 participants (Actual)Interventional2006-09-30Terminated(stopped due to Poor recruitment.)
Safety and Efficacy of Intravenous Infusion of Wharton's Jelly Derived Mesenchymal Stem Cell Plus Standard Therapy for the Treatment of Patients With Acute Respiratory Distress Syndrome Diagnosis Due to COVID 19: A Randomized Controlled Trial [NCT04390152]Phase 1/Phase 240 participants (Anticipated)Interventional2020-01-13Recruiting
A Pilot Feasibility Study of Metformin/Ritonavir Combination Treatment in Patients With Relapsed/Refractory Multiple Myeloma or Chronic Lymphocytic Leukemia [NCT02948283]Phase 13 participants (Actual)Interventional2017-09-05Completed
Pilot Study to Measure Exposure to Atazanavir, as a Component of Pharmacokinetic Parameters and Adherence Measured With MEMS in Naive HIV-infected Patients Treated Once Daily With Atazanavir Combined to Ritonavir and to Tenofovir/Emtricitabine. ANRS 134 C [NCT00528060]Phase 235 participants (Actual)Interventional2008-01-31Completed
[NCT00525239]60 participants (Anticipated)Interventional2004-03-31Recruiting
Phase IV Cohort Study Assessing Feasibility of Substituting Double Ritonavir-boosted Protease Inhibitors With Ritonavir-boosted Darunavir in HIV-infected Individuals With Viral Suppression on Highly Active Antiretroviral Therapy. [NCT00531557]Phase 412 participants (Actual)Interventional2007-09-30Completed
Characterization of Acute and Recent HIV-1 Infections in Zurich: a Long-term Observational Study [NCT00537966]2,017 participants (Anticipated)Interventional2002-01-31Recruiting
A Randomized, Open Label, Multicenter Study Comparing the Safety and Efficacy of Once Daily Regimen Containing Epzicom or Truvada Combined With Ritonavir Boosted Atazanavir as Initial Therapy for HIV-1 Infection (ET Study) [NCT00544128]Phase 4109 participants (Actual)Interventional2007-10-31Completed
Real-Life Effectiveness of the Kaletra Adherence Support Assistance (KASA) Program: A Prospective Observational Cohort Study (KASA PMOS) [NCT01662336]173 participants (Actual)Observational2012-06-30Completed
Randomized, Open-Label, Multicenter Study of Safety, Efficacy, and Tolerability of the Combination of RO5466731, RO5190591, Ritonavir, and Copegus With or Without RO5024048 in HCV Genotype 1 Infected Patients Who Are Either Treatment Naïve or Null Respond [NCT01628094]Phase 2110 participants (Actual)Interventional2012-06-30Completed
Double-blind, Randomized, Placebo-controlled, Multi-center Trial to Determine the Safety and Antiviral Effect of 3 Days of BZF961 With or Without Ritonavir Boosting in Hepatitis C Virus (HCV) Infected Patients [NCT01825980]Phase 1/Phase 254 participants (Actual)Interventional2013-03-31Completed
Monotherapy in Africa: Evaluation of New Therapy [NCT02155101]Phase 3120 participants (Actual)Interventional2014-05-31Completed
Real World Evidence of the Effectiveness of Paritaprevir/r - Ombitasvir, ± Dasabuvir, ± Ribavirin in Participants With Chronic Hepatitis C - An Observational Study in Greece [NCT02725866]216 participants (Actual)Observational2016-04-05Completed
A One-Directional, Open-label Drug Interaction Study to Investigate the Effects of Multiple-Dose Ritonavir on Single-Dose Pharmacokinetics of Colchicine in Healthy Volunteers [NCT00983515]Phase 124 participants (Actual)Interventional2008-07-31Completed
A Phase 3 Multicenter, Double-Blind, Randomized, Active Comparator-Controlled Clinical Trial to Evaluate the Safety and Efficacy of Doravirine (MK-1439) 100 mg Once Daily Versus Darunavir 800 mg Once Daily Plus Ritonavir 100 mg Once Daily, Each in Combina [NCT02275780]Phase 3769 participants (Actual)Interventional2014-12-01Completed
Evaluating the Effects of Tipranavir (With Ritonavir) Capsule and Liquid Formulation on Cytochrome P450 and P-glycoprotein Activity Using a Biomarker Cocktail in Healthy Human Volunteers [NCT02243553]Phase 134 participants (Actual)Interventional2006-01-31Completed
A Pilot Study of Once-Daily Therapy With Amprenavir, Ritonavir, Lamivudine and Abacavir in HIV-Infected, Antiretroviral-Naive Patients [NCT00001968]Phase 125 participants Interventional2000-01-31Completed
A Phase IIIB Open-Label, Comparative Study to Evaluate Saquinavir Soft Gel Capsule (SGC) Treatment in Combination With Other Antiretrovirals in HIV-1 Infected Antiretroviral-Naive Patients [NCT00002178]Phase 3375 participants InterventionalCompleted
Evaluation of Immune Reconstitution in HIV Infected Patients Treated With Fortovase (Saquinavir) SGC QD Plus Ritonavir QD Plus 2 NRTIs Vs Efavirenz QD Plus 2 NRTIs [NCT00002448]Phase 340 participants Interventional1999-10-31Completed
Phase I/II Study of ABT-378/Ritonavir in Combination With Reverse Transcriptase Inhibitors in Antiretroviral Naive HIV-Infected Patients [NCT00004578]Phase 1/Phase 2100 participants (Actual)Interventional1997-11-30Completed
A Multicenter, Open-Label, 24-Week Study to Evaluate the Efficacy and Safety of Indinavir Sulfate 800 Mg and Ritonavir 200 Mg b.i.d. Plus 2 NRTIs b.i.d. in HIV-1 Infected Individuals Who Require Early Treatment Intervention [NCT00005118]Phase 2100 participants InterventionalActive, not recruiting
A Phase II, Open-label Trial, to Investigate Pharmacokinetics, Safety, Tolerability and Antiviral Activity of TMC114/Rtv b.i.d in Treatment-Experienced HIV-1 Infected Children and Adolescents [NCT00355524]Phase 280 participants (Actual)Interventional2006-06-30Completed
Randomized, Open-Label, Multiple-Dose Study to Evaluate the Effect of Omeprazole 20 mg on the Pharmacokinetics of Atazanavir Administered With Ritonavir in Healthy Subjects [NCT00357240]Phase 156 participants Interventional2006-06-30Completed
Tipranavir (PNU-140690): A Fourteen Day Dose-response Study Using a Prototype Self-emulsifying Drug Delivery System (SEDDS) Formulation in Treatment-naive HIV-1 Infected Patients. Report on the Post-study Option, a 46-week Treatment Period of Triple Thera [NCT02249130]Phase 218 participants (Actual)Interventional1999-03-31Completed
Comparison Atazanavir/Ritonavir (ATV/r) vs Nevirapine (NVP) Twice a Day (Bid) on Truvada Backbone [NCT00552240]Phase 4154 participants (Actual)Interventional2007-09-30Completed
A Phase III, Open Label, Randomized, Comparative Study of the Antiviral Efficacy of ARV Therapy With Lopinavir/Ritonavir (LPV/r-Kaletra) in Combination With Tenofovir (TDF) Versus Standard of Care (Kaletra in Combination With 2 Nucleoside RTIs) in naïve-H [NCT00234910]Phase 3152 participants (Actual)Interventional2005-01-31Completed
A Phase III Multicenter, Open-Label, Randomized Study to Evaluate a Switch to MK-1439A in HIV-1-Infected Subjects Virologically Suppressed on a Regimen of a Ritonavir-boosted Protease Inhibitor and Two Nucleoside Reverse Transcriptase Inhibitors (NRTIs) [NCT02397096]Phase 3673 participants (Actual)Interventional2015-06-09Completed
An Open-Label, Randomized Study to Determine the Impact of Antiretroviral Treatment in HCV/HIV-Coinfected Subjects With High CD4+ Cell Count on the Efficacy of Hepatitis C Treatment With Pegylated Interferon Alfa-2A and Ribavirin [NCT00100581]2 participants (Actual)InterventionalCompleted
A Phase I Trial of the Safety and Pharmacokinetics of Fortovase (Saquinavir-SGC) Co-Administered With Low Dose (Ritonavir) RTV, ZDV and 3TC in HIV Seropositive Pregnant Women During Gestation and Postpartum, and in Their Infant's Post-Maternal Dosing [NCT00000920]Phase 124 participants InterventionalCompleted
Efficacy and Tolerability of an Antiretroviral bi-Therapy in HIV-1 Infected Patients With Multidrug Resistant HIV ANRS 109 Vista Trial. [NCT00120783]Phase 240 participants Interventional2002-02-28Terminated
A Randomized, Open-Label Study Assessing Safety, Tolerability and Efficacy of an Induction-Maintenance Treatment Strategy Including Lopinavir/Ritonavir Plus Tenofovir Disoproxil Fumarate and Emtricitabine Versus Efavirenz Plus Tenofovir Disoproxil Fumarat [NCT00121017]Phase 2200 participants InterventionalWithdrawn
MEDICLAS Study (Metabolic Effects of Different Classes of AntiretroviralS) [NCT00122226]Phase 450 participants Interventional2003-01-31Active, not recruiting
A Retrospective Observational Non-Interventional Study (NIS) to Assess Patient Characteristics Among COVID-19 Patients Receiving Treatment With Nirmatrelvir/Ritonavir (PAXLOVIDTM) in Morocco. [NCT05997485]150 participants (Anticipated)Observational2023-10-01Not yet recruiting
Open-Label, Single-Sequence Study to Evaluate the Effects of Darunavir/Ritonavir and/or Etravirine on the Pharmacokinetics of GSK3640254 and the Effects of GSK3640254 on the Pharmacokinetics of Darunavir/Ritonavir and/or Etravirine in Heathy Adults [NCT04630002]Phase 154 participants (Actual)Interventional2020-10-28Completed
Very Early Intensive Treatment of HIV-Infected Infants to Achieve HIV Remission: A Phase I/II Proof of Concept Study [NCT02140255]Phase 1/Phase 2905 participants (Anticipated)Interventional2015-01-23Recruiting
Randomized Open Label Study Assessing the Antiviral Activity, Toxicity and Pharmacologic Interaction of Tenofovir DF/Atazanavir Enhanced With Low Dose of Ritonavir as Part of a Salvage Regimen in HIV Infected Patients With Multiple Treatment Failures (ANR [NCT00122577]Phase 250 participants Interventional2002-03-31Terminated
Special Investigation of Kaletra in Pregnant Women [NCT01076985]24 participants (Actual)Observational2000-12-31Completed
See Detailed Description [NCT00085943]Phase 3866 participants Interventional2004-05-31Completed
The Pharmacokinetics of Lopinavir/Ritonavir in Combination With Atazanavir in HIV-Infected Subjects [NCT00420355]Phase 419 participants (Actual)Interventional2007-04-30Terminated(stopped due to Unexpected adverse event)
DRIVESHAFT: Phase IV Randomized, Open-Label Study in HIV-1 Virologically-suppressed Patients On Regimens With Darunavir 600mg/Ritonavir 100mg Twice-daily Switching to Darunavir 800mg/Ritonavir 100mg Once-daily Vs. Continuing Twice-daily Darunavir/Ritonavi [NCT01423812]Phase 460 participants (Actual)Interventional2012-01-31Completed
An Open Label, Non-randomized Treatment Protocol of Tipranavir Co-administered With Low-dose Ritonavir (TPV/r) in Protease Inhibitor-experienced Patients With HIV-1 Infection (the Tipranavir Expanded Access Program) [NCT00097799]0 participants Expanded Access2004-12-31Approved for marketing
A Randomized Clinical Trial Assessing Continuous HAART Versus Interrupted HAART in a Resource Poor Clinic [NCT00100646]30 participants (Actual)Interventional2007-03-31Completed
A Phase II, Randomized, Open-Label Study to Evaluate the Safety and Effectiveness of Two Antiretroviral Therapeutic Strategies: A Dual PI-Based HAART Regimen Versus a Multi-NRTI ART Regimen, in ART-Experienced Children and Youth Who Have Experienced Virol [NCT00102206]Phase 26 participants (Actual)InterventionalCompleted
Sex Differences in Lopinavir/Ritonavir Pharmacokinetics Among HIV-1-Infected Men and Women [NCT00102986]116 participants (Actual)Interventional2005-10-31Completed
Optimizing Pediatric HIV-1 Treatment in Infants With Prophylactic Exposure to Nevirapine, Nairobi, Kenya (6-12 Month RCT) [NCT00427297]Phase 334 participants (Actual)Interventional2007-09-30Terminated(stopped due to There is no longer equipoise. DSMB recommended termination.)
Phase III-IV, Comparative, Randomized, Open-Label, Study to Evaluate Safety and Efficacy of Suspending Nucleosides From a Triple-Drug Therapy Based on Lopinavir/Ritonavir Versus Continuing Triple-Drug Therapy in HIV-Infected Subjects With Undetectable Pla [NCT00114933]Phase 4200 participants Interventional2005-01-31Completed
A Randomized Comparison of Protease Inhibitor-based Versus Non-nucleoside Reverse Transcriptase Inhibitor-based Antiretroviral Therapy for Initial Treatment of Individuals With AIDS-related Kaposi's Sarcoma in Sub-Saharan Africa [NCT00444379]Phase 4224 participants (Actual)Interventional2007-04-30Completed
Pharmacokinetic and Efficacy of Saquinavir Mesylate Film Coated Tablet / Ritonavir 1500/100 Plus Tenofovir/Emtricitabine 300/200 mg Once Daily in HIV Pretreated Patients [NCT00476983]Phase 2/Phase 30 participants (Actual)InterventionalWithdrawn(stopped due to No funding)
A 48-Weeks National Multicenter Randomized Open Clinical Trial Evaluating Tolerance and Efficacy of a Treatment Simplification by Lopinavir/Ritonavir Versus Continuation of Current Treatment in HIV-Infected Patients With a Viral Load Inferior to 50 Copies [NCT00140751]Phase 3186 participants (Actual)Interventional2005-10-31Completed
An Open Label Safety Study of Tipranavir Co-Administered With Low-Dose Ritonavir (TVP/r) in Patients With Advanced HIV-1 Infection and Limited Treatment Options [NCT00144287]Phase 3255 participants (Actual)Interventional2004-05-31Completed
Pharmacokinetics of an Indinavir, Ritonavir and Amprenavir Regimen in HIV-Infected Individuals: A Pilot Study [NCT00148785]Phase 415 participants (Anticipated)Interventional2005-07-31Completed
Randomized, Single Oral Dose,Two Treatment,Four-period,Full-replicated,Cross-over Trial to Assess the BE of Orvical 200 mg/50 mg FT in Comparison With Kaletra 200 mg/50 mg FT in Healthy Male Subjects Under Fasting Conditions [NCT04386876]Phase 130 participants (Actual)Interventional2020-04-30Completed
The Regimen of Favipiravir Plus Hydroxychloroquine Can Accelerate Recovery of the COVID-19 Patients With Moderate Severity in Comparison to Lopinavir/Ritonavir Plus Hydroxychloroquine Regimen: an Open-label, Non-randomized Clinical Trial Study [NCT04376814]40 participants (Actual)Interventional2020-03-29Completed
A Randomized Trial of Efficacy and Safety of an Early OUTpatient Treatment of COVID-19 in Patients With Risk Factor for Poor Outcome: a Strategy to Prevent Hospitalization [NCT04365582]Phase 30 participants (Actual)Interventional2020-05-07Withdrawn(stopped due to The PI decided.)
Efficacy of Pragmatic Same-day Ring COVID-19 Prophylaxis for Adult Individuals Exposed to SARS-CoV-2 in Switzerland: an Open-label Cluster Randomized Trial [NCT04364022]Phase 3326 participants (Actual)Interventional2020-04-23Completed
Efficacy and Safety of Umifenovir as an Adjuvant Therapy Compared to the Control Therapeutic Regiment of Interferon Beta 1a, Lopinavir / Ritonavir and a Single Dose of Hydroxychloroquine in Moderate to Severe COVID-19: A Randomized, Double-Blind, Placebo- [NCT04350684]Phase 440 participants (Anticipated)Interventional2020-04-15Enrolling by invitation
A Multi-Center Comparison of Raltegravir to Lopinavir/Ritonavir, Both in Combination With Truvada, in HIV-Infected Individuals Naive to Antiretroviral Therapy [NCT00632970]Phase 46 participants (Actual)Interventional2008-02-29Terminated(stopped due to no patients completed)
A Proof of Concept Study for GSK2248761 (An Extension of NV-05A-002: A Phase I/IIa Double-Blind Study to Evaluate the Safety and Tolerability, Antiretroviral Activity, Pharmacokinetics and Pharmacodynamics of IDX12899 in Antiretroviral Treatment-Naive HIV [NCT00945282]Phase 28 participants (Actual)Interventional2009-10-20Completed
Bone and Body Comp: A Sub Study of the SECOND-LINE Study [NCT01513122]Phase 4210 participants (Actual)Interventional2010-02-28Completed
HIV Infection and Breastfeeding: Interventions for Maternal and Infant Health [NCT00164736]Phase 32,369 participants (Actual)Interventional2004-03-31Completed
"Study ACA-ARGE-04-001 A Pilot, Open-Label Study Assessing Safety, Tolerability, Efficacy of a Simplified Lopinavir/Ritonavir Induction/Maintenance Therapy in HIV-Infected Subjects on Their First Protease Inhibitor-Based Regimen." [NCT00159224]Phase 4100 participants (Actual)Interventional2005-04-30Completed
Switch to Unboosted Atazanavir With Tenofovir (SUAT) Study [NCT01351740]Phase 450 participants (Actual)Interventional2011-07-31Completed
A Phase II, Randomized, Open-Label Comparative Trial of Salvage Antiretroviral Therapies for HIV-Infected Individuals With Virological Evidence of Nelfinavir Treatment Failure as Reflected by Plasma HIV RNA Concentration of >= 1,000 Copies/ml [NCT00000918]Phase 2300 participants InterventionalCompleted
The Efficacy and Safety of Carrimycin Treatment in Patients With Novel Coronavirus Infectious Disease (COVID-19) : A Multicenter, Randomized, Open-controlled Study [NCT04286503]Phase 4520 participants (Anticipated)Interventional2020-02-23Not yet recruiting
A Randomized, Open-label, Controlled Study of the Efficacy of Lopinavir Plus Ritonavir and Arbidol for Treating With Patients With Novel Coronavirus Infection [NCT04252885]Phase 486 participants (Actual)Interventional2020-01-28Completed
Effects of Traditional Chinese Medicines (TCMs) on Patients With COVID-19 Infection: A Perspective, Open-labeled, Randomized, Controlled Trial [NCT04251871]150 participants (Anticipated)Interventional2020-01-22Recruiting
MERS-CoV Infection tReated With A Combination of Lopinavir /Ritonavir and Interferon Beta-1b: a Multicenter, Placebo-controlled, Double-blind Randomized Trial [NCT02845843]Phase 2/Phase 395 participants (Actual)Interventional2016-07-31Completed
Pharmacokinetic Evaluation of Single-dose Rosuvastatin 10 mg When Co-administered With Steady-state Tipranavir 500 mg/Ritonavir 200 mg TPV/r) B.I.D. in Healthy Adult Volunteers [NCT00344123]Phase 129 participants (Actual)Interventional2007-02-28Completed
Prospective Clinical Trial to Assess Safety and Efficacy of DRV/r(TMC 114/r), ETV(TMC 125) and MK-0518 in Addition to OBT in HIV-1 Infected Patients With Limited to No Treatment Options ANRS 139 TRIO [NCT00460382]Phase 2103 participants (Actual)Interventional2007-05-31Completed
An Open Label Study of the Impact on Insulin Sensitivity, Lipid Profile and Vascular Inflammation by Treatment With Lopinavir / Ritonavir (400 / 100 mg Twice Daily) or Raltegravir 400 mg Twice Daily in HIV Negative Male Volunteers. [NCT00531999]Phase 118 participants (Actual)Interventional2007-10-31Completed
A Drug-Drug Interaction Study Between Danoprevir/Low-dose Ritonavir and Cyclosporine, a Potent Inhibitor of OATP, in Healthy Subjects [NCT01514968]Phase 118 participants (Actual)Interventional2011-12-31Completed
A Study to Evaluate the Potential Drug-Drug Interaction Between Darunavir and Danoprevir When Administered Together With Low-Dose Ritonavir in Healthy Volunteers [NCT01519336]Phase 140 participants (Actual)Interventional2012-02-29Completed
Real World Evidence of the Effectiveness of Paritaprevir/r - Ombitasvir, ± Dasabuvir, ± Ribavirin in Patients With Chronic Hepatitis C -An Observational Study in Ireland [NCT02582671]101 participants (Actual)Observational2015-11-05Completed
A Phase IIa, Open-label, Randomized Trial in Treatment-naive HIV-1-infected Subjects to Determine the Antiviral Activity of 14 Days of Monotherapy With 4 Different Dose Regimens of TMC310911 Coadministered With Ritonavir [NCT00838162]Phase 233 participants (Actual)Interventional2009-06-30Completed
A Phase II Safety and Activity Study of Ritonavir in the Treatment of HIV-Associated Cutaneous Kaposi's Sarcoma [NCT00002366]Phase 20 participants InterventionalCompleted
Ex Vivo Study of Immune-Reconstitution Kinetics in HIV-infected ARV-naive Subjects, With Advanced Disease, Starting a Darunavir/Ritonavir or Efavirenz Based HAART (IMMUNO Study) [NCT01541085]33 participants (Actual)Observational2011-12-31Completed
Open-label, Comparative and Randomised Pilot Study to Evaluate the Efficacy and Safety of Saquinavir/Ritonavir in Single Therapy vs Standard HAART Therapy as Maintenance Therapy. [NCT00379405]Phase 430 participants (Actual)Interventional2006-06-30Completed
A Randomized Trial of Two Saquinavir-Containing Combination Treatment Regimens in Children With HIV Infection [NCT00000913]Phase 250 participants InterventionalCompleted
Pharmacokinetics of and Rate of HIV-1 RNA Decline in ARV-naive HIV-1 Infected Patients Treated With Low- or Standard-dose Saquinavir HGC (Invirase®) and Lopinavir/Ritonavir (Kaletra® [NCT00400738]Phase 248 participants (Actual)Interventional2004-03-31Completed
Evaluation of the Relationship Between Immunologic Recovery After Highly Active Antiretroviral Therapy and the Ability to Mobilize CD34+ Stem Cells Following G-CSF Administration [NCT00006578]0 participants (Actual)InterventionalWithdrawn
A Phase I/II, Randomized, Open-Label Study of the Safety and Pharmacokinetics of Indinavir + Ritonavir Therapy in HIV-Infected Subjects Failing Amprenavir, Nelfinavir, Saquinavir, or Nelfinavir/Saquinavir Combination Therapy [NCT00001133]Phase 150 participants InterventionalCompleted
Effect of Change to a Nucleoside Reverse Transcriptase Inhibitor (NRTI)-Sparing Regimen of Efavirenz (EFV) and Lopinavir/Ritonavir (LPV/r) on Liver Histology in HIV-1-Infected Individuals With Lactic Acidemia and Persistent Alanine Aminotransferase (ALT) [NCT00023218]0 participants (Actual)InterventionalWithdrawn
A Randomized, Comparative Study of Lopinavir/Ritonavir Versus GW433908 and Ritonavir Versus Lopinavir/Ritonavir and GW433908 (In Combination With Tenofovir Disoproxil Fumarate and One or Two Nucleoside Reverse Transcriptase Inhibitors) in HIV-1-Infected S [NCT00028366]56 participants InterventionalCompleted
A Randomized, Open-Label Study of 800 Mg Lopinavir/200 Mg Ritonavir QD in Combination With Tenofovir and Emtricitabine Vs. 400 Mg Lopinavir /100 Mg Ritonavir BID in Combination With Tenofovir and Emtricitabine in HIV-Infected Antiretroviral Naïve Subjects [NCT00043966]Phase 3200 participants Interventional2002-07-31Completed
A Pilot Study of the Immunologic Reconstitution in HIV-1 Infected Children Receiving Highly Active Antiretroviral Therapy With Combination Ritonavir, Nevirapine and Stavudine [NCT00001688]Phase 225 participants Interventional1998-01-31Completed
A Randomised, Controlled, Open-Label, 48-Week, Study To Asses Differences in Changes In Plasma Lipid Profile Between Patients On Saquinavir/Ritonavir Or Atazanavir/Ritonavir In Combination With Tenofovir Disoproxil Fumarate And Emtricitabine As A First-li [NCT00389402]Phase 4120 participants (Anticipated)Interventional2006-07-31Completed
A Phase II, Randomized, Open-Label Study Comparing Fixed-Dose Versus Concentration-Adjusted Lopinavir/Ritonavir Therapy in HIV-Infected Subjects on Salvage Therapy [NCT00046033]Phase 2118 participants InterventionalCompleted
Pharmacokinetic Interactions Between Ritonavir, Amprenavir and Efavirenz and Nelfinavir, Amprenavir, and Efavirenz in People Infected With HIV [NCT00001766]Phase 122 participants Interventional1998-08-31Completed
A Phase IV Study of Antiretroviral Therapy for HIV Infected Adults Presenting With Acute Opportunistic Infections: Immediate Versus Deferred Initiation of Antiretroviral Therapy [NCT00055120]Phase 4283 participants (Actual)Interventional2003-03-31Completed
A Comparative Trial of Protease-Containing and Protease-Sparing HAART Regimens in HIV-Infected Adolescents With an Evaluation of Therapeutic Drug Monitoring [NCT00075907]Phase 3240 participants Interventional2004-07-31Completed
A Pilot Study of a Nucleoside Analogue Reverse Transcriptase Inhibitor Sparing Regimen in Antiretroviral-Naïve, HIV-infected Patients [NCT00143689]Phase 413 participants (Actual)Interventional2002-04-30Completed
Effect of Moderate Liver Impairment on the Pharmacokinetics of Saquinavir After Administration of Saquinavir/Ritonavir 1000/100mg BID in HIV Patients [NCT00435929]Phase 116 participants (Actual)Interventional2006-09-30Completed
An Open-Label, Non-Randomized Study of Pharmacokinetic Interactions Between Depo-Medroxyprogesterone Acetate (DMPA, Depo-Provera) and Selected Protease Inhibitor (PI) and Nonnucleoside Reverse Transcriptase Inhibitor (NNRTI) Therapies Among HIV-Infected W [NCT00016601]76 participants Interventional2001-06-30Completed
Phase 2 Study To Investigate the Efficacy, Safety And Pharmacokinetics Of Ravidasvir In Combination With Ritonavir-boosted Danoprevir And Ribavirin In Treatment-naive Non-cirrhotic Taiwanese Patients Who Have Chronic Hepatitis C Genotype 1 [NCT03020095]Phase 238 participants (Actual)Interventional2015-08-31Completed
A Phase II Study of the Predictive Value of Pharmacokinetic-Adjusted Phenotypic Susceptibility (C12h/IC50) on Antiretroviral Response to Ritonavir-Enhanced Protease Inhibitors in Subjects With Failure of Previous Protease Inhibitor-Based Regimens [NCT00027339]Phase 253 participants (Actual)InterventionalCompleted
A Restrictively Randomized, Open-Label, Controlled, Pilot Study of the Effect of a Thymidine Analogue Substitution or Change to a Nucleoside-Sparing Regimen on Peripheral Fat Wasting [NCT00028314]150 participants Interventional2002-03-31Completed
Immunologic Consequences of Antiretroviral Therapy Intensification in Subjects With Moderately Advanced HIV-1 Disease: A Follow-Up Study to ACTG 315/375 [NCT00034086]22 participants InterventionalCompleted
A Phase I/II, Open Label Study to Evaluate the Ability of Combination Therapy With ABT-378/Ritonavir (Kaletra), Lamivudine (Epivir), Efavirenz (Sustiva)and Tenofovir DF to Completely Suppress Viral Replication in Subjects Infected With HIV-1 [NCT00038220]Phase 240 participants Interventional2000-07-31Completed
An Open Label, Phase II Study of Amprenavir/Ritonavir, Saquinavir/Ritonavir or Efavirenz in HIV-Infected Subjects Following Failure With Kaletra (ABT-378/Ritonavir) as Their First Protease Inhibitor Based HAART [NCT00038532]Phase 224 participants Interventional2001-04-30Completed
Evaluation of Potential Pharmacokinetic Interactions Between HIV Protease Inhibitors and Calcium Channel Blockers [NCT00039975]Phase 132 participants InterventionalCompleted
A Phase III, Randomized, Open-Label Comparison of Lopinavir/Ritonavir Plus Efavirenz Versus Lopinavir/Ritonavir Plus 2 NRTIs Versus Efavirenz Plus 2 NRTIs as Initial Therapy for HIV-1 Infection [NCT00050895]Phase 3775 participants InterventionalCompleted
A Pilot Study to Measure the Clearance of Replication-Competent HIV-1 in Resting Memory CD4+ Cells in HIV-1-Infected Subjects Who Receive Enfuvirtide Plus Oral Combination Antiretroviral Therapy [NCT00051831]19 participants (Actual)Interventional2003-10-31Completed
A Phase III, Randomized, Multicenter, Parallel Group, Open-Label, Three Arm Study to Compare the Efficacy and Safety of Two Dosing Regimens of GW433908/Ritonavir (700mg/100mg Twice Daily or 1400mg/200mg Once Daily) Versus Lopinavir/Ritonavir (400mg/100mg [NCT00025727]Phase 3330 participants Interventional2001-05-31Active, not recruiting
A Phase 2b, Open-Label, Randomized Study of the Safety, Tolerability, and Pharmacodynamic Activity of Lonafarnib With or Without Ritonavir in Patients Chronically Infected With Hepatitis Delta Virus (LOWR-5) [NCT02968641]Phase 20 participants (Actual)InterventionalWithdrawn(stopped due to This study was initially planned to enroll patients in Mongolia. However, due to challenges in setting it up in Mongolia, this study was later determined not to be initiated.)
An Investigation Into Beneficial Effects of Interferon Beta 1a, Compared to Interferon Beta 1b And The Base Therapeutic Regiment in Moderate to Severe COVID-19: A Randomized Clinical Trial [NCT04343768]Phase 260 participants (Actual)Interventional2020-04-09Completed
A Phase 1, Dose-Rising Study of AMD11070 in HIV-Seronegative Men to Assess the Safety and Pharmacokinetics After Single or Multiple Doses [NCT00063804]Phase 144 participants (Actual)InterventionalCompleted
An Open Label, Phase II Study of Amprenavir/Ritonavir or Saquinavir/Ritonavir in HIV-Infected Subjects Following Failure With Kaletra (Lopinavir/Ritonavir) as Their Second Protease Inhibitor. [NCT00038519]Phase 2/Phase 316 participants Interventional2001-04-30Completed
A Study of Two Different Doses of ABT-378/Ritonavir in HIV-Infected Patients Who Have Taken Protease Inhibitors and Non-Nucleoside Reverse Transcriptase Inhibitors [NCT00038636]Phase 336 participants Interventional2000-09-30Completed
A Phase II Pharmacokinetic Study of the Transdermal Contraceptive System and Oral Contraceptive in HIV-1 Infected Women on Lopinavir/Ritonavir [NCT00125983]Phase 232 participants (Actual)InterventionalCompleted
A Randomized, Open-Label Study Exploring a Simplified Kaletra® (Lopinavir/Ritonavir)-Based Therapy Versus a Sustiva® (Efavirenz)-Based Standard of Care in Previously Non-Treated HIV-Infected Subjects [NCT00075231]Phase 2150 participants Interventional2003-12-31Completed
The Pharmacokinetics of SAquinavir (Invirase New Tablet Formulation) 1,000mg + Ritonavir (Norvir) 100mg q12h in HIV-infected Pregnant Women (SARA) [NCT00145561]Phase 1/Phase 240 participants (Actual)Interventional2005-08-31Completed
Clinical Pilot Trial to Evaluate the Influence of Nevirapine in Exposure to Atazanavir in Steady State Equilibrium in HIV-infected Adult Patients. [NCT00355719]Phase 414 participants (Actual)Interventional2007-01-31Completed
A Phase II Study of Lopinavir/Ritonavir in Combination With Saquinavir Mesylate or Lamivudine/Zidovudine to Explore Metabolic Toxicities in Antiretroviral HIV-Infected Subjects [NCT00043953]Phase 230 participants (Actual)Interventional2002-08-31Completed
Boosted PI VS. NNRTI Based Therapy as Initial Treatment for HIV-1 Infected Patients With Advanced Disease [NCT00162643]Phase 4300 participants Interventional2004-12-31Recruiting
Maternal and Infant Peripartum Nevirapine, Versus Infant Only Peripartum Nevirapine, or Maternal Lopinavir/Ritonavir in Addition to Standard Zidovudine Prophylaxis for the Prevention of Perinatal HIV in Thailand. [NCT00409591]Phase 3435 participants (Actual)Interventional2008-07-31Terminated(stopped due to Change in National PMTCT guidelines in Thailand)
Latency and Early Neonatal Provision of Antiretroviral Drugs Clinical Trial [NCT02431975]Phase 473 participants (Actual)Interventional2015-08-31Completed
Randomized, Open-Label 2x2 Factorial Study to Compare the Safety and Efficacy of Different Combination Antiretroviral Therapy Regimens in Treatment Naive Patients With Advanced HIV Disease and/or CD4+ Cell Counts Less Than 200 Cells/MicroL [NCT00342355]Phase 41,771 participants (Actual)Interventional2004-01-31Completed
Relative Bioavailability of a Single Oral Dose of 40 mg Afatinib Given Alone Compared to Concomitant and Timed Administration of Multiple Oral Doses of Ritonavir - an Open-label, Randomised, Three-way Crossover Trial in Healthy Male Volunteers [NCT01426958]Phase 124 participants (Actual)Interventional2011-08-31Completed
Mechanisms of Immune Reconstitution & Reduced Immune Activation Following Darunavir-based ART [NCT01869634]Phase 437 participants (Actual)Interventional2013-06-30Completed
The Safety of Paxlovid (Nirmatrelvir/Ritonavir) in Hemodialysis Patients With SARS-CoV-2 Infection [NCT05366192]Phase 418 participants (Actual)Interventional2022-05-06Completed
An Open-Label, Fixed-Sequence Study in Healthy Subjects to Assess the Effect of Ritonavir on the Single-Dose Pharmacokinetics of JNJ-61393215 [NCT03593954]Phase 112 participants (Actual)Interventional2018-07-31Completed
A Phase II, Randomized Trial of Open-Label Truvada With Darunavir/Ritonavir Versus Multiclass Therapy With Truvada, Darunavir/Ritonavir, Maraviroc and Raltegravir in Acutely HIV-1 Infected Antiretroviral-Naïve Subjects [NCT00525733]40 participants (Actual)Interventional2007-10-31Completed
An Open-Label Study to Evaluate the Safety and Efficacy of Ombitasvir/Paritaprevir/Ritonavir With or Without Dasabuvir in Adults With Genotype 1a or Genotype 4 Chronic Hepatitis C Virus (HCV) Infection, With Severe Renal Impairment or End-Stage Renal Dise [NCT02487199]Phase 318 participants (Actual)Interventional2015-09-30Completed
An Open-Label, Multicenter Study to Evaluate the Safety and Efficacy of Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir With or Without Ribavirin (RBV) in US Veterans With Genotype 1 Chronic Hepatitis C Virus (HCV) Infection (TOPAZ-VA) [NCT02442284]Phase 399 participants (Actual)Interventional2015-05-13Completed
Pharmacokinetic Interactions Between Buprenorphine and Kaletra (Lopinavir/Ritonavir) [NCT00571961]12 participants (Actual)Interventional2007-01-31Completed
Evaluation of Efficacy and Safety of Ombitasvir, Paritaprevir, and Ritonavir Plus Ribavirin Based Therapy for Chronic Hepatitis C With or Without Compensated Cirrhosis in Haemodialysis Patients [NCT03067883]Phase 240 participants (Anticipated)Interventional2016-11-09Recruiting
An Open-label Randomized Controlled Trial on Lopinavir/ Ritonavir, Ribavirin and Interferon Beta 1b Combination Versus Lopinavir/ Ritonavir Alone, as Treatment for 2019 Novel Coronavirus Infection [NCT04276688]Phase 2127 participants (Actual)Interventional2020-02-10Completed
A Randomized Multicenter Study With Non-inferiority Hypothesis, Comparing the Availability to Maintain a Complete Viral Suppression by a Monotherapy of Darunavir/r to a NRTI Containing Regimen Including Darunavir/r, in HIV-1 Infected Patients With Previou [NCT00421551]Phase 3225 participants (Actual)Interventional2007-03-31Completed
CID 0821 - Pilot Study to Evaluate HIV Viremia and Persistence in Acutely HIV-Infected Antiretroviral Naïve Patients Treated With Darunavir/Ritonavir and Etravirine [NCT00855413]Phase 415 participants (Actual)Interventional2009-03-31Terminated(stopped due to Study halted by sponsor due to slow enrollment.)
An Open-label, Randomized Bioequivalence Study (Preliminary Study) of Ritonavir Versus NORVIR in Healthy Chinese Subjects [NCT03214159]Phase 112 participants (Actual)Interventional2017-07-11Completed
Selective Trial Of Paxlovid for PASC (STOP-PASC): Randomized Double-Blind Placebo-Controlled Pilot Trial of Paxlovid for the Treatment of PASC [NCT05576662]Phase 2168 participants (Actual)Interventional2022-11-08Completed
Pharmacokinetic Properties of Antiretroviral and Anti-Tuberculosis Drugs During Pregnancy and Postpartum [NCT04518228]325 participants (Anticipated)Observational2021-06-08Recruiting
Food Effects Study of GST-HG171 Tablets Combined With Ritonavir in Healthy Chinese Participants [NCT06084507]Phase 118 participants (Anticipated)Interventional2023-05-04Active, not recruiting
Pharmacokinetic Interactions Between Antiretroviral Agents, Lopinavir/Ritonavir and Efavirenz and Antimalarial Drug Combination, Artemether/Lumefantrine [NCT00266058]Phase 133 participants (Actual)Interventional2005-12-31Completed
A Pilot, Open-Label, Randomized, Comparative Study of the Antiviral Efficacy of Lopinavir/Ritonavir Single-Drug Regimen Versus Lopinavir/Ritonavir in Combination With Lamivudine/Zidovudine in Antiretroviral Naïve Patients [NCT00234923]Phase 3138 participants (Actual)Interventional2003-08-31Completed
Factors Associated With Adherence in a Cohort of HIV Positive Subjects on a First Time PI Containing HAART Regimen: Observational Study of the Impact of Adherence on Viral Load for a HAART Regimen Containing Kaletra vs Other Selected PI Containing HAART. [NCT00234962]Phase 4200 participants Interventional2002-08-31Terminated
An Open-Label Study to Evaluate the Safety and Efficacy of Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir With Ribavirin in Adults With Genotype 1 and Ombitasvir/Paritaprevir/Ritonavir With Ribavirin in Adults With Genotype 4 Chronic Hepatitis C Virus In [NCT02219477]Phase 336 participants (Actual)Interventional2014-11-24Completed
Phase IIB, Randomized, Multicenter, Parallel Group, Study to Evaluate the Safety, Pharmacokinetics and Antiviral Effect of Four Blinded Dosing Regimens of GW640385X/Ritonavir Compared to Open-label Current PI Therapy in HIV-1 Infected, Protease Inhibitor [NCT00242879]Phase 2130 participants (Actual)Interventional2005-08-31Terminated
A Pilot, Phase II, Open-label, Single Arm Study to Evaluate the Efficacy, Safety, Tolerability, and Pharmacokinetics of GW640385 When Administered With Ritonavir in Combination With NRTIs for 48 Weeks in HIV-1 Infected Adults [NCT00257621]Phase 230 participants (Actual)Interventional2004-10-31Completed
A Phase IIIb, Randomized, Open-label Study of the Safety and Efficacy of Dolutegravir/Abacavir/Lamivudine Once Daily Compared to Atazanavir and Ritonavir Plus Tenofovir/Emtricitabine Once Daily in HIV-1 Infected Antiretroviral Therapy Naïve Women [NCT01910402]Phase 3499 participants (Actual)Interventional2013-08-22Completed
Bioequivalence of Two Different Oral Solutions of 500 mg of Tipranavir (New Formulation vs. Current Formulation) Administered in Combination With 200 mg of Ritonavir (Oral Solution) to Healthy Volunteers (an Open-label, Randomised, Single Dose, Two-way Cr [NCT02244190]Phase 132 participants (Actual)Interventional2008-04-30Completed
A Phase II, Open-Label Trial, to Evaluate Pharmacokinetics, Safety, Tolerability and Antiviral Activity of Drv/Rtv Once Daily in Treatment-Naive HIV-1 Infected Adolescents Aged Between 12 and < 18 Years [NCT00915655]Phase 212 participants (Actual)Interventional2009-07-31Completed
Cross-sectional Study for the Characterisation of the Pharmacokinetic Parameters of Protease Inhibitors and Non-nucleoside Analog Reverse Transcriptase Inhibitors in the Spanish Population of HIV-infected Subjects [NCT00307502]Phase 1675 participants (Actual)Interventional2005-01-31Completed
A 48-Week, Randomized, Open-Label Phase 3B Study Comparing the Antiviral Efficacy and Safety of ATV/RTV 3TC With ATV/RTV Plus TDF/FTC In HIV-1-Infected, Treatment-Naïve Subjects, Followed By a 48-Week Period on ATV/RTV Plus 3TC [NCT01620944]Phase 33 participants (Actual)Interventional2012-07-31Terminated(stopped due to Business objectives have changed)
Low Dose Anti-inflammatory Radiotherapy for the Treatment of Pneumonia by COVID-19: Multi-central Prospective Study [NCT04380818]106 participants (Anticipated)Interventional2020-06-05Recruiting
Maraviroc Switch Central Nervous System (CNS) Substudy: a Substudy of MARCH, a Randomised, Open-label Study to Evaluate the Efficacy and Safety of Maraviroc (MVC) as a Switch for Either Nucleoside or Nucleotide Analogue Reverse Transcriptase Inhibitors (N [NCT01637233]28 participants (Actual)Observational2012-06-30Completed
Maraviroc Switch Collaborative Study Renal Substudy [NCT01637259]Phase 476 participants (Actual)Interventional2012-06-30Completed
Optimal Dosing of 1st Line Antituberculosis and Antiretroviral Drugs in Children (a Pharmacokinetic Study) [NCT01637558]Phase 4200 participants (Actual)Interventional2012-11-30Completed
To Determine the Effect of the Modified SQV/r (Saquinavir-boosted by Ritonavir) Regimen (500/100 mg for the 1st Week Followed by 1000/100 mg for the 2nd Week) on the QTc Interval, Pharmacokinetics, and Antiviral Activity in Treatment-naive HIV-1 Infected [NCT01638650]Phase 123 participants (Actual)Interventional2012-01-31Completed
A Three-Part Study Evaluating the Pharmacokinetics of Intravenous (IV) Danoprevir (DNV)/Oral Low-Dose Ritonavir (RTV), the Absolute Bioavailability of DNV With and Without Oral Low-Dose RTV, and the Effect of Oral Cyclosporine on IV DNV/Oral Low-Dose RTV [NCT01654211]Phase 133 participants (Actual)Interventional2012-07-31Completed
A Phase 1, Open-Label, Parallel Study to Evaluate the Effect of Multiple Doses of Isavuconazole on the Pharmacokinetics of Multiple Doses of Lopinavir/Ritonavir and the Effects of Lopinavir/Ritonavir on the Pharmacokinetics of Multiple Doses of Isavuconaz [NCT01660477]Phase 168 participants (Actual)Interventional2012-06-30Completed
Efficacy and Safety of Treatment Against Hepatitis C Virus Infection Based on Direct-acting Antivirals in Real-life Conditions: The GEHEP Cohort [NCT02333292]1,128 participants (Actual)Observational2014-12-31Completed
A Pharmacokinetic Study of Super-boosted Lopinavir/Ritonavir in Combination With Rifampin in HIV-1-infected Patients With Tuberculosis. [NCT01700790]Phase 49 participants (Actual)Interventional2016-02-29Terminated(stopped due to No Further Funding)
Early Access to TMC114 in Combination With Low-dose Ritonavir (TMC114/r) and Other Antiretrovirals (ARVs) for Treatment-naive or TMC114-naive, Early Treatment Experienced in HIV-1 Infected Patients [NCT01702090]Phase 410 participants (Actual)Interventional2012-02-29Completed
A Phase IIIb, Randomized, Open-label Study of the Safety and Efficacy of GSK1349572 (Dolutegravir, DTG) 50 mg Once Daily Compared to Darunavir/Ritonavir (DRV/r) 800 mg/100 mg Once Daily Each Administered With Fixed-dose Dual Nucleoside Reverse Transcripta [NCT01449929]Phase 3488 participants (Actual)Interventional2011-10-31Completed
A Multi Center, Sequential, Open-Label, Multiple-Dose Study of Setrobuvir (STV) Alone and With Co-Administration of Ritonavir-boosted Danoprevir to Evaluate the Safety, Tolerability and Pharmacokinetics of STV, DNV, and Ritonavir (RTV) in Subjects With Mi [NCT01714154]Phase 118 participants (Actual)Interventional2012-11-30Completed
A Phase IV, Open-label Single-arm Study Investigating the Pharmacokinetics and Pharmacodynamics of the Antiretroviral Combination of Rilpivirine and Ritonavirboosted Darunavir in Therapy-naive HIV-1 Infected Patients. [NCT01736761]Phase 436 participants (Actual)Interventional2012-12-31Completed
STUDY TO EVALUATE SAFETY, TOLERABILITY, PHARMACOKINETICS, AND ANTIVIRAL ACTIVITY OF RITONAVIR-BOOSTED DANOPREVIR IN COMBINATION WITH PEGINTERFERON ALFA-2A PLUS RIBAVIRIN IN TREATMENT-NAÏVE PATIENTS OF ASIAN ORIGIN WHO HAVE CHRONIC HEPATITIS C GENOTYPE 1 W [NCT01749150]Phase 261 participants (Actual)Interventional2013-04-30Completed
An Open-Label Phase 3B Study in HIV-Infected Individuals With Viremia on or After Their First-Line Non-Nucleoside Reverse Transcriptase Inhibitor or Integrase Inhibitor-Based Regimen and Starting a Second-Line Regimen Consisting of ATV/RTV or DRV/RTV With [NCT01605084]Phase 30 participants (Actual)Interventional2012-06-30Withdrawn
Effects of Gastric pH on the Pharmacokinetics of Atazanavir in Healthy Volunteers [NCT01759875]Phase 18 participants (Actual)Interventional2013-01-31Completed
Nevirapine vs Ritonavir-boosted Lopinavir in ART HIV-infected Adults in a Resource-limited Setting; a Randomized, Multicenter, Parallel Group Study [NCT01772940]Phase 4425 participants (Actual)Interventional2008-12-31Completed
A PHASE I, MULTIPLE DOSE, OPEN-LABEL PHARMACOKINETIC STUDY OF NIRMATRELVIR/RITONAVIR IN HEALTHY LACTATING WOMEN [NCT05441215]Phase 18 participants (Anticipated)Interventional2022-12-12Recruiting
A Double-Blind, Randomized, Placebo-Controlled, Single Dose Escalation First Time in Human Study to Investigate the Safety, Tolerability and Pharmacokinetics of GSK2838232 and to Evaluate the Effect of Food and Ritonavir on GSK2838232 in Healthy Subjects [NCT01802918]Phase 117 participants (Actual)Interventional2013-02-18Completed
A Study to Evaluate the Pharmacokinetics and Safety of Oral Single-Dose JNS011 Tablet in Combination With Low-Dose Ritonavir Capsule in Healthy Japanese Adult Males [NCT01810887]Phase 48 participants (Actual)Interventional2008-05-31Completed
Pharmacokinetic Interactions Between Telaprevir and Un-boosted Atazanavir in HIV/HCV-co-infected Patients Under Treatment for Genotype 1 Chronic Hepatitis C. [NCT01818856]Phase 114 participants (Anticipated)Interventional2012-12-31Completed
An Open-Label Pilot Study to Evaluate the Antiviral Activity, Safety, Tolerability, and Pharmacokinetics of ABT-450 With Ritonavir (ABT-450/r) Dosed in Combination With ABT-530, With and Without Ribavirin (RBV) in Treatment-Naïve Subjects With Genotype 3 [NCT02068222]Phase 210 participants (Actual)Interventional2014-04-30Completed
A Pilot Randomized, Open Label Study to Evaluate Efficacy and Safety of the Combination of RAL+ATV/r in Comparison With TDF/FTC+ATV/r in HIV Infected Patients, Who Failed an Initial NNRTI Containing Regimen [NCT01829802]Phase 450 participants (Anticipated)Interventional2014-05-31Active, not recruiting
A Phase2/3, Multi-center, Randomized, Double-blind, Placebo-parallel Controlled Study to Investigate the Efficacy and Safety of Ravidasvir in Combination With Danoprevir/r and Ribavirin(RBV) in Treatment-naive, Non-cirrhotic, Chronic Hepatitis C Genotype [NCT03362814]Phase 2/Phase 3425 participants (Actual)Interventional2017-07-01Completed
A Multicenter, Randomized, Active Controlled, Open Label, Platform Trial on the Efficacy and Safety of Experimental Therapeutics for Patients With COVID-19 (Caused by Infection With Severe Acute Respiratory Syndrome Coronavirus-2) [NCT04351724]Phase 2/Phase 3500 participants (Anticipated)Interventional2020-04-16Recruiting
An Open-Label Study to Evaluate the Safety, Efficacy, and Pharmacokinetics of Ombitasvir/ABT-450/Ritonavir (Ombitasvir/ABT-450/r) and Dasabuvir Co-administered With or Without Sofosbuvir (SOF) and Ribavirin (RBV) in Direct-Acting Antiviral Agent (DAA) Tre [NCT02356562]Phase 229 participants (Actual)Interventional2015-02-03Completed
An Investigation Into Beneficial Effects of Interferon Beta 1a, Compared to The Base Therapeutic Regiment in Moderate to Severe COVID-19: A Randomized, Double-Blind, Placebo-Controlled, Clinical Trial [NCT04350671]Phase 440 participants (Anticipated)Interventional2020-04-15Enrolling by invitation
The Effect of Protease Inhibitors on the Pharmacokinetics of Oral Norethindrone Contraception [NCT01667978]35 participants (Actual)Interventional2012-06-30Completed
An Open-Label, Treatment Duration-Ranging Study to Evaluate the Safety and Efficacy of Ombitasvir/Paritaprevir/ Ritonavir (Ombitasvir/ABT-450/r) and Dasabuvir Co-administered With Sofosbuvir (SOF) With and Without Ribavirin (RBV) in Direct-Acting Antivira [NCT02399345]Phase 310 participants (Actual)Interventional2015-03-31Completed
Randomized, Placebo-Controlled, Multiple-Dose Study to Evaluate the Pharmacodynamics, Safety and Pharmacokinetics of BMS-955176 (Double-Blinded) and BMS-955176 With Atazanavir +/- Ritonavir (Open-Labeled) in HIV-1 Infected Subjects [NCT01803074]Phase 2107 participants (Actual)Interventional2013-04-04Completed
Post-marketing Surveillance Study of the Effectiveness and Safety of New Oral Antivirals for Outpatients With Mild-moderate COVID-19 [NCT05894603]211 participants (Actual)Observational2022-08-01Active, not recruiting
A Multicenter, Open-Label, 24-Week Study to Evaluate the Safety and Activity of Indinavir Sulfate 800 Mg b.i.d. in Combination With Ritonavir 200 Mg b.i.d. and Two NRTIs in HIV-1 Infected Patients Who Failed PI Therapy [NCT00002361]Phase 230 participants InterventionalCompleted
Ritonavir (ABT-538)/Indinavir Combination Treatment in HIV-Infected Patients Previously Receiving Indinavir 800 mg TID [NCT00002223]0 participants InterventionalCompleted
A Multicenter, Open-Label, 24-Week Pilot Study to Evaluate the Safety and Efficacy of Indinavir Sulfate 800 Mg b.i.d. in Combination With Ritonavir 100 Mg/d4T/3TC b.i.d. in HIV-Infected Individuals [NCT00002241]Phase 280 participants InterventionalActive, not recruiting
A Single Centre, Open-label, Randomized, Parallel Group, Multiple Dose Comparison of the Effect of TPV 750 mg and RTV 200 mg or TPV 500 mg and RTV 100 mg, Administered Twice Daily, on the Pharmacokinetic Characteristics of Norethindrone-Ethinyl Estradiol [NCT02245438]Phase 152 participants (Actual)Interventional2002-05-31Terminated
Prophylactic Effect of Nirmatrelvir/Ritonavir and Ursodeoxycholic Acid on Reducing Complications After Cardiac Surgery During COVID-19 Pandemics: A Prospective, Randomized Controlled, Multicenter Trial [NCT05690646]Phase 4491 participants (Actual)Interventional2023-01-28Active, not recruiting
A Phase 2b, Double-Blind, Placebo-Controlled, Exploratory Randomized Trial to Determine the Bone, Immunologic, Virologic, and Neurocognitive Effects of a Novel Maraviroc-Containing Antiretroviral Regimen in Treatment-Naïve Patients Infected With R5-Tropic [NCT01400412]Phase 2262 participants (Actual)Interventional2012-01-17Completed
A Phase IIb Randomized, Controlled, Partially-Blinded Trial to Investigate Safety, Efficacy and Dose-Response of BMS-663068 in Treatment-experienced HIV-1 Subjects, Followed by an Open-Label Period on the Recommended Dose [NCT01384734]Phase 2254 participants (Actual)Interventional2011-07-26Completed
A Study on ART Naïve Patients On Different Regimens to Treat Hiv (a Phase 4 Study) [NCT01445223]Phase 4242 participants (Actual)Interventional2004-04-30Completed
Interferon-Free Therapy for Chronic Hepatitis C Virus Genotype 1 Infection in Participants With HIV-1 Coinfection Receiving Concurrent Antiretroviral Therapy (C_ASCENT) [NCT02194998]Phase 246 participants (Actual)Interventional2015-09-16Terminated(stopped due to The study closed to accrual before the planned accrual goal was attained due to the availability of newer directly-acting antiviral (DAA) treatments for HCV.)
A Study to Evaluate the Safety, Tolerability, Pharmacokinetics of RAY1216 and the Effect of Food on RAY1216 Pharmacokinetics in Healthy Adult Participants [NCT05829551]Phase 188 participants (Actual)Interventional2022-05-20Completed
A PHASE 1, OPEN-LABEL, FIXED SEQUENCE, 2-PERIOD CROSSOVER STUDY TO ESTIMATE THE EFFECT OF CARBAMAZEPINE ON THE PHARMACOKINETICS OF PF-07321332 BOOSTED WITH RITONAVIR IN HEALTHY PARTICIPANTS [NCT04962230]Phase 112 participants (Actual)Interventional2021-07-15Completed
HERV-K Suppression Using Antiretroviral Therapy in Volunteers With Amyotrophic Lateral Sclerosis (ALS) [NCT02437110]Phase 1122 participants (Actual)Interventional2019-04-01Active, not recruiting
Pharmacokinetics of Plasma Lopinavir/Ritonavir Over a 12 Hour Dosing Interval Following Administration of 400/100, 200/150, and 200/50 mg Twice Daily to HIV-negative Healthy Volunteers [NCT00985543]Phase 122 participants (Actual)Interventional2009-10-31Completed
A Single-centre, Open-label Study of Multiple Doses of Tipranavir 500 mg and Ritonavir 200 mg (Twice Daily) on the Pharmacokinetic Characteristics of Methadone Administered as a Single Dose in Healthy Volunteers [NCT02245451]Phase 115 participants (Actual)Interventional2005-01-31Completed
A Study on Safety, Tolerability and Pharmacokinetics of Ritonavir-boosted Danoprevir in Single and Multiple Doses in Healthy Volunteers [NCT03019991]Phase 118 participants (Actual)Interventional2015-10-31Completed
A Pilot, Open-Label Study of Adjunctive Therapy With Lovaza® in Hypertriglyceridemic, HIV-Infected Subjects Who Switched Protease Inhibitor to Once-Daily Lexiva® 1400mg Plus Norvir® 100mg Plus Optimized Background [NCT01010399]Phase 436 participants (Actual)Interventional2009-09-30Completed
An Open Label, Multicentre, International Pilot Study of Paritaprevir/Ritonavir, Ombitasvir, Dasabuvir With or Without Ribavirin or Glecaprevir/Pibrentasvir for People With Recently Acquired Hepatitis C Virus Infection With or Without HIV Co-infection. [NCT02634008]Phase 383 participants (Actual)Interventional2016-06-30Completed
Multi-Drug Antiretroviral Therapy for Heavily Pretreated Pediatric AIDS Patients: A Phase I Proof of Concept Trial [NCT00001108]Phase 16 participants InterventionalCompleted
A Phase IIIB Open-Label Trial Replacing Saquinavir HGC (Ro 31-8959) With Saquinavir SGC (Ro 31-8959) in Combination With Other Antiretroviral Drugs in Patients With HIV-1 Infection [NCT00002374]Phase 3120 participants InterventionalCompleted
An Interventional, Double-Blinded, 2-Arm Study to Investigate the Efficacy of Orally Administered Nirmatrelvir/Ritonavir Compared With Placebo/Ritonavir in Non-hospitalized Adult Participants Suffering From Post-COVID [NCT05823896]Phase 2400 participants (Anticipated)Interventional2023-05-01Recruiting
An Open-Label, Phase II Trial to Evaluate the Steady-State Pharmacokinetics, Safety, and Efficacy of 1592U89 in Combination With Selected HIV-1 Protease Inhibitors in Antiretroviral-Naive, HIV-1-Infected Patients. [NCT00002440]Phase 280 participants InterventionalCompleted
A Phase II Study of ABT-378/Ritonavir and Efavirenz in Multiple Protease Inhibitor-Experienced Subjects [NCT00004582]Phase 20 participants InterventionalCompleted
Evaluation of Additional Treatments for COVID-19: a Randomized Trial in Niger [NCT04409483]Phase 30 participants (Actual)Interventional2020-06-01Withdrawn(stopped due to Epidemic dynamics)
Open Multicentre Study for Assessment of the Safety and Efficacy Against COVID-19 of the Drug JTBC00201 (PROMOMED RUS LLC, Russia) in the Adult Population [NCT05601167]Phase 3264 participants (Actual)Interventional2021-02-17Completed
Cost-effectiveness of Different Antiretroviral Treatment in Patients HIV Naive. Randomized Clinical, Not Masked, Trial Comparing DRVr3TC, ABC3TC (Kivexa) RPV, or EVG COBI FTC TDF (Stribild) for 48 Weeks [NCT02470650]Phase 4150 participants (Anticipated)Interventional2015-06-30Recruiting
A Phase I, Single-center, Open Label Clinical Study, to Evaluate the Pharmacokinetic Character of GLS4 Combined With RTV or TAF Alone or GLS4 and RTV and TAF Combination Administration in Healthy Subjects [NCT04551261]Phase 128 participants (Actual)Interventional2021-01-10Completed
A Randomised, Controlled, Open-Label Trial to Compare Brachial Artery Reactivity and Cardiovascular Risk of a Treatment Simplification by Darunavir/Ritonavir (DRV/r) 800/100 mg O.D. Versus a Triple Combination Therapy Containing DRV/r in HIV-1 Infected Su [NCT01391013]Phase 230 participants (Actual)Interventional2009-06-30Completed
Drug-Drug Interaction Study to Assess the Effects of Steady-State Darunavir/Ritonavir on Steady-State Pitavastatin in Healthy Adult Volunteers [NCT01422369]Phase 428 participants (Actual)Interventional2011-04-30Completed
Comparative Therapeutic Efficacy and Safety of Remdesivir Plus Lopinavir/ Ritonavir and Tocilizumab Versus Hydroxychloroquine Plus Ivermectin and Tocilizumab in COVID-19 Patients. [NCT04779047]Phase 4150 participants (Anticipated)Interventional2020-10-01Recruiting
Efficacy of Novel Agents for Treatment of SARS-CoV-2 Infection Among High-Risk Outpatient Adults: An Adaptive Randomized Platform Trial [NCT04354428]Phase 2/Phase 3289 participants (Actual)Interventional2020-04-16Terminated(stopped due to Low number of events contributing to primary outcome)
A Single-centre, Open-label, Randomised, Crossover, Drug-drug Interaction Study to Investigate the Effect of a Single Oral Dose of Ritonavir on the Single Dose Pharmacokinetics of ASP2151 in Healthy Men [NCT02223351]Phase 148 participants (Actual)Interventional2014-09-30Completed
A Phase 2 Study to Evaluate the Safety, Tolerability, Antiviral Activity, and Pharmacokinetics of ABT-450 With Ritonavir (ABT-450/r) and ABT-267 in Japanese Adults With Chronic Hepatitis C Virus Infection [NCT01672983]Phase 2110 participants (Actual)Interventional2012-07-31Completed
A Randomized Phase II Study of the Safety, Immunologic, and Virologic Effects of Cyclosporine A in Conjunction With Trizivir(R) and Kaletra(R) Versus Trizivir(R) and Kaletra(R) Alone During Primary HIV-1 Infection [NCT00084149]Phase 254 participants (Actual)Interventional2004-02-29Completed
An Open-Label, Multicenter Study to Evaluate the Efficacy and Safety of Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir With Low-Dose Ribavirin QD in Subjects With Genotype 1a Chronic Hepatitis C Virus Infection (GEODE II) [NCT02609659]Phase 3105 participants (Actual)Interventional2015-10-28Completed
An Exploratory Study to Evaluate Immune Restoration Following Removal of Viral Antigen in Treatment-Naïve and Treatment-Experienced Adults With Genotype (GT) 1a Chronic Hepatitis C Virus (HCV) Infection Administered Ombitasvir/ ABT-450/Ritonavir With Dasa [NCT02476617]Phase 325 participants (Actual)Interventional2015-06-30Completed
Real World Evidence of the Effectiveness of Paritaprevir/Ritonavir - Ombitasvir, ± Dasabuvir, ± Ribavirin in Patients With Chronic Hepatitis C - An Observational Study in Belgium [NCT02581163]314 participants (Actual)Observational2015-10-07Completed
An Exploratory Study to Evaluate the Kinetics of Viral Load Decline With Ombitasvir/ABT 450/Ritonavir (Ombitasvir/ABT-450/r) and Dasabuvir Therapy With Low Dose Ribavirin (RBV), Full Dose RBV or RBV Add-On in Treatment Naïve Adults With Genotype 1a Chroni [NCT02493855]Phase 246 participants (Actual)Interventional2015-06-30Completed
An Open-Label, Proof of Concept Study To Evaluate the Efficacy and Justification Of OBV/PTV/r and DSV In Adults With Chronic Hepatitis C Virus (HCV) Infection Genotype 2K/1B- NINJA Study [NCT03050905]Phase 47 participants (Actual)Interventional2017-06-22Completed
Real World Evidence of the Effectiveness of Paritaprevir/r - Ombitasvir, ± Dasabuvir, ± Ribavirin in Patients With Chronic Hepatitis C - An Observational Study in Canada (AMBER) [NCT02581189]565 participants (Actual)Observational2015-10-13Completed
A Phase I, Three-Arm Safety, Tolerability, and Pharmacokinetic Interaction Study of PA-824, an Investigational Nitroimidazole for the Treatment of Tuberculosis, Together With Efavirenz, Ritonavir-Boosted Lopinavir, or Rifampin [NCT01571414]Phase 152 participants (Actual)Interventional2012-05-31Completed
A Randomized Study to Evaluate the Effect of Switching From Efavirenz to Atazanavir/ Ritonavir on Lipoatrophy and Mitochondrial Dysfunction in HIV-infected Subjects With Good Virologic Suppression [NCT01194856]Phase 41 participants (Actual)Interventional2010-10-31Terminated(stopped due to Closed due to low enrollment)
Evaluation of Efficacy of Pharmacotherapy Treatment of COVID- 19 Infection Using Oral Levamisole and Formoterol+Budesonide Inhaler and Comparison of This Treatment Protocol With Standard National Treatment of the Disease [NCT04331470]Phase 2/Phase 330 participants (Anticipated)Interventional2020-04-04Recruiting
[NCT01579019]Phase 20 participants (Actual)Interventional2012-07-31Withdrawn
A Randomized, Single-center, Open-label, One-sequence, Two-period Crossover Study in 3 Parts to Investigate the Effects of Multiple Doses of Ketoconazole (Part 1), Rifampicin (Part 2), and Ritonavir-boosted Atazanavir (Part 3) on the PK of a Single Dose o [NCT01591850]Phase 151 participants (Actual)Interventional2011-09-30Completed
A Study to Evaluate the Potential Drug-Drug Interaction Between Danoprevir When Coadministered With Low-Dose Ritonavir and Tenofovir Disoproxil Fumarate or Atazanavir in Healthy Adult Volunteers [NCT01592305]Phase 140 participants (Actual)Interventional2012-05-31Completed
An Up to Two-Part Relative Bioavailability Study of Ritonavir-Boosted Danoprevir Fixed Dose Combination Tablets as Compared to the Reference Phase 2 Ad Hoc Combination Tablets in Healthy Adult Volunteers [NCT01592318]Phase 142 participants (Actual)Interventional2012-05-31Completed
Safety and Efficacy of Switching a Stable Combined Antiretroviral Therapeutic Regimen to Atazanavir With Ritonavir Plus Lamivudine in Treatment Experienced HIV Positive Patients With Full and Stable Virological Suppression [NCT01599364]Phase 4266 participants (Actual)Interventional2014-04-30Completed
Randomized Controlled Pilot Study of Highly Active Anti-Retroviral Therapy for Patients With Primary Biliary Cirrhosis [NCT01614405]13 participants (Actual)Interventional2012-06-30Completed
PREZISTA or INTELENCE Switch Evaluation in Virologically Suppressed Patients Naïve to Darunavir or Etravirine and Who Are Intolerant of Their Current or Prior Combination Antiretroviral Therapy Regimen: A Phase IV, Open-label, Multicentre Observational Tr [NCT01615601]77 participants (Actual)Observational2011-10-31Completed
Phase 1, Open Label Study to Evaluate the Effect of Omeprazole and Ritonavir on GSK2336805 Pharmacokinetics in Healthy Adults [NCT01458054]Phase 128 participants (Actual)Interventional2011-09-27Completed
A Phase 1, Single Center, Open-label Study of PF-07321332 Administrated as Multiple Oral Doses in Healthy Chinese Participants. [NCT05339334]Phase 114 participants (Actual)Interventional2022-03-10Completed
An Open-Label Pilot Study to Evaluate the Safety, Tolerability, Pharmacokinetics, and Antiviral Activity of ABT-450 With Ritonavir (ABT-450/r) Dosed in Combination With ABT-072 and Ribavirin (RBV) in Treatment-Naive Subjects With Genotype 1 Chronic Hepati [NCT01221298]Phase 211 participants (Actual)Interventional2010-10-31Completed
A Phase 1, Multicenter, Open-label, Single Sequence Crossover Study to Evaluate Drug-drug Interaction Potential of OATP1B/CYP3A Inhibitor on the Pharmacokinetics of DS-8201a in Subjects With HER2-expressing Advanced Solid Malignant Tumors [NCT03383692]Phase 140 participants (Actual)Interventional2018-01-12Completed
A Phase 1, Randomized, Double-blind, Single-and Multiple-dose Escalation, Placebo-controlled Study to Evaluate the Safety, Tolerability, and Pharmacokinetics of QLS1128 and in Combination With Ritonavir in Healthy Participants [NCT05458076]Phase 186 participants (Anticipated)Interventional2022-07-03Recruiting
An Open Label, Randomized, Parallel-group Pharmacokinetics Trial of Tipranavir / Ritonavir (TPV/RTV), Alone or in Combination With RTV-boosted Saquinavir (SQV), Amprenavir (APV), or Lopinavir (LPV), Plus an Optimized Background Regimen, in Multiple Antire [NCT00056641]Phase 2328 participants Interventional2003-02-18Completed
Effect of Multiple Dosing With 240 mg QD BI 201335 on the Steady-state Pharmacokinetics of 800 mg QD Darunavir Coadministered With 100 mg QD Ritonavir (DRV/r) in Healthy Male and Female Volunteers (an Open-label, Multiple-dose, Single Group, Single Fixed [NCT01374802]Phase 114 participants (Actual)Interventional2011-06-30Completed
RAD-1: A Phase I/II Antiretroviral Management Algorithm for Pediatric Subjects of Four-Drug Combination Therapies Based on Prior Antiretroviral Experience [NCT00000902]Phase 1217 participants InterventionalCompleted
A Randomized Study of the Virologic Efficacy of Different Antiretroviral (AR) Treatment Strategies in HIV-Infected Individuals With Detectable Plasma HIV RNA Measurements After at Least 16 Weeks on Their Initial Protease Inhibitor-Containing AR Regimens [NCT00000914]800 participants InterventionalCompleted
A Phase II/III Prospective, Multicenter, Randomized, Controlled Trial Comparing the Safety and Efficacy of Three Clarithromycin-Containing Combination Drug Regimens for the Treatment of Disseminated Mycobacterium Avium Complex (MAC) Disease in Persons Wit [NCT00001058]Phase 2246 participants InterventionalCompleted
A Randomized Phase IIIB Comparative Study to Evaluate Saquinavir Soft Gel Capsule (SGC) TID Regimen in Combination With Two NRTIs Versus Saquinavir Soft Gel Capsule (SGC) BID Regimen in Combination With Two NRTIs Versus Saquinavir Soft Gel Capsule (SGC) B [NCT00002378]Phase 3825 participants InterventionalCompleted
Efficacy and Safety of All-Oral Combination of Narlaprevir/Ritonavir and Sofosbuvir in Treatment-naïve Patients With Chronic Hepatitis C Genotype 1 [NCT04246723]Phase 285 participants (Actual)Interventional2019-05-06Completed
A Controlled Phase 2 Trial Assessing Three Doses of T-20 in Combination With Abacavir, Amprenavir, Ritonavir, and Efavirenz in HIV-1 Infected Adults [NCT00002239]Phase 268 participants Interventional1999-05-31Completed
Pharmacokinetic Interaction Studies of Amprenavir (APV), Efavirenz (EFV), and a Second Protease Inhibitor in HIV-Seronegative Volunteers [NCT00005762]90 participants Interventional2001-03-31Completed
A Randomized, Open-Label, Pilot Treatment Trial Evaluating Cellular Dynamics and Immune Restoration in Treatment-Naive HIV-Infected Subjects Receiving Either the Protease Inhibitor LPV/r or the Nucleoside Analogue Reverse Transcriptase Inhibitors d4T/3TC/ [NCT00004855]55 participants InterventionalCompleted
A Phase IV, Open-Label, Randomized Study to Compare the Efficacy and Safety of Epivir/Ziagen/Zerit (3TC/ABC/d4T) Versus Epivir/Ziagen/Sustiva (3TC/ABC/EFV) Versus Epivir/Ziagen/Agenerase/Norvir (3TC/ABC/APV/RTV) for 96 Weeks in the Treatment of HIV-1 Infe [NCT00005017]Phase 4300 participants InterventionalActive, not recruiting
A Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Efficacy and Safety of ABT-450/Ritonavir/ABT-267 (ABT-450/r/ABT-267) and ABT-333 in Treatment-Naïve and Treatment-Experienced, Non-Cirrhotic Asian Adults With Subgenotype 1b Chronic Hepa [NCT02517515]Phase 3650 participants (Actual)Interventional2015-07-31Completed
A Phase IV, Multisite Study of the Treatment of Chronic Hepatitis C Virus Infection Genotype 1 in a Real World Large Health Maintenance Organization: An Evaluation of Real World Sustained Virological Response and Patient Reported Outcomes [NCT02461745]Phase 4200 participants (Actual)Interventional2015-06-30Completed
Open-label Study to Evaluate the Safety and Efficacy of the Combination of Ombitasvir, Paritaprevir/r ± Dasabuvir With or Without Ribavirin (RBV) in Adult Patients With GT1 or GT4 Chronic HCV Infection and Response to Prior Treatment of Early Stage Hepato [NCT02504099]Phase 33 participants (Actual)Interventional2015-07-31Terminated(stopped due to Study stopped due to low enrollment)
A Follow-up Study to Assess Resistance and Durability of Response to AbbVie Direct-Acting Antiviral Agent (DAA) Therapy in Subjects Who Participated in Phase 2 or 3 Clinical Studies for the Treatment of Chronic Hepatitis C Virus (HCV) Infection [NCT01773070]Phase 3478 participants (Actual)Interventional2013-06-30Completed
A Phase II Rolling Arm Master Protocol (PRAM) of Novel Antiretroviral Therapy in Stable Experienced HIV-Infected Children. PRAM-2: A Phase I/II Randomized, Multicenter Protocol Comparing Four Antiretroviral Regimens Containing Combinations of Protease Inh [NCT00001091]Phase 1200 participants InterventionalCompleted
An Open Label, Randomized, Multicenter Study to Evaluate Fortovase (Saquinavir) SGC QD, Norvir (Ritonavir) QD Plus Two NRTIs Vs Sustiva (Efavirenz) QD Plus Two NRTIs in HIV Infected Patients [NCT00002447]Phase 3146 participants Interventional1999-10-31Completed
Safety and Efficacy of Ritonavir (ABT-538) in Combination With Nelfinavir in HIV-Infected Subjects. [NCT00002201]0 participants InterventionalCompleted
Appropriate Evaluation of Antiviral Therapy With Nirmatrelvir-ritonavir in Hospitalized Patients: a Multicenter Retrospective Study in China [NCT05749445]200 participants (Anticipated)Observational2022-12-15Recruiting
AN INTERVENTIONAL EFFICACY AND SAFETY, PHASE 2, DOUBLE-BLIND, 2-ARM STUDY TO INVESTIGATE ORALLY ADMINISTERED NIRMATRELVIR/RITONAVIR COMPARED WITH PLACEBO/RITONAVIR FOR THE TREATMENT OF SEVERE COVID-19 IN HOSPITALIZED PARTICIPANTS WHO ARE IMMUNOCOMPROMISED [NCT05545319]Phase 20 participants (Actual)Interventional2022-12-13Withdrawn(stopped due to Termination due to challenges related to the operational feasibility of the study, taking into account the current epidemiology and declining hospitalization rates for severe COVID-19.)
Chemoprophylaxis of SARS-CoV-2 Infection (COVID-19) in Exposed Healthcare Workers : A Randomized Double-blind Placebo-controlled Clinical Trial [NCT04328285]Phase 3118 participants (Actual)Interventional2020-04-14Terminated(stopped due to French authority's decision)
A Multipart, Open-label Study to Evaluate the Safety and Efficacy of Ombitasvir/Paritaprevir/Ritonavir With and Without Dasabuvir Coadministered With and Without Ribavirin in Adults With Genotype 1 or 4 Chronic Hepatitis C Virus Infection and Human Immuno [NCT01939197]Phase 2/Phase 3318 participants (Actual)Interventional2013-08-30Completed
Randomized, Open-Label, Multiple-Dose Study to Evaluate the Effect of Famotidine on the Pharmacokinetics of Atazanavir/Ritonavir/Tenofovir in Healthy Subjects [NCT00365339]Phase 140 participants Interventional2006-04-30Completed
A PHASE 1, OPEN-LABEL, RANDOMIZED, SINGLE DOSE, CROSSOVER STUDY TO ESTIMATE THE RELATIVE BIOAVAILABILITY OF NIRMATRELVIR (PF-07321332) /RITONAVIR ORAL POWDER IN 3 DIFFERENT FOOD DELIVERY VEHICLES RELATIVE TO THE NIRMATRELVIR (PF-07321332) /RITONAVIR COMME [NCT05544786]Phase 112 participants (Actual)Interventional2022-09-28Completed
International, Multicenter, Randomized, Double Blind, Active-controlled, Parallel-group Phase III Study of Narlaprevir/Ritonavir and Pegylated Interferon/Ribavirin in 2 Patient Populations - naïve and Treatment Failure Patients With Genotype 1 Chronic Hep [NCT03833362]Phase 3420 participants (Actual)Interventional2014-05-07Completed
Open-Label, Multiple Dose Study to Determine the Relative Bioavailability of Atazanavir (ATV) 400 mg Administered With Ritonavir (RTV) and Efavirenz (EFV) Compared to Atazanavir 300 mg Administered With Ritonavir Alone in Healthy Subjects [NCT00357188]Phase 122 participants Interventional2006-07-31Completed
AN INTERVENTIONAL EFFICACY AND SAFETY, PHASE 2/3, DOUBLE-BLIND, 2-ARM STUDY TO INVESTIGATE ORALLY ADMINISTERED PF-07321332/RITONAVIR COMPARED WITH PLACEBO IN NONHOSPITALIZED SYMPTOMATIC ADULT PARTICIPANTS WITH COVID-19 WHO ARE AT INCREASED RISK OF PROGRES [NCT04960202]Phase 2/Phase 32,246 participants (Actual)Interventional2021-07-16Completed
Effect of Atazanavir Administered With and Without Ritonavir on the Pharmacokinetics of the Cytochrome P450 2C8 Substrate Rosiglitazone in Healthy Subjects [NCT00362726]Phase 114 participants Interventional2006-09-30Completed
Understanding HCV Reinfection Rates in an Incarcerated Population After Cure With Interferon Free HCV Treatment: A Pilot Project [NCT02460133]Phase 444 participants (Actual)Interventional2015-07-31Active, not recruiting
An Open-Label Study to Evaluate the Safety and Efficacy of Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir With or Without Ribavirin (RBV) in Adults With Genotype 1 Chronic Hepatitis C Virus (HCV) Infection, With Severe Renal Impairment or End-Stage Renal [NCT02207088]Phase 368 participants (Actual)Interventional2014-09-23Completed
Comparative Therapeutic Efficacy and Safety of Remdesivir Versus Lopinavir/ Ritonavir and Remdesivir Combination in COVID-19 Patients [NCT04738045]Phase 490 participants (Anticipated)Interventional2020-11-01Recruiting
A Phase 1 Clinical Study to Assess the Effect of Darunavir/Ritonavir or Lopinavir/Ritonavir on the Pharmacokinetics of Daclatasvir in Healthy Subjects [NCT02159352]Phase 149 participants (Actual)Interventional2014-06-30Completed
A Drug Interaction Study to Assess the Pharmacokinetics of Narlaprevir and Antiretroviral Drugs [NCT03537404]Phase 136 participants (Actual)Interventional2017-04-24Completed
An Open-Label, Multiple Ascending Dose Study to Assess the Safety, Tolerability, Pharmacokinetics and Antiviral Activity of ABT-267 in HCV Infected Subjects [NCT01563536]Phase 212 participants (Actual)Interventional2012-02-29Completed
An Open-Label, Sequential Arm, Multicenter Study to Evaluate the Antiviral Activity, Safety and Pharmacokinetics of ABT-450 With Ritonavir (ABT-450/r) Dosed in Combination With ABT-267 With and Without Ribavirin (RBV) in Treatment-Naïve Subjects With Geno [NCT01458535]Phase 261 participants (Actual)Interventional2011-09-30Completed
A Randomized, Open-Label, Multicenter Study to Evaluate the Antiviral Activity, Safety, and Pharmacokinetics, of ABT-450 With Ritonavir (ABT-450/r) in Combination With ABT-267 and/or ABT-333 With and Without Ribavirin (RBV) for 8, 12 or 24 Weeks in Treatm [NCT01464827]Phase 2580 participants (Actual)Interventional2011-10-31Completed
The Influence of Lopinavir/Ritonavir on Gemfibrozil Pharmacokinetics in Healthy Volunteers [NCT00474201]15 participants (Actual)Interventional2007-05-31Completed
An Open-Label, Multicenter Study to Evaluate Long-term Outcomes With ABT-450/Ritonavir/ABT-267 (ABT-450/r/ABT-267) and ABT-333 With or Without Ribavirin (RBV) in Adults With Genotype 1 Chronic Hepatitis C Virus (HCV) Infection (TOPAZ-II) [NCT02167945]Phase 3615 participants (Actual)Interventional2014-06-12Completed
Neuropsychiatric Adverse Effects of Efavirenz in Children Living With HIV in Kilimanjaro, Tanzania [NCT03227653]144 participants (Actual)Observational2017-06-19Completed
A Phase 3, Matrix Design, Partially Double-Blind, Randomized Study of the Efficacy and Safety of 50 mg Lonafarnib/100 mg Ritonavir BID With and Without 180 mcg PEG IFN-alfa-2a for 48 Weeks Compared With PEG IFN-alfa-2a Monotherapy and Placebo Treatment in [NCT03719313]Phase 3407 participants (Actual)Interventional2018-12-01Completed
Influence of Efavirenz and Ritonavir on Human Brain P-Glycoprotein Activity Using PET Imaging [NCT01668147]Phase 1/Phase 213 participants (Actual)Interventional2012-08-31Completed
Switching From Regimens Consisting of a RTV-Boosted Protease Inhibitor Plus TDF/FTC to a Combination of Raltegravir Plus Nevirapine and Lamivudine in HIV Patients With Suppressed Viremia and Impaired Renal Function (RANIA Study) (Pilot Study) Protocol MK- [NCT02116660]Phase 211 participants (Actual)Interventional2014-09-03Terminated(stopped due to This study was terminated early due to poor recruitment.)
Pharmacokinetics Study of Tenofovir in HIV-infected Thai Children Using Tenofovir-based Regimen [NCT02404259]32 participants (Actual)Interventional2010-06-30Completed
Phase I/II Study of ABT-378/Ritonavir in Protease Inhibitor Experienced HIV-Infected Patients [NCT00004580]Phase 10 participants InterventionalCompleted
A Pharmacokinetics Study Comparing Lopinavir Plasma Exposure When Given as Lopinavir/Ritonavir (1:1) in the Presence of Rifampicin and Lopinavir/Ritonavir (4:1) Without Rifampicin in HIV and TB Co-infected Children in South Africa. [NCT02348177]Phase 496 participants (Actual)Interventional2013-01-31Completed
Dose Adaptation to Offset the Pharmacokinetic Interaction Between Ticagrelor and Ritonavir in Healthy Volunteers by Population-based PK Modeling (Simcyp®) [NCT02435563]Phase 222 participants (Actual)Interventional2014-08-31Completed
Population Pharmacokinetics of Antiretroviral in Children [NCT03194165]65 participants (Actual)Observational2017-06-16Completed
Adaptive, Randomized, Non-inferiority Trial on the Use of Monoclonal Antibodies or Antivirals in Outpatients With Mild or Moderate COVID-19 [NCT05321394]Phase 3536 participants (Actual)Interventional2022-03-07Completed
A Prospective Multicenter, Randomized, Controlled Clinical Trial of the Safety and Efficacy of Azvudine vs. Nirmatrelvir-Ritonavir in Hospitalized Patients With Moderate to Severe COVID-19 Infection [NCT05697055]Phase 4410 participants (Anticipated)Interventional2023-01-21Recruiting
Efficacy and Safety of Albuvirtide for Injection Combined With LPV/r for Treatment of HIV-1-Infected Patients Failed First-line Antiretroviral Therapy [NCT02369965]Phase 3418 participants (Actual)Interventional2014-02-19Completed
AN INTERVENTIONAL EFFICACY AND SAFETY, PHASE 2/3, DOUBLE-BLIND, 2 ARM STUDY TO INVESTIGATE ORALLY ADMINISTERED PF 07321332/RITONAVIR COMPARED WITH PLACEBO IN NONHOSPITALIZED SYMPTOMATIC ADULT PARTICIPANTS WITH COVID-19 WHO ARE AT LOW RISK OF PROGRESSING T [NCT05011513]Phase 2/Phase 31,440 participants (Actual)Interventional2021-08-25Terminated(stopped due to Enrollment ceased due to a very low rate of hospitalization or death observed in the standard-risk patient population)
A Phase 2, Open-Label Study of the Safety, Tolerability, Pharmacokinetics, and Pharmacodynamic Activity of a Titrating-Dose Lonafarnib/Ritonavir in Patients Chronically Infected With Hepatitis Delta Virus [NCT02527707]Phase 215 participants (Actual)Interventional2015-09-30Completed
PROTEAse Inhibitor (DRV/Rtv) in Mono- or Triple Therapy in Suppressed HIV-1 Infected Subjects [NCT01448707]Phase 3274 participants (Actual)Interventional2012-03-15Completed
HIV Postexposure Prophylaxis With Darunavir/r (PEPDar) [NCT01516970]Phase 3312 participants (Actual)Interventional2011-11-25Completed
A Pilot Study to Assess Virologic Suppression and Immune Recovery With Raltegravir and Lopinavir/Ritonavir and Raltegravir and Emtricitabine/Tenofovir in HIV-1 Infected Treatment-naïve Subjects [NCT00654147]Phase 244 participants (Actual)Interventional2008-04-30Completed
Lopinavir/Ritonavir/Combivir vs. Abacavir/Zidovudine/Lamivudine for Virologic Efficacy and the Prevention of Mother-to-Child HIV Transmission Among Breastfeeding Women With CD4 Counts Greater Than or Equal to 200 Cells/mm3 in Botswana [NCT00270296]Phase 2730 participants (Actual)Interventional2006-06-30Completed
An Open-Label Study to Evaluate the Safety and Efficacy of ABT-450/Ritonavir/ABT-267 (ABT 450/r/ABT-267) and ABT-333 Coadministered With Ribavirin (RBV) in Treatment-Naïve and Treatment-Experienced Asian Adults With GT1b Chronic Hepatitis C Virus (HCV) In [NCT02517528]Phase 3104 participants (Actual)Interventional2015-07-20Completed
An Open-Label, Multicenter Study to Evaluate the Pharmacokinetics, Safety, and Efficacy of Ombitasvir (OBV), Paritaprevir (PTV), Ritonavir (RTV) With or Without Dasabuvir (DSV) and With or Without Ribavirin (RBV) in Pediatric Subjects With Genotype 1 or 4 [NCT02486406]Phase 2/Phase 364 participants (Actual)Interventional2015-10-28Completed
Drug Use Investigation of Kaletra [NCT01076972]1,184 participants (Actual)Observational2000-12-31Completed
A Phase 2b Randomized, Active-Controlled, Staged, Open-Label Trial to Investigate Safety and Efficacy of BMS-955176/GSK3532795 in Combination With Dolutegravir and Atazanavir (With or Without Ritonavir) in Treatment-Experienced HIV-1 Infected Adults [NCT02386098]Phase 286 participants (Actual)Interventional2015-07-08Terminated(stopped due to GI Intolerability)
An Open-Label, Multicenter Study to Evaluate the Efficacy and Safety of Ombitasvir/ABT-450/Ritonavir and Dasabuvir With or Without Ribavirin (RBV) in Treatment-Naïve or Treatment-Experienced Adults in Brazil With Genotype 1 Chronic Hepatitis C Virus (HCV) [NCT02442271]Phase 3222 participants (Actual)Interventional2015-04-27Completed
A Randomized, Open-Label Study to Evaluate the Safety and Efficacy of Ombitasvir/ABT-450/Ritonavir Co-administered With Ribavirin (RBV) in Adults With Genotype 4 Chronic Hepatitis C Virus (HCV) Infection and Cirrhosis (AGATE-1) [NCT02265237]Phase 3184 participants (Actual)Interventional2014-10-28Completed
Phase I Study of Pharmacokinetics and Safety of Ravidasvir in Combination With Ritonavir-boosted Danoprevir in Single and Multiple Doses in Healthy Participants [NCT03020134]Phase 118 participants (Actual)Interventional2016-07-15Completed
Part A: Drug Interaction Study Between GS-7977 and Antiretroviral Therapy (ARV) Combinations of Efavirenz, Tenofovir and Emtricitabine; Efavirenz, Zidovudine and Lamivudine; Atazanavir/Ritonavir, Tenofovir and Emtricitabine; Darunavir/Ritonavir, Tenofovir [NCT01565889]Phase 1/Phase 252 participants (Actual)Interventional2012-03-31Completed
Comparative Randomized, Single Dose, Three-way, Three-sequence, Two Treatment, Partial Replicate, Crossover, Open-label Study to Determine the Bioequivalence of Nirmatrelvir & Ritonavir From Copaxid 150 +100 mg Tablets (Eva Pharma, Egypt) Versus Paxlovid [NCT05491330]Phase 128 participants (Actual)Interventional2022-08-21Completed
Retrospective Cohort to Evaluate the Effectiveness and Safety of Xiyanping Injection Combined With Conventional Treatment for New Coronavirus Infection Pneumonia (Common Type) [NCT04275388]426 participants (Anticipated)Observational2020-05-15Not yet recruiting
THE PRIORITIZE STUDY: A Pragmatic, Randomized Study of Oral Regimens for Hepatitis C: Transforming Decision-Making for Patients, Providers, and Stakeholders [NCT02786537]Phase 41,275 participants (Actual)Interventional2016-06-30Completed
Immune Reconstitution as a Determinant of Adverse Effects to New Antiretroviral Therapy in Persons With Advanced HIV Infection [NCT00885664]Phase 460 participants (Actual)Interventional2005-10-31Completed
An Open-Label Study to Evaluate the Safety, Antiviral Activity and Pharmacokinetics of Direct-Acting Antiviral Agent (DAA) Treatment in Combination With Peginterferon Alpha-2a and Ribavirin (pegIFN/RBV) in Chronic Hepatitis C Virus (HCV) Infected Subjects [NCT01609933]Phase 232 participants (Actual)Interventional2012-12-18Completed
An Open Clinical Trial to Evaluate Danoprevir Sodium Tablets Combined With Ritonavir in the Treatment of SARS-CoV-2 Infection [NCT04345276]Phase 410 participants (Actual)Interventional2020-03-18Completed
A Single Center, Open-Label, Randomised, Parallel, Multiple Dose Comparison of the Effect of Tipranavir 500 mg and Ritonavir 100 mg or Tipranavir 750 mg and Ritonavir 200 mg Twice a Day for 11.5 Days on the Pharmacokinetic Characteristics of Zidovudine 30 [NCT02249416]Phase 160 participants (Actual)Interventional2001-11-30Completed
Evaluating Pharmacokinetic Interactions With Vaginal Ring Contraceptives and Antiretroviral Therapy (ART) [NCT01903031]Phase 284 participants (Actual)Interventional2014-12-30Completed
A Phase IIa, Open-label Trial to Evaluate the Safety, Tolerability and Efficacy of a 12 Weeks Combination Therapy of TMC647055 and TMC435 With and Without GSK23336805 With a Pharmacokinetic Enhancer With and Without Ribavirin in Chronic Genotype 1 Hepatit [NCT01724086]Phase 290 participants (Actual)Interventional2012-10-31Completed
Baricitinib Therapy in COVID-19: A Pilot Study on Safety and Clinical Impact [NCT04358614]Phase 2/Phase 312 participants (Actual)Interventional2020-03-16Completed
A Pilot Project to Operationalize the Prevention Strategy of Post Exposure Prophylaxis Following Sexual Exposure to HIV in Combination With Educational Programming and Behavioral Risk Reduction Strategies in Los Angeles County [NCT00949234]Phase 2267 participants (Actual)Interventional2010-03-31Completed
A 48 Week, Phase II, Non-Comparative, Open-label, Multi-Cohort, Multicenter Study to Evaluate the Safety, Tolerability, Pharmacokinetics and Antiviral Activity of GW433908/Ritonavir BID When Administered to HIV-1 Infected, PI-Naïve and Experienced, Pediat [NCT00089583]Phase 2110 participants (Actual)Interventional2004-07-31Completed
"Hydroxychloroquine and Lopinavir/ Ritonavir for Hospitalization and Mortality Reduction in Patients With COVID-19 and Mild Disease Symptoms: The Hope Coalition" [NCT04403100]Phase 31,968 participants (Anticipated)Interventional2020-06-03Recruiting
Antiretroviral Regime for Viral Eradication in Newborns After Intervention Failure of Mother-to-child Transmission of HIV [NCT02712801]Phase 4600 participants (Actual)Interventional2016-04-30Completed
A Randomized, Double-Blind, Phase III Study of ABT-378/Ritonavir Plus Stavudine and Lamivudine vs Nelfinavir Plus Stavudine and Lamivudine in Antiretroviral Naive HIV-Infected Subjects [NCT00004583]Phase 3660 participants Interventional1999-03-31Completed
An Open-Label Randomized Clinical Trial to Evaluate the Efficacy and Safety of Short Course Antiretroviral Therapy for Acute or Recent HIV-1 Infection in Zimbabwe and the United States [NCT00414518]16 participants (Actual)Interventional2007-01-31Completed
Efficacy of Atazanavir / Ritonavir Monotherapy as Maintenance in Patients With Viral Suppression. Randomized, Open Label Non Inferiority Trial. A Phase 3 Study. [NCT01511809]Phase 3117 participants (Actual)Interventional2010-09-30Completed
Genetic Predictors of Pharmacokinetic Variability in the Drug-drug Interaction Between Darunavir/Ritonavir and Pravastatin: the Role of SLCO1B1 Polymorphisms. [NCT00630734]Phase 432 participants (Actual)Interventional2008-02-29Completed
An Open-Label, Single-Arm Study to Evaluate the Safety and Efficacy of Ombitasvir/ABT-450/Ritonavir and Dasabuvir in Adults With Genotype 1b Chronic Hepatitis C Virus (HCV) Infection and Cirrhosis (TURQUOISE-III) [NCT02219503]Phase 360 participants (Actual)Interventional2014-09-30Completed
INFORM-SVR: A Randomized, Multi-Center Study of Interferon-Free Treatment With a Combination of a Polymerase Inhibitor (RO5024048) and a Ritonavir Boosted HCV Protease Inhibitor (RO5190591/r, DNV/r) With or Without Copegus® in Interferon Naïve HCV Genotyp [NCT01278134]Phase 2170 participants (Actual)Interventional2011-02-28Completed
Comparison of Liquid Kaletra and Low Dose Kaletra Tablets in HIV-Positive Children [NCT00762320]8 participants (Actual)Interventional2008-10-31Completed
An Open-label Study to Evaluate the Efficacy and Safety of ABT-450/Ritonavir/ABT-267 (ABT-450/r/ABT-267) Co-administered With Ribavirin (RBV) for 12 or 16 Weeks in Treatment-Naïve and Treatment-Experienced Japanese Adults With Genotype 2 Chronic Hepatitis [NCT02023112]Phase 3171 participants (Actual)Interventional2014-01-31Completed
Open-Label, Phase 2 Study to Evaluate the Safety and Efficacy of the Combination of ABT-450/Ritonavir/ABT-267 With ABT-333 and With or Without RBV in HCV Genotype 1 and ABT-450/r/ABT-267 With RBV in HCV GT4-Infected Adult Liver or Renal Transplant Recipie [NCT01782495]Phase 2129 participants (Actual)Interventional2013-02-25Completed
Randomized Clinical Trial to Evaluate the Interest of a Down-scaled Treatment Strategy Using Dual Therapy (Nucleoside Analogs) in HIV Infected Patients Already Being Treated Using Triple Therapy, Who Present With a Successful Virological Control and for W [NCT02302547]Phase 3224 participants (Actual)Interventional2014-12-31Completed
An Open-Label, Single-Arm Study to Evaluate the Safety and Efficacy of ABT-450/Ritonavir/ABT-267 (ABT-450/r/ABT-267) and ABT-333 Co-administered With Ribavirin (RBV) in Adults With Genotype 1b Chronic Hepatitis C Virus (HCV) Infection and Cirrhosis (Turqu [NCT02216422]Phase 336 participants (Actual)Interventional2014-09-30Completed
Evaluation of the Clinical Effects of Ombitasvir/Paritaprevir/Ritonavir Regimen in the Treatment of Chronic HCV Patients in CKD Versus ESRD Patients in Assiut University Hospital [NCT03341988]Phase 1100 participants (Actual)Interventional2017-11-22Completed
Evaluation of Pharmacokinetic Interaction Between Nitazoxanide and Atazanavir/Ritonavir in Healthy Volunteers [NCT05680792]17 participants (Actual)Interventional2020-09-10Completed
A Single Center, Non-randomized, Open-label Phase I Study to Investigate the Pharmacokinetics and Safety of SIM0417/Ritonavir After Single Dose Administration in Healthy Elderly Subjects [NCT05826249]Phase 120 participants (Anticipated)Interventional2023-04-07Recruiting
Phase I, Open-label Trial in Healthy Subjects to Evaluate the Drug-drug Interaction Between Ritonavir at Steady-state and TMC435350, a Viral Protease Inhibitor Against Hepatitis C Virus, After the First and the Last Dose of a Multiple Dosing Regimen [NCT01891851]Phase 112 participants (Actual)Interventional2007-10-31Completed
A Phase I Study to Assess the Pharmacokinetics and Safety of Single Dose of Ravidasvir and Danoprevir/r and Repeated Doses of Ravidasvir in Combination With Danoprevir/r in Healthy Volunteers. [NCT03288636]Phase 118 participants (Actual)Interventional2017-08-08Completed
IMPAACT 1092: Phase IV Evaluation Of The Steady State Pharmacokinetics Of Zidovudine, Lamivudine, and Lopinavir/Ritonavir in Severely Malnourished HIV-1-Infected Children [NCT01818258]Phase 452 participants (Actual)Interventional2015-10-26Completed
The Effect of Efavirenz and Ritonavir-boosted Darunavir on the Pharmacokinetics of the HMG CoA Reductase Inhibitor Pitavastatin [NCT01695954]Phase 134 participants (Actual)Interventional2012-05-31Completed
Efficacy and Safety of Ombitasvir/ Paritaprevir / Ritonavir Plus Ribavirin in Management HCV Genotype 4 and End-stage Kidney Disease With or Without Hemodialysis (An Open Label- Multicenter Prospective Study) [NCT03499639]Phase 4110 participants (Actual)Interventional2018-05-01Completed
COVID-19: A PHASE 1, OPEN-LABEL, FIXED SEQUENCE, 2-PERIOD CROSSOVER STUDY TO ESTIMATE THE EFFECT OF ITRACONAZOLE ON THE PHARMACOKINETICS OF PF-07321332/RITONAVIR IN HEALTHY PARTICIPANTS [NCT04962022]Phase 112 participants (Actual)Interventional2021-07-20Completed
A Randomized, Phase 2b Study of a Double-Dose Lopinavir/Ritonavir-Based Antiretroviral Regimen With Rifampin-Based Tuberculosis Treatment Versus a Standard-Dose Lopinavir/Ritonavir-Based Antiretroviral Regimen With Rifabutin-Based Tuberculosis Treatment W [NCT01601626]Phase 271 participants (Actual)Interventional2013-07-13Terminated(stopped due to The study was stopped early due to feasibility concerns.)
A Phase IIIb, Open -Label, Randomized Multi-center Study Comparing the Antiviral Efficacy, Safety, and Effect on Serum Lipids of Atazanavir/Ritonavir Versus Lopinavir/Ritonavir, in Combination With Two Nucleoside or Nucleotide Reverse Transcriptase Inhibi [NCT00135395]Phase 3200 participants (Anticipated)Interventional2004-05-31Completed
A Randomized Open-label Study of the Antiviral Efficacy and Safety of Atazanavir Versus Lopinavir/Ritonavir(LPV/RTV), Each in Combination With Two Nucleosides in Subjects Who Have Experienced Virologic Failure With Prior Protease Inhibitor-Containing HAAR [NCT00028301]Phase 30 participants Interventional2001-02-28Completed
A PHASE 1, OPEN-LABEL, RANDOMIZED, SINGLE-DOSE, CROSSOVER STUDY TO ESTIMATE THE RELATIVE BIOAVAILABILITY OF PF-07321332 FOLLOWING ORAL ADMINISTRATION OF 4 DIFFERENT FORMULATIONS RELATIVE TO THE COMMERCIAL TABLET FORMULATION IN HEALTHY ADULT PARTICIPANTS U [NCT05263895]Phase 112 participants (Actual)Interventional2022-03-03Completed
Comparative Efficacy of Ivermectin Versus Combination of Hydroxychloroquine Plus Darunavir/ Ritonavir for Shortening Duration of SARS-CoV2 Detection From Respiratory Secretion Among Asymptomatic or Afebrile COVID-19 Infection [NCT04435587]Phase 480 participants (Anticipated)Interventional2020-07-13Recruiting
A Pharmacokinetic Evaluation of Levonorgestrel Implant in HIV-Infected Women on Darunavir Versus Rilpivirine-based Antiretroviral Therapy [NCT03589027]Phase 260 participants (Anticipated)Interventional2018-08-07Recruiting
A Randomized, Double-blind, Placebo-Controlled Study to Evaluate the Efficacy and Safety of ABT-450/Ritonavir/ABT-267 (ABT-450/r/ABT-267) in Treatment-Naïve and Treatment-Experienced Japanese Adults With Subgenotype 1b Chronic Hepatitis C Virus (HCV) Infe [NCT02023099]Phase 3363 participants (Actual)Interventional2013-12-31Completed
An Open-Label, Proof of Concept, Randomized Trial Comparing a LPV/r-Based to an nNRTI-Based Antiretroviral Therapy Regimen for Clearance of Plasmodium Falciparum Subclinical Parasitemia in HIV-infected Adults With CD4+ Counts >200 and <500 Cells/mm^3 [NCT01632891]Phase 1/Phase 252 participants (Actual)Interventional2014-01-10Completed
COVID-19: A PHASE 1, OPEN-LABEL, 3-TREATMENT, 6-SEQUENCE, 3-PERIOD CROSSOVER STUDY TO ESTIMATE THE EFFECT OF PF-07321332/RITONAVIR AND RITONAVIR ON THE PHARMACOKINETICS OF MIDAZOLAM IN HEALTHY PARTICIPANTS. [NCT05032950]Phase 112 participants (Actual)Interventional2021-09-17Completed
A 48 Week, Phase II, Open-label, 2-cohort, Multicenter Study to Evaluate the Pharmacokinetics, Safety, Tolerability and Antiviral Activity of GW433908 and GW433908/RTV When Administered to HIV-1 Infected Protease Inhibitor (PI) Naive and PI-experienced Pe [NCT00071760]Phase 259 participants (Actual)Interventional2003-10-23Completed
The Effect of the Co-administration of Atazanavir (ATV) and Ritonavir (RTV) on the Pharmacokinetics of a Combined Oral Contraceptive Containing Ethinyl Estradiol and Norgestimate in Healthy Female Subjects [NCT00357604]Phase 122 participants Interventional2006-07-31Completed
A Multicenter, Single-blind, Randomized, Controlled Study to Evaluate the Efficacy and Safety of JT001 (VV116) Compared With Paxlovid for the Early Treatment of COVID-19 in Participants With Mild to Moderate COVID-19 [NCT05341609]Phase 3822 participants (Actual)Interventional2022-04-04Completed
Treatment of Chronic Delta Hepatitis With Lonafarnib and Ritonavir [NCT02511431]Phase 222 participants (Actual)Interventional2015-07-29Completed
To Evaluate the Efficacy and Safety of Nirmatrelvir/Ritonavir in the Treatment of the Omicron Variant of COVID-19 [NCT05813600]58 participants (Actual)Interventional2022-08-31Completed
Favipiravir, Lopinavir/Ritonavir or Combination Therapy: a Randomised, Double Blind, 2x2 Factorial Placebo-controlled Trial of Early Antiviral Therapy in COVID-19 [NCT04499677]Phase 2240 participants (Actual)Interventional2020-09-24Completed
A Randomized, Open-Label Study to Evaluate the Safety and Efficacy of the Co-Administration of Ombitasvir/ABT-450/Ritonavir (Ombitasvir/ABT-450/r) With Sofosbuvir (SOF) With or Without Ribavirin (RBV) in Subjects With Genotype 2 Chronic Hepatitis C Virus [NCT02292719]Phase 270 participants (Actual)Interventional2014-12-19Completed
A Prospective Single Arm, Open-label, International, Multicenter Study to Evaluate the Safety of Atazanavir (ATV) Capsule Boosted With Ritonavir (RTV) With an Optimized NRTI Background Therapy, in HIV Infected, Antiretroviral Naive and Experienced Pediatr [NCT01691794]Phase 4108 participants (Actual)Interventional2012-11-30Completed
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00006604 (15) [back to overview]Pharmacokinetic (PK) Parameter: Maximum Plasma Concentration (Cmax)
NCT00006604 (15) [back to overview]Pharmacokinetic (PK) Parameter: Minimum Plasma Concentration (C24)
NCT00006604 (15) [back to overview]Number of Participants Who Died
NCT00006604 (15) [back to overview]Change in CD4 Count (Cells/mm^3) From Baseline to Week 48
NCT00006604 (15) [back to overview]Change in CD4 Count (Cells/mm^3) From Baseline to Week 96
NCT00006604 (15) [back to overview]Change in CD4 Percent From Baseline to Week 20
NCT00006604 (15) [back to overview]Change in CD4 Percent From Baseline to Week 96
NCT00006604 (15) [back to overview]Change in CD4 Percent From Baseline to Week 48
NCT00006604 (15) [back to overview]Change in CD4 Count (Cells/mm^3) From Baseline to Week 20
NCT00006604 (15) [back to overview]Pharmacokinetic (PK) Parameter: Clearance (CL/F)
NCT00006604 (15) [back to overview]Number of Participants Who Experienced a Safety Endpoint of Interest Attributed to ATV
NCT00006604 (15) [back to overview]Percentage of Participants With HIV RNA <400 Copies/mL at Week 24
NCT00006604 (15) [back to overview]Percentage of Participants With HIV RNA <400 Copies/mL at Week 48
NCT00006604 (15) [back to overview]Percentage of Participants With HIV RNA <400 Copies/mL at Week 96
NCT00006604 (15) [back to overview]Pharmacokinetic (PK) Parameter: Area Under the Curve (AUC24h)
NCT00035932 (38) [back to overview]Participants Achieving Virologic Half Log Suppression (LOQ = 400 c/mL) at Week 48, (Overall and by PI Sensitivity)
NCT00035932 (38) [back to overview]Participants Achieving Treatment Response (LOQ = 50 c/mL) Without Prior Failure at Week 48
NCT00035932 (38) [back to overview]Participants Achieving Treatment Response (LOQ = 50 c/mL) Without Prior Failure at Week 96
NCT00035932 (38) [back to overview]Participants Achieving Virologic Half Log Suppression (LOQ = 400 c/mL) at Week 96
NCT00035932 (38) [back to overview]Participants Achieving Virologic Half Log Suppression (LOQ = 50 c/mL) at Week 24
NCT00035932 (38) [back to overview]Participants Achieving Virologic Half Log Suppression (LOQ = 50 c/mL) at Week 48
NCT00035932 (38) [back to overview]Adherence to Regimen Though Week 48 Based on MACS the Multicenter AIDS Cohort Study (MACS) Adherence Questionnaire
NCT00035932 (38) [back to overview]Correlation of ATV Minimum Plasma Concentration (Cmin) Inhibitory Quotient (IQ), and Number of PI Mutations at Baseline and CD4 Cell Count Change From Baseline at Week 24
NCT00035932 (38) [back to overview]Correlation of ATV Minimum Plasma Concentration (Cmin), Inhibitory Quotient (IQ), and Number of Protease Inhibitor (PI) Mutations at Baseline With HIV RNA Change From Baseline at Week 24
NCT00035932 (38) [back to overview]Deaths, Serious Adverse Events (SAEs), and Adverse Events (AEs) Through Week 48
NCT00035932 (38) [back to overview]Grade 3/4 Laboratory Abnormalities Through Week 48
NCT00035932 (38) [back to overview]Change From Baseline in CD4 Cell Count at Week 48
NCT00035932 (38) [back to overview]HIV RNA Level - Treated Subjects With Evaluable Cmins at Week 24
NCT00035932 (38) [back to overview]Lipid Mean Percent Change From Baseline at Week 24
NCT00035932 (38) [back to overview]Lipid Mean Percent Change From Baseline at Week 48
NCT00035932 (38) [back to overview]Lipid Mean Percent Change From Baseline at Week 96, Observed Values
NCT00035932 (38) [back to overview]Mean ATV, RTV and SQV Minimum Concentration (Cmin) Values
NCT00035932 (38) [back to overview]Mean Score of European Quality of Life-5 Dimensions (EQ-5D) Health Index Score at Baseline, Mid-Study (Week 24), and Final (Week 48)
NCT00035932 (38) [back to overview]Mean Score of European Quality of Life-5 Dimensions (EQ-5D) Visual Analog Scale (VAS) at Baseline, Mid-Study (Week 24), and Final (Week 48)
NCT00035932 (38) [back to overview]Participants Achieving Virologic Half Log Suppression (Limit of Quantification [LOQ] = 400 c/mL) at Week 24 (Overall and by Protease Inhibitor [PI] Sensitivity)
NCT00035932 (38) [back to overview]Fridericia-corrected QT (QTcF) Interval and Change From Baseline by Analysis Time Point
NCT00035932 (38) [back to overview]Change From Baseline in CD4 Cell Count at Week 24
NCT00035932 (38) [back to overview]PR Interval and Change From Baseline by Analysis Time Point
NCT00035932 (38) [back to overview]Change From Baseline in CD4 Cell Count at Week 96
NCT00035932 (38) [back to overview]Fasting Glucose Mean Change From Baseline at Week 24
NCT00035932 (38) [back to overview]Most Common AEs and AEs of Interest Through Week 48
NCT00035932 (38) [back to overview]Mean Change From Baseline in HIV RNA at Week 48
NCT00035932 (38) [back to overview]Fasting Glucose Mean Change From Baseline at Week 48
NCT00035932 (38) [back to overview]HIV IC50 at Week 24
NCT00035932 (38) [back to overview]Inhibitory Quotient at Week 24
NCT00035932 (38) [back to overview]Mean Change From Baseline in HIV Ribonucleic Acid (RNA) at Week 24
NCT00035932 (38) [back to overview]Mean Change From Baseline in HIV RNA at Week 2
NCT00035932 (38) [back to overview]Participants Achieving Virologic Half Log Suppression (LOQ = 50 c/mL) at Week 96
NCT00035932 (38) [back to overview]Mean Change From Baseline in HIV RNA at Week 96
NCT00035932 (38) [back to overview]Participants Achieving Treatment Response (LOQ = 400 c/mL) Without Prior Failure at Week 24
NCT00035932 (38) [back to overview]Participants Achieving Treatment Response (LOQ = 400 c/mL) Without Prior Failure at Week 48
NCT00035932 (38) [back to overview]Participants Achieving Treatment Response (LOQ = 400 c/mL) Without Prior Failure at Week 96
NCT00035932 (38) [back to overview]Participants Achieving Treatment Response (LOQ = 50 c/mL) Without Prior Failure at Week 24
NCT00040664 (16) [back to overview]Geometric Mean of Steady State Plasma Amprenavir (APV) Parameter: AUC(0-tau)
NCT00040664 (16) [back to overview]Least Squares Mean of Plasma APV Parameter: Ctau
NCT00040664 (16) [back to overview]Percentage of Participants With HIV-1 RNA <400 Copies Per mL at Weeks 12, 48, 96, and 168 (Time to Loss of Virologic Response [TLOVR] Analysis)
NCT00040664 (16) [back to overview]Number of Participants With Any Drug-related Grade 2 to 4 Adverse Event
NCT00040664 (16) [back to overview]Number of Participants Who Discontinued Treatment Due to Adverse Events
NCT00040664 (16) [back to overview]Median Change From Baseline HIV-1 RNA Values at Weeks 12, 48, 96, and 168 Visits
NCT00040664 (16) [back to overview]Number of Participants With APV Resistance Associated HIV-1 RNA Genotypic Mutations and Phenotypic Resistance at Time of Virologic Failure Not Present at Baseline
NCT00040664 (16) [back to overview]Median Steady State Plasma APV Tmax
NCT00040664 (16) [back to overview]Least Squares Mean of Plasma APV Parameter: Cmax
NCT00040664 (16) [back to overview]Least Squares Mean of Plasma APV Parameter: AUC0-tau
NCT00040664 (16) [back to overview]Median Change From Baseline in CD4+ Values at Week 12, 48, 96, and 168 Visits
NCT00040664 (16) [back to overview]Number of Participants With Grade 3 or 4 Treatment-emergent Laboratory Abnormalities
NCT00040664 (16) [back to overview]Geometric Mean of Steady State Plasma APV Parameter: t1/2
NCT00040664 (16) [back to overview]Geometric Mean of Steady State Plasma APV Parameter: Cmax
NCT00040664 (16) [back to overview]Geometric Mean of Steady State Plasma APV Parameter: CL/F
NCT00040664 (16) [back to overview]Geometric Mean of Steady State Plasma APV Parameter: CL/F
NCT00042289 (26) [back to overview]PK Parameter: Trough Concentration (C12) With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Trough Concentration (C24) With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Trough Concentration (C24) With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]Number of Women Who Met PK Target of Area Under the Curve (AUC) for ARVs
NCT00042289 (26) [back to overview]PK Parameter: Maximum Concentration (Cmax) in ng/mL With Median (95% CI) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Cord/Maternal Blood Concentration Ratio With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]Number of Women Who Met PK Target of Area Under the Curve (AUC) for ARVs
NCT00042289 (26) [back to overview]Area Under the Curve From 0 to 24 Hours (AUC24) of ARVs for Contraceptive Arms
NCT00042289 (26) [back to overview]Area Under the Curve From 0 to 12 Hours (AUC12) of ARVs for Contraceptive Arms
NCT00042289 (26) [back to overview]Plasma Concentration for Contraceptives
NCT00042289 (26) [back to overview]PK Parameter: Trough Concentration (C24) With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Cord/Maternal Blood Concentration Ratio With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]Pharmacokinetic (PK) Parameter: Infant Plasma Washout Half-life (T1/2) of ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Maximum Concentration (Cmax) in mg/L With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Maximum Concentration (Cmax) in mg/L With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Maximum Concentration (Cmax) in mg/L With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Maximum Concentration (Cmax) in ng/mL With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Trough Concentration (C12) With Geometric Mean (95% CI) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Trough Concentration (C12) With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Area Under the Curve From 0 to 24 Hours (AUC24) With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Area Under the Curve From 0 to 24 Hours (AUC24) With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Area Under the Curve From 0 to 24 Hours (AUC24) With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Area Under the Curve From 0 to 12 Hours (AUC12) With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Area Under the Curve From 0 to 12 Hours (AUC12) With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Area Under the Curve From 0 to 12 Hours (AUC12) With Geometric Mean (95% CI) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]Pharmacokinetic (PK) Parameter: Infant Plasma Washout Concentration of ARVs and TB Drugs
NCT00054717 (90) [back to overview]Mean Change From Baseline to Week 40 in CD4+ Cell Count
NCT00054717 (90) [back to overview]Mean Change From Baseline to Week 48 in CD4+ Cell Count
NCT00054717 (90) [back to overview]Mean Change From Baseline to Week 56 in CD4+ Cell Count
NCT00054717 (90) [back to overview]Mean Change From Baseline to Week 64 in CD4+ Cell Count
NCT00054717 (90) [back to overview]Mean Change From Baseline to Week 72 in CD4+ Cell Count
NCT00054717 (90) [back to overview]Mean Change From Baseline to Week 8 in CD4+ Cell Count
NCT00054717 (90) [back to overview]Mean Change From Baseline to Week 80 in CD4+ Cell Count
NCT00054717 (90) [back to overview]Mean Change From Baseline to Week 88 in CD4+ Cell Count
NCT00054717 (90) [back to overview]Mean Change From Baseline to Week 96 in CD4+ Cell Count
NCT00054717 (90) [back to overview]Median Change From Baseline in Viral Load to Week 2
NCT00054717 (90) [back to overview]Median Change From Baseline in Viral Load to Week 24
NCT00054717 (90) [back to overview]Median Change From Baseline in Viral Load to Week 32
NCT00054717 (90) [back to overview]Median Change From Baseline in Viral Load to Week 4
NCT00054717 (90) [back to overview]Median Change From Baseline in Viral Load to Week 40
NCT00054717 (90) [back to overview]Median Change From Baseline in Viral Load to Week 48
NCT00054717 (90) [back to overview]Median Change From Baseline in Viral Load to Week 56
NCT00054717 (90) [back to overview]Median Change From Baseline in Viral Load to Week 64
NCT00054717 (90) [back to overview]Median Change From Baseline in Viral Load to Week 72
NCT00054717 (90) [back to overview]Median Change From Baseline in Viral Load to Week 80
NCT00054717 (90) [back to overview]Median Change From Baseline in Viral Load to Week 88
NCT00054717 (90) [back to overview]Median Change From Baseline in Viral Load to Week 96
NCT00054717 (90) [back to overview]Time to Confirmed Virologic Failure Through 48 Weeks of Treatment
NCT00054717 (90) [back to overview]Time to Confirmed Virologic Failure Through 96 Weeks of Treatment
NCT00054717 (90) [back to overview]Time to New CDC Class C Progression Event or Death.
NCT00054717 (90) [back to overview]Time to Treatment Failure Through 48 Weeks of Treatment
NCT00054717 (90) [back to overview]Time to Treatment Failure Through 96 Weeks of Treatment
NCT00054717 (90) [back to overview]Treatment Response at Week 16
NCT00054717 (90) [back to overview]Treatment Response at Week 2
NCT00054717 (90) [back to overview]Treatment Response at Week 24
NCT00054717 (90) [back to overview]Treatment Response at Week 32
NCT00054717 (90) [back to overview]Treatment Response at Week 4
NCT00054717 (90) [back to overview]Treatment Response at Week 40
NCT00054717 (90) [back to overview]Treatment Response at Week 48
NCT00054717 (90) [back to overview]Treatment Response at Week 48
NCT00054717 (90) [back to overview]Treatment Response at Week 56
NCT00054717 (90) [back to overview]Treatment Response at Week 64
NCT00054717 (90) [back to overview]Treatment Response at Week 72
NCT00054717 (90) [back to overview]Treatment Response at Week 8
NCT00054717 (90) [back to overview]Treatment Response at Week 80
NCT00054717 (90) [back to overview]Treatment Response at Week 88
NCT00054717 (90) [back to overview]Treatment Response at Week 96
NCT00054717 (90) [back to overview]Virologic Response (Viral Load >= 1 Log Drop) at Viral Load Nadir, LOCF
NCT00054717 (90) [back to overview]Virologic Response (Viral Load >= 1 Log Drop) at Week 16
NCT00054717 (90) [back to overview]Virologic Response (Viral Load >= 1 Log Drop) at Week 2
NCT00054717 (90) [back to overview]Virologic Response (Viral Load >= 1 Log Drop) at Week 24
NCT00054717 (90) [back to overview]Virologic Response (Viral Load >= 1 Log Drop) at Week 32
NCT00054717 (90) [back to overview]Virologic Response (Viral Load >= 1 Log Drop) at Week 4
NCT00054717 (90) [back to overview]Virologic Response (Viral Load >= 1 Log Drop) at Week 40
NCT00054717 (90) [back to overview]Virologic Response (Viral Load >= 1 Log Drop) at Week 48
NCT00054717 (90) [back to overview]Virologic Response (Viral Load >= 1 Log Drop) at Week 56
NCT00054717 (90) [back to overview]Virologic Response (Viral Load >= 1 Log Drop) at Week 64
NCT00054717 (90) [back to overview]Virologic Response (Viral Load >= 1 Log Drop) at Week 8
NCT00054717 (90) [back to overview]Virologic Response (VL < 400 Copies/ml) at Week 16
NCT00054717 (90) [back to overview]Virologic Response (VL < 400 Copies/ml) at Week 2
NCT00054717 (90) [back to overview]Virologic Response (VL < 400 Copies/ml) at Week 24
NCT00054717 (90) [back to overview]Virologic Response (VL < 400 Copies/ml) at Week 32
NCT00054717 (90) [back to overview]Virologic Response (VL < 400 Copies/ml) at Week 4
NCT00054717 (90) [back to overview]Virologic Response (VL < 400 Copies/ml) at Week 40
NCT00054717 (90) [back to overview]Virologic Response (VL < 400 Copies/ml) at Week 48
NCT00054717 (90) [back to overview]Virologic Response (VL < 400 Copies/ml) at Week 56
NCT00054717 (90) [back to overview]Virologic Response (VL < 400 Copies/ml) at Week 64
NCT00054717 (90) [back to overview]Virologic Response (VL < 400 Copies/ml) at Week 72
NCT00054717 (90) [back to overview]Virologic Response (VL < 400 Copies/ml) at Week 8
NCT00054717 (90) [back to overview]Virologic Response (VL < 400 Copies/ml) at Week 80
NCT00054717 (90) [back to overview]Virologic Response (VL < 400 Copies/ml) at Week 88
NCT00054717 (90) [back to overview]Virologic Response (VL < 400 Copies/ml) at Week 96
NCT00054717 (90) [back to overview]Virologic Response (VL < 50 Copies/ml) at Viral Load Nadir, LOCF
NCT00054717 (90) [back to overview]Virologic Response (VL < 50 Copies/ml) at Week 16
NCT00054717 (90) [back to overview]Virologic Response (VL < 50 Copies/ml) at Week 2
NCT00054717 (90) [back to overview]Virologic Response (VL < 50 Copies/ml) at Week 24
NCT00054717 (90) [back to overview]Virologic Response (VL < 50 Copies/ml) at Week 32
NCT00054717 (90) [back to overview]Virologic Response (VL < 50 Copies/ml) at Week 4
NCT00054717 (90) [back to overview]Virologic Response (VL < 50 Copies/ml) at Week 40
NCT00054717 (90) [back to overview]Virologic Response (VL < 50 Copies/ml) at Week 48
NCT00054717 (90) [back to overview]Virologic Response (VL < 50 Copies/ml) at Week 56
NCT00054717 (90) [back to overview]Virologic Response (VL < 50 Copies/ml) at Week 64
NCT00054717 (90) [back to overview]Virologic Response (VL < 50 Copies/ml) at Week 72
NCT00054717 (90) [back to overview]Virologic Response (VL < 50 Copies/ml) at Week 8
NCT00054717 (90) [back to overview]Virologic Response (VL < 50 Copies/ml) at Week 80
NCT00054717 (90) [back to overview]Virologic Response (VL < 50 Copies/ml) at Week 88
NCT00054717 (90) [back to overview]Virologic Response (VL < 50 Copies/ml) at Week 96
NCT00054717 (90) [back to overview]Percentage of Patients With Division of Acquired Immunodeficiency Syndrome (DAIDS) Grade 3 or 4 Laboratory Abnormalities
NCT00054717 (90) [back to overview]Virologic Response (VL < 400 Copies/ml) at Viral Load Nadir, LOCF
NCT00054717 (90) [back to overview]Median Change From Baseline in Viral Load to Week 8
NCT00054717 (90) [back to overview]Median Change From Baseline in Viral Load to Week 16
NCT00054717 (90) [back to overview]Mean Change From Baseline to Week 16 in CD4+ Cell Count
NCT00054717 (90) [back to overview]Mean Change From Baseline to Week 2 in CD4+ Cell Count
NCT00054717 (90) [back to overview]Mean Change From Baseline to Week 24 in CD4+ Cell Count
NCT00054717 (90) [back to overview]Mean Change From Baseline to Week 32 in CD4+ Cell Count
NCT00054717 (90) [back to overview]Mean Change From Baseline to Week 4 in CD4+ Cell Count
NCT00071760 (51) [back to overview]Number of Confirmed Virologic Failure Participants (Par.) With Treatment-emergent Reductions in Drug Susceptibility (DS)
NCT00071760 (51) [back to overview]Number of Participants With Plasma HIV-1 Ribonucleic Acid (RNA) <400 Copies Per Milliliter at Baseline and Weeks 4, 12, 24, 36, and 48 (MSD=F)
NCT00071760 (51) [back to overview]Number of Participants With Hematology Toxicities
NCT00071760 (51) [back to overview]Number of Participants With Clinical Chemistry Toxicities
NCT00071760 (51) [back to overview]Absolute Values of Cholesterol, Glucose, High Density Lipoprotein (HDL) Cholesterol, Low Density Lipoprotein (LDL) and Triglyceride (TG) at Baseline (Day 1), Weeks 4, 12, 24, 36, and 48
NCT00071760 (51) [back to overview]Number of Participants With at Least a 1.0 log10 HIV-1 RNA Decrease From Baseline at Weeks 4, 12, 24, 36, and 48 (MSD=F Analysis)
NCT00071760 (51) [back to overview]Number of Participants Reporting Perfect Adherence at Weeks 2, 12, 24, and 48 as Assessed by the Study Coordinator Using the Adherence Questionnaire
NCT00071760 (51) [back to overview]Number of Participants With Treatment-Emergent Changes in HIV-1 Phenotypic Drug Susceptibility
NCT00071760 (51) [back to overview]Number of Confirmed Virologic Failure Participants (Par.) With Treatment-emergent HIV Genotypic Resistance in Reverse Transcriptase and Protease
NCT00071760 (51) [back to overview]Median Plasma HIV-1 RNA (log10 Copies/mL) at Baseline and Weeks 4, 12, 24, 36, and 48 (Observed Analysis)
NCT00071760 (51) [back to overview]Median Percent Cluster of Differentiation Antigen 4 (CD4+) Cell Count at Baseline and at Weeks 4, 12, 24, 36, and 48
NCT00071760 (51) [back to overview]Median Percent Change From Baseline in CD4+ Cell Count at Weeks 4, 12, 24, 36, and 48
NCT00071760 (51) [back to overview]Number of Participants With the Indicated Treatment-emergent (TE) Grade 3/4 Laboratory Abnormalities
NCT00071760 (51) [back to overview]Number of Participants With the Indicated Virological Outcome at Week 48
NCT00071760 (51) [back to overview]Number of Participants With the Indicated Response Scores for the Parent/Guardian (P/G) Perception of FPV Oral Suspension Questionnaire: Items 1 to 4
NCT00071760 (51) [back to overview]Number of Participants With Treatment-Emergent Mutations at Virologic Failure Timepoint
NCT00071760 (51) [back to overview]Plasma Amprenavir (APV) AUC (0-tau[τ])
NCT00071760 (51) [back to overview]Plasma Amprenavir (APV) AUC (0-tau[τ])
NCT00071760 (51) [back to overview]Median Change From Baseline in Plasma HIV-1 RNA (log10 Copies/mL) at Weeks 4, 12, 24, 36, and 48 (Observed Analysis)
NCT00071760 (51) [back to overview]Change From Baseline in Plasma HIV-1 RNA Levels at Each Study Visit
NCT00071760 (51) [back to overview]Change From Baseline in Plasma HIV-1 RNA Levels at Each Study Visit
NCT00071760 (51) [back to overview]Number of Participants With the Indicated Response Scores for the Parent/Guardian Perception of the Child's Assessment of FPV Oral Suspension Questionnaire: Items (I) 5 to 10
NCT00071760 (51) [back to overview]Absolute Values of Serum Lipase at Baseline (Day 1), Weeks 4, 12, 24, and 48
NCT00071760 (51) [back to overview]Absolute Values of Serum Lipase at Baseline (Day 1), Weeks 4, 12, 24, and 48
NCT00071760 (51) [back to overview]Absolute Values of Cluster of Differentiation 4 (CD4+) Cell Counts by Study Visit
NCT00071760 (51) [back to overview]Absolute Values of Cluster of Differentiation 4 (CD4+) Cell Counts by Study Visit
NCT00071760 (51) [back to overview]Plasma APV CL/F Following Dosing Expressed in mL/Min
NCT00071760 (51) [back to overview]Plasma APV CL/F Following Dosing Expressed in mL/Min
NCT00071760 (51) [back to overview]Plasma APV CL/F Following Dosing Expressed in mL/Min/kg
NCT00071760 (51) [back to overview]Plasma APV CL/F Following Dosing Expressed in mL/Min/kg
NCT00071760 (51) [back to overview]Number of Participants With Treatment Limiting Toxicities
NCT00071760 (51) [back to overview]Plasma APV Cmax
NCT00071760 (51) [back to overview]Absolute Values of Cholesterol, Glucose, High Density Lipoprotein (HDL) Cholesterol, Low Density Lipoprotein (LDL) and Triglyceride (TG) at Baseline (Day 1), Weeks 4, 12, 24, 36, and 48
NCT00071760 (51) [back to overview]Number of Participants Who Permanently Discontinued the Treatment Due to Adverse Event
NCT00071760 (51) [back to overview]Plasma APV Cmax
NCT00071760 (51) [back to overview]Number of Participants With Treatment Emergent Adverse Events (AEs)
NCT00071760 (51) [back to overview]Absolute Values of Alanine Amino Transferase (ALT) and Aspartate Amino Transferase (AST) at Baseline (Day 1), Weeks 4, 12, 24, 36, and 48
NCT00071760 (51) [back to overview]Plasma Unbound APV Percent Protein Binding (%Cτ)
NCT00071760 (51) [back to overview]Plasma RTV CL/F Expressed in mL/Min
NCT00071760 (51) [back to overview]Plasma Unbound APV Percent Protein Binding (%Cτ)
NCT00071760 (51) [back to overview]Plasma Unbound APV Cτ
NCT00071760 (51) [back to overview]Plasma Unbound APV Cτ
NCT00071760 (51) [back to overview]Plasma RTV Cτ
NCT00071760 (51) [back to overview]Plasma RTV Cmax
NCT00071760 (51) [back to overview]Plasma RTV CL/F Expressed in mL/Min/kg
NCT00071760 (51) [back to overview]Number of Participants With Plasma Human Immuno Virus Type 1 (HIV-1) Ribonucleic Acid (RNA) Levels < 400 Copies/Milliliter at Each Study Visit
NCT00071760 (51) [back to overview]Plasma Ritonavir (RTV) AUC (0-τ)
NCT00071760 (51) [back to overview]Plasma APV Cτ
NCT00071760 (51) [back to overview]Number of Participants With the Indicated Treatment-emergent (TE) Grade 3/4 Adverse Events (AE)
NCT00071760 (51) [back to overview]Number of Participants Who Permanently Discontinued the Treatment Due to an AE
NCT00071760 (51) [back to overview]Plasma APV Cτ
NCT00074581 (2) [back to overview]All Partner HIV Infection Rates in Early-ART and Delayed-ART Arms
NCT00074581 (2) [back to overview]Linked Partner HIV Infection Rates in Early-ART and Delayed-ART Arms
NCT00084149 (7) [back to overview]Levels of Proviral DNA in Peripheral Blood Mononuclear Cells (PBMC) (log10)
NCT00084149 (7) [back to overview]Adverse Events Related to Study Medication
NCT00084149 (7) [back to overview]CD4 T Cell Levels
NCT00084149 (7) [back to overview]HIV-1 Viral Load Levels
NCT00084149 (7) [back to overview]Number of Patients With Viral Load Less Than 50 Copies/ml
NCT00084149 (7) [back to overview]Proviral DNA (log10)
NCT00084149 (7) [back to overview]Proviral DNA Levels (log10)
NCT00089505 (9) [back to overview]Number of Participants Who Experienced Virologic Failure or Died.
NCT00089505 (9) [back to overview]Number of Participants Who Experienced Treatment-related Toxicity That Led to Discontinuation of Randomized Regimen.
NCT00089505 (9) [back to overview]Number of Participants Who Experienced HIV-related Disease Progression or Death
NCT00089505 (9) [back to overview]Percent of Participants Who Reported to Never Missed Any of the Study Drug Regimen in the Past Month
NCT00089505 (9) [back to overview]Percent of Participants Who Experienced Virologic Failure or Died
NCT00089505 (9) [back to overview]CD4 Count Change From Randomization
NCT00089505 (9) [back to overview]Number of Participants Who Received NVP-containing Regimens at Randomization and Experienced NVP-associated Rash or Grade 2+ Liver Lab Abnormality
NCT00089505 (9) [back to overview]Time From Randomization to Virologic Failure or Death for Participants Without SD NVP Exposure Prior to Study Entry
NCT00089505 (9) [back to overview]Time From Randomization to Virologic Failure or Death for Participants Who Had SD NVP Exposure Prior to Study Entry
NCT00089583 (33) [back to overview]Plasma APV t1/2
NCT00089583 (33) [back to overview]Number of Participants (Par.) With Plasma HIV-1 Ribonucleic Acid (RNA) <400 Copies Per Milliliter at Baseline and Weeks 2,12, 24, and 48 (MSD=F)
NCT00089583 (33) [back to overview]Change From Baseline in the Percentage of Total Lymphocytes (TLs) That Are CD4+ Cells at Weeks 2, 12, 24, and 48
NCT00089583 (33) [back to overview]Change From Baseline in Triglycerides, Total Cholesterol, Low-density Lipoprotein (LDL) Cholesterol, High-density Lipoprotein (HDL) Cholesterol, and Serum Glucose at Week 48
NCT00089583 (33) [back to overview]Cluster of Differentiation Antigen 4 (CD4+) Cell Count at Baseline and at Weeks 2, 12, 24, and 48
NCT00089583 (33) [back to overview]Median Change From Plasma HIV-1 RNA (log10 Copies/mL) at Weeks 2, 12, 24, and 48 (Observed Analysis)
NCT00089583 (33) [back to overview]Number of Confirmed Virologic Failure Participants (Par.) With Treatment-emergent Reductions in Drug Susceptibility (DS)
NCT00089583 (33) [back to overview]Number of Participants With Treatment-emergent (TE) Grade 3/4 Clinical Chemistry Laboratory Abnormalities
NCT00089583 (33) [back to overview]Number of Confirmed Virologic Failure Participants (Par.) With Treatment-emergent HIV Genotypic Resistance in Reverse Transcriptase and Protease
NCT00089583 (33) [back to overview]Median Plasma HIV-1 RNA (log10 Copies/mL) at Baseline and Weeks 2, 12, 24, and 48 (Observed Analysis)
NCT00089583 (33) [back to overview]Change From Baseline in Aspartate Aminotransferase (AST) and Alanine Aminotransferase (ALT) at Week 48
NCT00089583 (33) [back to overview]Change From Baseline in CD4+ Cell Count at Weeks 2, 12, 24, and 48
NCT00089583 (33) [back to overview]Number of Participants Who Permanently Discontinued the Treatment Due to Any Adverse Event (AE)
NCT00089583 (33) [back to overview]Change From Baseline in Serum Lipase at Week 48
NCT00089583 (33) [back to overview]Plasma RTV CL/F Following Dosing Expressed in mg/kg
NCT00089583 (33) [back to overview]Plasma RTV Tmax
NCT00089583 (33) [back to overview]Number of Confirmed Virologic Failure Participants (Par.) Since the Week 48 Analysis With Treatment-emergent HIV Genotypic Resistance in Reverse Transcriptase and Protease
NCT00089583 (33) [back to overview]Plasma RTV t1/2
NCT00089583 (33) [back to overview]Plasma APV Tmax
NCT00089583 (33) [back to overview]Plasma RTV Cτ
NCT00089583 (33) [back to overview]Plasma RTV Cmax
NCT00089583 (33) [back to overview]Plasma RTV CL/F Following Dosing Expressed in mg
NCT00089583 (33) [back to overview]Plasma Ritonavir (RTV) AUC (0-τ)
NCT00089583 (33) [back to overview]Plasma APV CL/F Following Dosing Expressed in mg
NCT00089583 (33) [back to overview]Number of Confirmed Virologic Failure Participants (Par.) Since the Week 48 Analysis With Treatment-emergent Reductions in Drug Susceptibility (DS)
NCT00089583 (33) [back to overview]Plasma APV Cτ
NCT00089583 (33) [back to overview]Plasma APV Cmax
NCT00089583 (33) [back to overview]Plasma APV CL/F Following Dosing Expressed in mg/kg
NCT00089583 (33) [back to overview]Plasma Amprenavir (APV) AUC (0-tau[τ])
NCT00089583 (33) [back to overview]Percentage of Total Lymphocytes (TLs) That Are CD4+ Cells at Baseline and Weeks 2, 12, 24, and 48
NCT00089583 (33) [back to overview]Number of Participants With at Least a 1.0 log10 HIV-1 RNA Decrease From Baseline at Weeks 2, 12, 24, and 48 (Observed Analysis)
NCT00089583 (33) [back to overview]Number of Participants Reporting Perfect Adherence Over the 3 Days Prior to the Study Visits at Weeks 2, 12, 24, and 48 as Assessed by Study Coordinator Using the Pediatric AIDS Clinical Trials Group (PACTG) Adherence Questionnaire
NCT00089583 (33) [back to overview]Number of Participants (Par.) With Virological Outcome (Plasma HIV-1 Ribonucleic Acid [RNA] <400 Copies/mL) at Week 48
NCT00090779 (9) [back to overview]Change in CD4 Counts Cells/mm^3 From Week 36 for IT Arm and From Week 0 for DT Arm
NCT00090779 (9) [back to overview]Time From Study Entry in DT Arm Participants or From Week 36 in IT Arm Participants to Meeting the Clinical, Virologic, or Immunologic Criteria for Treatment Initiation or Re-initiation
NCT00090779 (9) [back to overview]Time to Meeting the Clinical, Virologic, or Immunologic Criteria for Treatment Initiation or Re-initiation
NCT00090779 (9) [back to overview]Time to Treatment Initiation or Death
NCT00090779 (9) [back to overview]Number of Participants Experiencing Either a CDC Category B or C Diagnosis, CD4<200 Cells/mm^3 or CD4 Percent <14%.
NCT00090779 (9) [back to overview]Number of Participants in IT Arm Off Treatment Before 36 Weeks
NCT00090779 (9) [back to overview]Ranked Log10 HIV-1 RNA Viral Load (log10 Copies/mL) Averaged at 72 and 76 Weeks for the IT Arm and DT Arm
NCT00090779 (9) [back to overview]Ranked log10 HIV-1 RNA Viral Load (log10 Copies/mL) Averaged at Weeks 72 and 76 for the IT Arm and Ranked log10 HIV-1 RNA Viral Load (log10 Copies/mL) Averaged at Weeks 36 and 40 for the DT Arm
NCT00090779 (9) [back to overview]Number of Participants Meeting Clinical, Virologic, or Immunologic Criteria for Treatment Initiation or Re-initiation
NCT00099632 (5) [back to overview]Severe (Grade 3) and Higher Adverse Events and Any Grade Adverse Event That Leads to a Treatment Change From First Day of Study Treatment to Week 12
NCT00099632 (5) [back to overview]Number of Participants With New PI-resistant Variants as Detected by Standard Composite (Bulk) Genotyping.
NCT00099632 (5) [back to overview]Number of Participants With New Circulating NRTI-resistant Variants Detected by Standard Composite (Bulk) Genotyping.
NCT00099632 (5) [back to overview]Number of Participants Who Discontinued Study Treatment Prematurely
NCT00099632 (5) [back to overview]Number of Participants With New Circulating Nonnucleoside Reverse Transcriptase Inhibitor (NNRTI)-Resistant Variants as Detected by Standard Composite (Bulk) Genotyping
NCT00102960 (13) [back to overview]Time to First Hospitalization
NCT00102960 (13) [back to overview]Total Occurrence of Grade 3 or 4 Clinical Events
NCT00102960 (13) [back to overview]Total Occurrence of Grade 3 or 4 Laboratory Events
NCT00102960 (13) [back to overview]Number of Children Experiencing Severe CDC Stage B or Stage C Disease or Death (Cumulative After 3.5 Years)
NCT00102960 (13) [back to overview]Duration of Hospitalisation
NCT00102960 (13) [back to overview]Hospitalization Rates
NCT00102960 (13) [back to overview]Number of Participants Who Experienced Clinical Failure (Defined as Development of Severe CDC Stage B or Stage C Disease.) on Therapy.
NCT00102960 (13) [back to overview]Number of Participants Who Experienced Immunological Failure Defined as Failure of CD4% to Reach 20% or CD4% Falls Below 20% on Two Occasions, Within 4 Weeks, at Any Time After the First 24 Weeks of Therapy (Initial Therapy or Restart)
NCT00102960 (13) [back to overview]Number of Participants Who Experienced Regimen-limiting ART Drug Toxicity
NCT00102960 (13) [back to overview]Number of Participants Who Experienced Virological Failure Defined as Confirmed HIV-1 RNA Value of at Least 10,000 Copies Per/ml Recorded on Two Consecutive Separate Occasions After 24 Weeks of Treatment (Initial Therapy or Restart)
NCT00102960 (13) [back to overview]Time From Randomization to Starting or Needing to Start Continuous Therapy
NCT00102960 (13) [back to overview]Time to Death Alone or Death Plus Life Threatening Stage C Events or HIV Events Associated With Permanent End-organ Damage.
NCT00102960 (13) [back to overview]Time to Failure of First Line Therapy or Death
NCT00105079 (6) [back to overview]Number of Participants Assessed for Adverse Events (AEs)
NCT00105079 (6) [back to overview]Number of Patients Who Discontinued Treatment Due to Abnormal Laboratory Parameters
NCT00105079 (6) [back to overview]Change From Baseline in Cluster Differentiation Antigen 4 Positive (CD4+) Lymphocyte Count
NCT00105079 (6) [back to overview]Change From Baseline in HIV-1 RNA Viral Load
NCT00105079 (6) [back to overview]Number of Patients With HIV-1 RNA Viral Load <50 and <400 Copies/mL
NCT00105079 (6) [back to overview]Number of Patients With Human Immunodeficiency Virus Type 1 (HIV-1) Ribonucleic Acid (RNA) Viral Load <50 Copies/mL
NCT00109590 (12) [back to overview]Pre-dose Concentration Pharmacokinetic Outcome for LPV/r (Cpredose ug/mL).
NCT00109590 (12) [back to overview]Median HIV-1 Viral Load at 24 Weeks Postpartum in Women
NCT00109590 (12) [back to overview]Number of Women With Grade >=3 Events After Start of Study Treatment
NCT00109590 (12) [back to overview]Four (4) Hour Concentration Pharmacokinetic Outcome for LPV/r (C4hour ug/mL).
NCT00109590 (12) [back to overview]Resistance Mutations in HIV Infected Infants
NCT00109590 (12) [back to overview]The Proportion of Women in Each Randomized Arm Who Have One or More New NVP Resistance Mutations as Identified by Consensus Sequencing or Oligonucleotide Ligation Assay (OLA) in Plasma
NCT00109590 (12) [back to overview]The Proportion of Women in Each Randomized Arm Who Have One or More New NVP Resistance Mutations for the Subgroup of Women With Plasma HIV RNA >= 500 Copies/ml At Entry
NCT00109590 (12) [back to overview]The Proportion of Women Who Develop One or More New NVP Resistance Mutations as Identified by Consensus Sequencing or Oligonucleotide Ligation Assay in Plasma (Sampling Was Done at Days 10,21,30, and Weeks 5,6, and 8 Postpartum).
NCT00109590 (12) [back to overview]Proportion of Women With New NVP Resistance Mutation Within 8 Weeks Postpartum Who Had a NVP Resistance Mutation Detected at 72 Weeks Postpartum.
NCT00109590 (12) [back to overview]The Proportion of Women With Any New ZDV, ddI, or LPV/r Resistance Mutations.
NCT00109590 (12) [back to overview]Maximum Concentration Pharmacokinetic Outcome for LPV/r (Cmax ug/mL) .
NCT00109590 (12) [back to overview]Area Under the Curve Pharmacokinetic Outcome for LPV/r. (AUC ug*hr/mL)
NCT00118898 (22) [back to overview]Time From Treatment Dispensation to Regimen Failure (First Occurrence of Virologic Failure or Treatment Modification)
NCT00118898 (22) [back to overview]Time From Treatment Dispensation to Treatment Modification
NCT00118898 (22) [back to overview]Number of Participants With Virologic Failure
NCT00118898 (22) [back to overview]Number of Participants With Virologic Failure and Emergence of Major Resistance
NCT00118898 (22) [back to overview]Change in CD4 Count (Cells/mm3) From Baseline
NCT00118898 (22) [back to overview]Change in Fasting High-density Lipoprotein (HDL) Cholesterol Level From Baseline
NCT00118898 (22) [back to overview]Change in Fasting Non-high Density Lipoprotein (Non-HDL) Cholesterol Level From Baseline
NCT00118898 (22) [back to overview]Change in Fasting Total Cholesterol Level From Baseline
NCT00118898 (22) [back to overview]Change in Fasting Triglyceride Level From Baseline
NCT00118898 (22) [back to overview]Time From Treatment Dispensation to a Grade 3/4 Safety Event
NCT00118898 (22) [back to overview]Cumulative Probability of Not Experiencing a Grade 3/4 Safety Event
NCT00118898 (22) [back to overview]Cumulative Probability of Not Experiencing Regimen Failure
NCT00118898 (22) [back to overview]Cumulative Probability of Not Experiencing Treatment Modification
NCT00118898 (22) [back to overview]Cumulative Probability of Not Experiencing Virologic Failure
NCT00118898 (22) [back to overview]Number of Participants Experiencing Certain Targeted Clinical Events, Including Death, AIDS-defining Illness, and HIV-1 Related Events.
NCT00118898 (22) [back to overview]Number of Participants With HIV-1 RNA Levels Less Than 200 Copies/mL
NCT00118898 (22) [back to overview]The Number of Participants With HIV-1 RNA Levels Less Than 50 Copies/mL
NCT00118898 (22) [back to overview]Amount of Study Follow-up
NCT00118898 (22) [back to overview]Time From Randomization to Virologic Failure
NCT00118898 (22) [back to overview]Number of Participants With Regimen Failure
NCT00118898 (22) [back to overview]Number of Participants With Treatment Modification
NCT00118898 (22) [back to overview]Number of Participants With a Grade 3/4 Safety Event
NCT00135356 (18) [back to overview]Mean Percent Change From Baseline in Visceral Adipose Tissue (VAT) Area by Computed Tomography (CT) Scans and in Trunk Fat by DEXA.
NCT00135356 (18) [back to overview]Mean Percent Changes From Baseline in Fasting Lipids
NCT00135356 (18) [back to overview]Percentage of Participants With Abnormal Liver Function Tests
NCT00135356 (18) [back to overview]Percentage of Participants With Adverse Events (AEs) Leading to Discontinuation
NCT00135356 (18) [back to overview]Percentage of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs), Deaths, and AEs Leading to Discontinuation
NCT00135356 (18) [back to overview]Mean Changes From Baseline in Body Weight at Week 48 and Week 96
NCT00135356 (18) [back to overview]Change From Baseline in Trunk-to-limb Fat Ratio as Measured by DEXA at Week 96
NCT00135356 (18) [back to overview]Change From Baseline in Trunk-to-limb Fat Ratio as Measured by Dual Energy X-Ray Absortiometry (DEXA) at Week 48
NCT00135356 (18) [back to overview]Kaplan-Meier Cumulative Proportion of Participants Without Virologic Rebound (HIV RNA ≥400 c/mL) at Timepoints up to Week 96 in Treated Participants With HIV RNA <400 c/mL at Baseline
NCT00135356 (18) [back to overview]Mean Change From Baseline in CD4 Count
NCT00135356 (18) [back to overview]Mean Changes From Baseline in Body Mass Index at Week 48 and Week 96
NCT00135356 (18) [back to overview]Mean Changes From Baseline in Fasting Glucose at Week 48 and Week 96
NCT00135356 (18) [back to overview]Mean Changes From Baseline in Fasting Homeostasis Model Assessment of Insulin Resistance (HOMA-IR)
NCT00135356 (18) [back to overview]Mean Changes From Baseline in Fasting Insulin at Week 48 and Week 96
NCT00135356 (18) [back to overview]Mean Changes From Baseline in Waist Circumference at Week 48 and Week 96
NCT00135356 (18) [back to overview]Mean Changes From Baseline in Waist-to-Hip Ratio at Week 48 and Week 96
NCT00135356 (18) [back to overview]Mean Percent Change From Baseline in Peripheral Adipose Tissue (Limb Fat) by DEXA and by Changes in Subcutaneous Adipose Tissue (SAT) Area by CT Scans
NCT00135356 (18) [back to overview]Mean Percent Change From Baseline in Total Body Fat by DEXA and in Total Adipose Tissue (TAT) Area by CT Scans
NCT00144170 (92) [back to overview]Virologic Response at Week 96
NCT00144170 (92) [back to overview]Virologic Response at Week 32
NCT00144170 (92) [back to overview]Virologic Response at Week 32
NCT00144170 (92) [back to overview]Treatment Response at Week 72
NCT00144170 (92) [back to overview]Virologic Response at Week 4
NCT00144170 (92) [back to overview]Virologic Response at Week 4
NCT00144170 (92) [back to overview]Virologic Response at Week 4
NCT00144170 (92) [back to overview]Virologic Response at Week 40
NCT00144170 (92) [back to overview]Median Change From Baseline in Viral Load (Week 8)
NCT00144170 (92) [back to overview]Median Change From Baseline in Viral Load (Week 80)
NCT00144170 (92) [back to overview]Median Change From Baseline in Viral Load (Week 88)
NCT00144170 (92) [back to overview]Median Change From Baseline in Viral Load (Week 96)
NCT00144170 (92) [back to overview]Time to Confirmed Virologic Failure Through 48 Weeks of Treatment
NCT00144170 (92) [back to overview]Time to Confirmed Virologic Failure Through 96 Weeks of Treatment
NCT00144170 (92) [back to overview]Time to New Centers for Disease Control (CDC) Class C Progression Event or Death.
NCT00144170 (92) [back to overview]Time to Treatment Failure Through 48 Weeks of Treatment
NCT00144170 (92) [back to overview]Mean Change From Baseline in CD4+ Cell Count (Week 56)
NCT00144170 (92) [back to overview]Mean Change From Baseline in CD4+ Cell Count (Week 64)
NCT00144170 (92) [back to overview]Mean Change From Baseline in CD4+ Cell Count (Week 72)
NCT00144170 (92) [back to overview]Mean Change From Baseline in CD4+ Cell Count (Week 8)
NCT00144170 (92) [back to overview]Mean Change From Baseline in CD4+ Cell Count (Week 80)
NCT00144170 (92) [back to overview]Mean Change From Baseline in CD4+ Cell Count (Week 88)
NCT00144170 (92) [back to overview]Mean Change From Baseline in CD4+ Cell Count (Week 96)
NCT00144170 (92) [back to overview]Median Change From Baseline in Viral Load (Week 16)
NCT00144170 (92) [back to overview]Virologic Response at Week 40
NCT00144170 (92) [back to overview]Virologic Response at Week 40
NCT00144170 (92) [back to overview]Virologic Response at Week 48
NCT00144170 (92) [back to overview]Virologic Response at Week 48
NCT00144170 (92) [back to overview]Virologic Response at Week 96
NCT00144170 (92) [back to overview]Virologic Response at Week 48
NCT00144170 (92) [back to overview]Virologic Response at Week 96
NCT00144170 (92) [back to overview]Time to Treatment Failure Through 96 Weeks of Treatment
NCT00144170 (92) [back to overview]Virologic Response at Week 88
NCT00144170 (92) [back to overview]Median Change From Baseline in Viral Load (Week 2)
NCT00144170 (92) [back to overview]Virologic Response at Week 88
NCT00144170 (92) [back to overview]Virologic Response at Week 88
NCT00144170 (92) [back to overview]Virologic Response at Week 80
NCT00144170 (92) [back to overview]Virologic Response at Week 80
NCT00144170 (92) [back to overview]Virologic Response at Week 80
NCT00144170 (92) [back to overview]Virologic Response at Week 8
NCT00144170 (92) [back to overview]Virologic Response at Week 8
NCT00144170 (92) [back to overview]Virologic Response at Week 8
NCT00144170 (92) [back to overview]Virologic Response at Week 72
NCT00144170 (92) [back to overview]Virologic Response at Week 72
NCT00144170 (92) [back to overview]Virologic Response at Week 72
NCT00144170 (92) [back to overview]Virologic Response at Week 64
NCT00144170 (92) [back to overview]Virologic Response at Week 64
NCT00144170 (92) [back to overview]Median Change From Baseline in Viral Load (Week 24)
NCT00144170 (92) [back to overview]Median Change From Baseline in Viral Load (Week 32)
NCT00144170 (92) [back to overview]Median Change From Baseline in Viral Load (Week 4)
NCT00144170 (92) [back to overview]Median Change From Baseline in Viral Load (Week 40)
NCT00144170 (92) [back to overview]Median Change From Baseline in Viral Load (Week 48)
NCT00144170 (92) [back to overview]Mean Change From Baseline in CD4+ Cell Count (Week 48)
NCT00144170 (92) [back to overview]Median Change From Baseline in Viral Load (Week 56)
NCT00144170 (92) [back to overview]Median Change From Baseline in Viral Load (Week 64)
NCT00144170 (92) [back to overview]Median Change From Baseline in Viral Load (Week 72)
NCT00144170 (92) [back to overview]Mean Change From Baseline in CD4+ Cell Count (Week 40)
NCT00144170 (92) [back to overview]Mean Change From Baseline in CD4+ Cell Count (Week 4)
NCT00144170 (92) [back to overview]Mean Change From Baseline in CD4+ Cell Count (Week 32)
NCT00144170 (92) [back to overview]Treatment Response at Week 16
NCT00144170 (92) [back to overview]Treatment Response at Week 2
NCT00144170 (92) [back to overview]Mean Change From Baseline in CD4+ Cell Count (Week 24)
NCT00144170 (92) [back to overview]Treatment Response at Week 24
NCT00144170 (92) [back to overview]Virologic Response at Week 64
NCT00144170 (92) [back to overview]Virologic Response at Week 56
NCT00144170 (92) [back to overview]Virologic Response at Week 56
NCT00144170 (92) [back to overview]Virologic Response at Week 56
NCT00144170 (92) [back to overview]Treatment Response at Week 32
NCT00144170 (92) [back to overview]Treatment Response at Week 4
NCT00144170 (92) [back to overview]Treatment Response at Week 40
NCT00144170 (92) [back to overview]Treatment Response at Week 48
NCT00144170 (92) [back to overview]Mean Change From Baseline in CD4+ Cell Count (Week 2)
NCT00144170 (92) [back to overview]Mean Change From Baseline in CD4+ Cell Count (Week 16)
NCT00144170 (92) [back to overview]Treatment Response at Week 56
NCT00144170 (92) [back to overview]Treatment Response at Week 64
NCT00144170 (92) [back to overview]Treatment Response at Week 8
NCT00144170 (92) [back to overview]Treatment Response at Week 80
NCT00144170 (92) [back to overview]Treatment Response at Week 88
NCT00144170 (92) [back to overview]Treatment Response at Week 96
NCT00144170 (92) [back to overview]Virologic Response
NCT00144170 (92) [back to overview]Virologic Response
NCT00144170 (92) [back to overview]Virologic Response at Viral Load Nadir During Study Treatment Through 96 Weeks
NCT00144170 (92) [back to overview]Virologic Response at Week 16
NCT00144170 (92) [back to overview]Virologic Response at Week 16
NCT00144170 (92) [back to overview]Virologic Response at Week 16
NCT00144170 (92) [back to overview]Virologic Response at Week 2
NCT00144170 (92) [back to overview]Virologic Response at Week 2
NCT00144170 (92) [back to overview]Virologic Response at Week 2
NCT00144170 (92) [back to overview]Virologic Response at Week 24
NCT00144170 (92) [back to overview]Virologic Response at Week 24
NCT00144170 (92) [back to overview]Virologic Response at Week 24
NCT00144170 (92) [back to overview]Virologic Response at Week 32
NCT00145795 (10) [back to overview]Rates of Virologic Failure
NCT00145795 (10) [back to overview]Clinical HIV-related Events
NCT00145795 (10) [back to overview]Immune Reconstitution [3 Months]
NCT00145795 (10) [back to overview]Immune Reconstitution [6 Months]
NCT00145795 (10) [back to overview]Rates of ex Vivo T Cell Apoptosis: CD8+ Cell Population [3 Months]
NCT00145795 (10) [back to overview]Rates of ex Vivo T Cell Apoptosis: CD8+ Cell Population [6 Months]
NCT00145795 (10) [back to overview]Rates of ex Vivo T Cell Apoptosis: CD4+ naïve Cell Population [3 Months]
NCT00145795 (10) [back to overview]Rates of ex Vivo T Cell Apoptosis: CD4+ naïve Cell Population [6 Months]
NCT00145795 (10) [back to overview]Rates of ex Vivo T Cell Apoptosis: CD4+ Memory Cell Population [3 Months]
NCT00145795 (10) [back to overview]Rates of ex Vivo T Cell Apoptosis: CD4+ Memory Cell Population [6 Months]
NCT00207142 (17) [back to overview]Change From Baseline in HIV-1 RNA at Week 24 of the Induction Phase
NCT00207142 (17) [back to overview]Time to Suppression (Confirmed HIV-1 RNA < 400 c/mL) During Treatment Phase
NCT00207142 (17) [back to overview]Time to Suppression (Confirmed HIV-1 RNA < 50 c/mL) During Treatment Phase
NCT00207142 (17) [back to overview]Change From Baseline in CD4 Cell Count at Week 24 of Induction Phase
NCT00207142 (17) [back to overview]Change From Baseline in CD4 Cell Count at Week 48 of Rescue Phase
NCT00207142 (17) [back to overview]Kaplan-Meier Cumulative Proportion for Treatment Failure (HIV-1 RNA ≥50 c/mL) at Different Time Points Through Week 48 of the Maintenance Phase
NCT00207142 (17) [back to overview]Treatment Outcomes Based on Viral Loads (HIV-1 RNA ≥50 c/mL) Through the End of Rescue Phase
NCT00207142 (17) [back to overview]Change From End of Induction Phase in CD4 Cell Count at Week 48 of Maintenance Phase
NCT00207142 (17) [back to overview]Treatment Outcomes Based on Viral Loads (HIV-1 RNA ≥400 c/mL) Through the End of Rescue Phase
NCT00207142 (17) [back to overview]Change From Baseline in HIV-1 RNA at Week 48 of the Rescue Phase
NCT00207142 (17) [back to overview]Percentage of Participants With HIV-1 RNA <400 c/mL Through Week 48 of the Maintenance Phase
NCT00207142 (17) [back to overview]Percentage of Participants With HIV-1 RNA <50 Copies/mL (c/mL) Through Week 48 of the Maintenance Phase
NCT00207142 (17) [back to overview]Kaplan-Meier Cumulative Proportion for Treatment Failure (HIV-1 RNA ≥400 c/mL) at Different Time Points Through Week 48 of the Maintenance Phase
NCT00207142 (17) [back to overview]Percent Change From End of Induction Phase in Fasting Lipids at Week 48 of Maintenance Phase
NCT00207142 (17) [back to overview]Summary of Adverse Events During Induction Phase
NCT00207142 (17) [back to overview]Summary of Adverse Events During Maintenance Phase
NCT00207142 (17) [back to overview]Summary of Adverse Events During Rescue Phase
NCT00242216 (2) [back to overview]Proportion of Patient With Viral Load Less Than 400 Copies/mL
NCT00242216 (2) [back to overview]CD4 Cell Count Change From Baseline During Treatment.
NCT00255840 (1) [back to overview]Cumulative Treatment Failure Rate of Participants on First Line Antiretroviral Therapy Monitored by Primary Health Care Nurses (Investigative Arm)is Not Inferior to the Cumulative Treatment Failure Rate of Participants Monitored by Doctors (Control Arm).
NCT00262522 (4) [back to overview]Percentage of Subjects With Adverse Events of Diarrhea During the First 8 Weeks
NCT00262522 (4) [back to overview]Mean Change From Baseline to Week 96 in CD4+ T Cell Counts
NCT00262522 (4) [back to overview]Percentage of Subjects With Plasma Human Immunodeficiency Virus Type 1 (HIV-1) Ribonucleic Acid (RNA) Levels < 50 Copies/mL at Week 96
NCT00262522 (4) [back to overview]Percentage of Subjects With Plasma Human Immunodeficiency Virus Type 1 (HIV-1) Ribonucleic Acid (RNA) Levels < 50 Copies/mL at Week 48
NCT00270296 (2) [back to overview]Number of HIV+ Infants
NCT00270296 (2) [back to overview]Number of Participants With Virologic Suppression
NCT00296504 (21) [back to overview]Median Plasma HIV-1 RNA at Baseline and Weeks 12, 24, 48, 72, 96, 132, and 168
NCT00296504 (21) [back to overview]Median Plasma HIV-1 RNA at Baseline and Weeks 24, 48, 72, 96, 120, 144, 168, 180, 204, and 216
NCT00296504 (21) [back to overview]Median Plasma HIV-1 RNA at Weeks 180, 240, 300, 360, 420, and 432
NCT00296504 (21) [back to overview]Median Value of the Total Cholesterol/HDL Ratio at Weeks 120, 180, 204, 216, and 432
NCT00296504 (21) [back to overview]Number of Participants With Any Adverse Event (AE): Interim Analysis
NCT00296504 (21) [back to overview]Change From Baseline in Aspartate Aminotransferase (AST), Alanine Transaminase (ALT), and Serum Lipase at Weeks 48, 120, 180, 204, and 216
NCT00296504 (21) [back to overview]Change From Baseline in Aspartate Aminotransferase (AST), Alanine Transaminase (ALT), and Serum Lipase at Weeks 48, 96, 132, and 168
NCT00296504 (21) [back to overview]Change From Baseline in the Indicated Clinical Chemistry Parameters at Weeks 48, 96, 120, 132, 168, 180, 204, and 216
NCT00296504 (21) [back to overview]Change From Baseline in the Total Cholesterol/HDL Ratio at Weeks 48, 120, 180, 204, and 216
NCT00296504 (21) [back to overview]Median Values of the Indicated Clinical Chemistry Parameters at Weeks 120, 180, 204, 216, and 432
NCT00296504 (21) [back to overview]Number of Participants Enrolled in Studies APV30001 and APV300002 With the Indicated HIV-associated Conditions
NCT00296504 (21) [back to overview]Number of Participants Enrolled in Study APV30003 and Other Studies With the Indicated HIV-associated Conditions
NCT00296504 (21) [back to overview]Cluster of Differentiation Antigen 4 (CD4+) Cell Count at Baseline and Weeks 48, 120, 168, 180, 204, and 216: Observed Analysis
NCT00296504 (21) [back to overview]Number of Participants With HIV-1 Disease Progression to CDC Class C, or New CDC Class C or Death, From Baseline
NCT00296504 (21) [back to overview]Percentage of Participants With Plasma HIV-1RNA <400 and <50 Copies Per Milliliter at Baseline and Weeks 12, 24, 48, 60, 96, and 132 (MD=F and Observed)
NCT00296504 (21) [back to overview]Percentage of Participants With Plasma HIV-1RNA <50 Copies Per Milliliter at Baseline and Weeks 120, 180, 240, 300, 360, 420, and 432 (Observed)
NCT00296504 (21) [back to overview]Cluster of Differentiation Antigen 4 (CD4+) Cell Count at Baseline and Weeks 24, 48, 96, 132, and 168: Observed Analysis
NCT00296504 (21) [back to overview]Number of Participants With Any Adverse Event (AE): Final Analysis
NCT00296504 (21) [back to overview]Median Aspartate Aminotransferase (AST), Alanine Transaminase (ALT), and Serum Lipase Values at Weeks 120, 180, 204, 216, and 432
NCT00296504 (21) [back to overview]Change From Baseline in the Total Cholesterol/HDL Ratio at Weeks 48, 96, 132, and 168
NCT00296504 (21) [back to overview]Percentage of Participants With Plasma HIV-1 Ribonucleic Acid (RNA) <400 and <50 Copies Per Milliliter at Baseline and Weeks 48, 120, 180, and 216 (MD=F and Observed)
NCT00307151 (9) [back to overview]Change in CD4 Percent From Entry to Week 48
NCT00307151 (9) [back to overview]Percent of Participants Experiencing Virologic Failure
NCT00307151 (9) [back to overview]Time From Start of Study Treatment to First New Grade >=3 Lab Abnormality, Sign or Symptom Occurring on Study Treatment
NCT00307151 (9) [back to overview]Time From Randomization to Virologic Failure
NCT00307151 (9) [back to overview]Time From Randomization to Treatment Failure, Defined as Virologic Failure or Permanent Discontinuation of the Randomized NNRTI or PI Component of Study Treatment
NCT00307151 (9) [back to overview]Time From Randomization to HIV-related Disease Progression or Death
NCT00307151 (9) [back to overview]Time From Randomization to Death
NCT00307151 (9) [back to overview]Percent of Participants With Treatment Failure, Defined as a Confirmed Virologic Failure or Permanent Discontinuation of the Randomized NNRTI or PI Component of Study Treatment
NCT00307151 (9) [back to overview]Number of Participants Developing New NRTI, NNRTI or PI-resistant Virus
NCT00326716 (27) [back to overview]Infant HIV Status
NCT00326716 (27) [back to overview]Infant Gender
NCT00326716 (27) [back to overview]Median Change From Baseline to Day of Delivery in Maternal Cluster of Differentiation 4 (CD4) Cell Count
NCT00326716 (27) [back to overview]Mean RTV Trough Plasma Concentration (Cmin) 24 Hours Following the Daily Dose
NCT00326716 (27) [back to overview]Mean RTV Time of Maximum Observed Plasma Concentration (Tmax)
NCT00326716 (27) [back to overview]Mean RTV Terminal Elimination Half Life (T 1/2)
NCT00326716 (27) [back to overview]Mean RTV Maximum Plasma Concentration (Cmax) in One Dosing Interval
NCT00326716 (27) [back to overview]Mean RTV Area Under the Concentration Curve (AUC TAU)
NCT00326716 (27) [back to overview]Mean HIV RNA Level at Baseline
NCT00326716 (27) [back to overview]Mean CD4 Cell Count at Baseline
NCT00326716 (27) [back to overview]Mean ATV Trough Plasma Concentration (Cmin) 24 Hours Following the Daily Dose
NCT00326716 (27) [back to overview]Mean ATV Time of Maximum Observed Plasma Concentration (Tmax)
NCT00326716 (27) [back to overview]Mean ATV Terminal Elimination Half Life (T 1/2)
NCT00326716 (27) [back to overview]Mean ATV Maximum Plasma Concentration (Cmax) in One Dosing Interval
NCT00326716 (27) [back to overview]Mean ATV Area Under the Concentration Curve (AUC TAU)
NCT00326716 (27) [back to overview]Infant Gestational Age at Delivery
NCT00326716 (27) [back to overview]Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT00326716 (27) [back to overview]Number of Participants With Grade 2 to Grade 4 AEs and SAEs
NCT00326716 (27) [back to overview]SAEs in Enrolled Infants
NCT00326716 (27) [back to overview]SAEs in Enrolled Mothers
NCT00326716 (27) [back to overview]Maternal HIV Ribonucleic Acid (RNA) Level on Day of Delivery
NCT00326716 (27) [back to overview]Median Change From Baseline to Day of Delivery in Maternal HIV RNA Level
NCT00326716 (27) [back to overview]Mean Atazanavir Maternal Plasma Concentration and Neonatal Cord Blood Concentration
NCT00326716 (27) [back to overview]Mean Atazanavir Plasma Protein Binding
NCT00326716 (27) [back to overview]Median Infant Total Bilirubin Level
NCT00326716 (27) [back to overview]Multicenter AIDS Cohort Study (MACS) Participant Adherence to Regimen and Drug Components for ATV 300 mg / RTV 100 mg Test Dose
NCT00326716 (27) [back to overview]Infant Race
NCT00335322 (2) [back to overview]Time-weighted Mean Change From Baseline Plasma HIV-RNA.
NCT00335322 (2) [back to overview]Time Weighted Mean Change From Baseline Plasma HIV-RNA
NCT00337467 (13) [back to overview]Number of Participants With Genotype Substitutions for Virologic Rebounds (HIV-RNA ≥ 400 c/mL) Through Week 48
NCT00337467 (13) [back to overview]Percentage of Participants With Virological Rebound Through Week 96
NCT00337467 (13) [back to overview]Percentage of Participants With Treatment Failure Through Week 96
NCT00337467 (13) [back to overview]Percentage of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs), Deaths, and Discontinuations Due to AEs
NCT00337467 (13) [back to overview]Number of Participants With Genotype Substitutions for Virologic Rebounds (HIV-RNA ≥ 400 c/mL) Through Week 96
NCT00337467 (13) [back to overview]Mean Percent Changes From Baseline in Fasting Total Cholesterol, High Density Lipoprotein (HDL) Cholesterol, Non-HDL Cholesterol, Low Density Lipoprotein (LDL) Cholesterol, and Triglycerides at Week 96
NCT00337467 (13) [back to overview]Mean Percent Changes From Baseline in Fasting Total Cholesterol, High Density Lipoprotein (HDL) Cholesterol, Non-HDL Cholesterol, Low Density Lipoprotein (LDL) Cholesterol, and Triglycerides at Week 48
NCT00337467 (13) [back to overview]Mean Change From Baseline in Cluster of Differentiation 4 (CD4) Cell Count at Week 24
NCT00337467 (13) [back to overview]Mean Change From Baseline in CD4 Cell Count at Week 96
NCT00337467 (13) [back to overview]Mean Change From Baseline in CD4 Cell Count at Week 48
NCT00337467 (13) [back to overview]Cumulative Proportion of Participants Without Treatment Failure Through Week 100
NCT00337467 (13) [back to overview]Percentage of Participants With Treatment Failure Through Week 48
NCT00337467 (13) [back to overview]Percentage of Participants With Virological Rebound Through Week 48
NCT00342355 (2) [back to overview]Serious Adverse Events
NCT00342355 (2) [back to overview]Progression to AIDS or Death in tx naïve Pts With Adv HIV dx in the Four Randomly Assigned Regimens.
NCT00357552 (11) [back to overview]Number of Subjects With at Least One New PI-associated Resistance Mutation at Time of Virologic Failure.
NCT00357552 (11) [back to overview]Percentage of Subjects Reporting Not Skipping Medications in the Last Month.
NCT00357552 (11) [back to overview]Number of Screened Subjects With at Least One NNRTI, or NRTI-associated Resistance Mutation at A5230 Screening.
NCT00357552 (11) [back to overview]Proportion of Participants With Plasma HIV-1 RNA Levels < 400 Copies/mL From Baseline to Week 104
NCT00357552 (11) [back to overview]Level of HIV-1 RNA as Ascertained From Paired DBS and Plasma
NCT00357552 (11) [back to overview]Time to Treatment Failure, Defined as the First Occurrence of Death, Disease Progression, or Virologic Failure.
NCT00357552 (11) [back to overview]Time to First New Grade 3 or 4 Sign or Symptom or Laboratory Toxicity Following LPV/r Intensification
NCT00357552 (11) [back to overview]Probability of Grade 3 or 4 Sign or Symptom, or Laboratory Toxicity Over 24 Weeks on Study.
NCT00357552 (11) [back to overview]Percentage of Enrolled Participants With Virologic Success at Week 24 on LPV/r Monotherapy
NCT00357552 (11) [back to overview]Number of Participants With Study-targeted Diagnoses and Clinical Events
NCT00357552 (11) [back to overview]Change in CD4+ Cell Counts From Study Entry to Week 104
NCT00358917 (5) [back to overview]Virologic Response (HIV-1 RNA <50 Copies/mL) at Week 48 for Participants With 0-2 Protease Inhibitor Substitutions at Baseline Associated With Reduced Response to Lopinavir/Ritonavir
NCT00358917 (5) [back to overview]Percentage of Participants With Plasma Human Immunodeficiency Virus Type 1 (HIV-1) Ribonucleic Acid (RNA) Levels < 50 Copies/Milliliter (mL) at Week 48
NCT00358917 (5) [back to overview]Percentage of Participants With New Primary Protease Mutations at Week 48
NCT00358917 (5) [back to overview]Percentage of Participants Responding at Week 48 Based on the Food and Drug Administration (FDA) Time to Loss of Virologic Response (TLOVR) Algorithm
NCT00358917 (5) [back to overview]Mean Change From Baseline to Week 48 in Cluster of Differentiation 4 Single-Positive Thymocyte (CD4+ T) Cell Counts
NCT00363142 (11) [back to overview]Steady-State Plasma Levels of Amprenavir (APV) and Ritonavir (RTV) Ctau at Weeks 12 and 24
NCT00363142 (11) [back to overview]Number of Participants With Grade 2-4 Adverse Events Occurring in Greater Than or Equal to 2% of Subjects Through Week 24
NCT00363142 (11) [back to overview]Percentage of Participants With Plasma HIV-1 RNA <50 Copies/mL at Week 24, TLOVR Analysis
NCT00363142 (11) [back to overview]Percentage of Participants With Plasma Human Immunodeficiency Virus, Type 1, Ribonucleic Acid (HIV-1 RNA) <400 Copies/mL at Week 24, Time to Loss of Virologic Response (TLOVR) Analysis
NCT00363142 (11) [back to overview]Number of Participants Who Discontinued Treatment Due to Adverse Events Through Week 24
NCT00363142 (11) [back to overview]Percentage of Participants Not Meeting the Definition of Virologic Failure at or Prior to Week 24
NCT00363142 (11) [back to overview]Percent Change From Baseline in Low Density Lipoprotein (LDL) at Week 24
NCT00363142 (11) [back to overview]Number of Participants With Plasma HIV-1 RNA Genotypic Mutations and Phenotypic Resistance at Time of Virologic Failure Not Present at Baseline
NCT00363142 (11) [back to overview]Median Change From Baseline of CD4+ Cell Count at Week 24, Observed Analysis
NCT00363142 (11) [back to overview]Mean Change From Baseline of log10 Copies/mL Plasma HIV-1 RNA Levels at Week 24, Observed Analysis
NCT00363142 (11) [back to overview]Percent Change From Baseline in Total Cholesterol, High Density Lipoprotein (HDL), and Triglycerides at Week 24
NCT00381303 (10) [back to overview]Descriptive Statistics of ETR Subgroup - Change From Baseline in CD4+ Cell Count Using the Imputation Method of LOCF
NCT00381303 (10) [back to overview]Descriptive Statistics of Change From Baseline in CD4+ Cell Count Using Observed Values
NCT00381303 (10) [back to overview]Descriptive Statistics of Change From Baseline in CD4+ Cell Count Using the Imputation Method of Last Observation Carried Forward (LOCF)
NCT00381303 (10) [back to overview]Descriptive Statistics of [TLOVR Non-virologic Failure (VF) Censored] - VL < 50 HIV-1 RNA by Race
NCT00381303 (10) [back to overview]Descriptive Statistics of ETR Subgroup - Change From Baseline in CD4+ Cell Using Observed Values
NCT00381303 (10) [back to overview]Descriptive Statistics of ETR Subgroup [TLOVR Non-virologic Failure (VF) Censored] - VL < 50 HIV-1 RNA
NCT00381303 (10) [back to overview]Number of Etravirine-TMC125 (ETR) Subgroup- VL < 50 HIV-1 RNA Copies/mL (TLOVR) Subjects
NCT00381303 (10) [back to overview]Number of TLOVR Non-virologic Failure (VF) Censored - VL < 50 HIV-1 RNA Subjects by Sex
NCT00381303 (10) [back to overview]Number of Viral Load (VL) < 50 HIV-1 RNA Copies/mL (Time to Loss of Virologic Response[TLOVR]) Subjects by Sex
NCT00381303 (10) [back to overview]Number of VL < 50 HIV-1 RNA Copies/mL (TLOVR) Subjects by Race
NCT00413153 (9) [back to overview]Total Bilirubin
NCT00413153 (9) [back to overview]Insulin Sensitivity
NCT00413153 (9) [back to overview]Liver Enzymes -- Aspartate Aminotransferase (AST)
NCT00413153 (9) [back to overview]Body Composition - Visceral Adipose Tissue
NCT00413153 (9) [back to overview]Fasting Glucose
NCT00413153 (9) [back to overview]Glucose Trafficking
NCT00413153 (9) [back to overview]Immune Parameters -- CD4 Count
NCT00413153 (9) [back to overview]Liver Enzymes -- Alanine Aminotransferase (ALT)
NCT00413153 (9) [back to overview]Lipid Metabolism - Serum Triglyceride
NCT00414518 (3) [back to overview]Viral Set Point
NCT00414518 (3) [back to overview]Number of Participants Experiencing Either an AIDS-defining Event, a Grade 3 or 4 Adverse Event, or Acute Retroviral Syndrome
NCT00414518 (3) [back to overview]Plasma HIV-1 Viral Load (Copies/ml) at Week 24 as Compared Between the Two Arms
NCT00427297 (4) [back to overview]Incidence of Severe Adverse Events (Excluding Mortality)
NCT00427297 (4) [back to overview]Incidence of Mortality
NCT00427297 (4) [back to overview]Immunologic Failure
NCT00427297 (4) [back to overview]Viral Failure
NCT00435929 (11) [back to overview]Number Participants With Abnormal Vital Signs
NCT00435929 (11) [back to overview]Maximum Observed Plasma Concentration (Cmax) of SQV and RTV
NCT00435929 (11) [back to overview]Number of Participants With Abnormal Electrocardiogram (ECG) Findings
NCT00435929 (11) [back to overview]Number of Participants With the Indicated Grade 3 and Grade 4 Laboratory Parameters
NCT00435929 (11) [back to overview]Plasma Clearance After Oral Administration (CL/F) of SQV and RTV
NCT00435929 (11) [back to overview]Terminal Half-life (T1/2) of SQV and RTV
NCT00435929 (11) [back to overview]Minimum Observed Plasma Concentration (Cmin) of SQV and RTV
NCT00435929 (11) [back to overview]Time of Maximum Plasma Concentration (Tmax) of SQV and RTV
NCT00435929 (11) [back to overview]Area Under the Plasma Concentration-Time Curve From Time of Administration to 12 Hours After Dosing (AUC 0-12h) of Saquinavir (SQV) and Ritonavir (RTV)
NCT00435929 (11) [back to overview]Cluster of Differentiation 4 (CD4 ) Count
NCT00435929 (11) [back to overview]Volume of Distribution (Vd) of SQV and RTV
NCT00440947 (26) [back to overview]Mean Age at Baseline of Participants Randomized to Treatment for the 48-Week Randomized Phase
NCT00440947 (26) [back to overview]Change From Baseline in HIV-1 RNA at Week 84
NCT00440947 (26) [back to overview]Change From Baseline in HIV-1 RNA at Week 36
NCT00440947 (26) [back to overview]Change From Baseline in HIV-1 RNA at Week 144
NCT00440947 (26) [back to overview]Mean Percent Compliance at Week 84
NCT00440947 (26) [back to overview]Change From Baseline in CD4+ Cell Count at Week 84
NCT00440947 (26) [back to overview]Change From Baseline in CD4+ Cell Count at Week 36
NCT00440947 (26) [back to overview]Change From Baseline in CD4+ Cell Count at Week 144
NCT00440947 (26) [back to overview]Percentage of Participants Who Achieved Plasma HIV-1 RNA <50 c/ml at the Week 84 Visit
NCT00440947 (26) [back to overview]Percentage of Participants Who Achieved Plasma HIV-1 RNA <50 c/ml at the Week 36 Visit
NCT00440947 (26) [back to overview]Percentage of Participants Who Achieved Plasma HIV-1 RNA <50 c/ml at the Week 144 Visit
NCT00440947 (26) [back to overview]Percentage of Participants Who Achieved HIV-1 RNA <400 c/ml at the Week 144 Visit
NCT00440947 (26) [back to overview]Number of Participants Who Met the Protocol-defined Virologic Failure (PDVF) Criteria at Week 36
NCT00440947 (26) [back to overview]Number of Participants Who Met the PDVF Criteria at Week 84
NCT00440947 (26) [back to overview]Number of Participants Who Met the PDVF Criteria at Week 144
NCT00440947 (26) [back to overview]Number of Confirmed Virologic Failure Participants With Treatment-emergent HIV Genotypic Resistance in Reverse Transcriptase and Protease From Week 84 Through Week 144
NCT00440947 (26) [back to overview]Number of Confirmed Virologic Failure Participants With Treatment-emergent HIV Genotypic Resistance in Reverse Transcriptase and Protease From Randomization at Week 36 Through Week 84
NCT00440947 (26) [back to overview]Percentage of Participants Who Achieved HIV-1 RNA <400 c/ml at the Week 84 Visit
NCT00440947 (26) [back to overview]Number of Confirmed Virologic Failure Participants With Treatment-emergent HIV Genotypic Resistance in Reverse Transcriptase and Protease From Baseline Through Week 36
NCT00440947 (26) [back to overview]Percentage of Participants Who Achieved Plasma HIV-1 RNA <400 c/ml at the Week 36 Visit
NCT00440947 (26) [back to overview]Number of Confirmed Virologic Failure Participants From Week 84 Through Week 144 With Treatment-emergent Reductions in HIV Susceptibility to Abacavir, Lamivudine, Atazanavir, or Ritonavir
NCT00440947 (26) [back to overview]Number of Confirmed Virologic Failure Participants From Randomization at Week 36 Through Week 84 With Treatment-emergent Reductions in HIV Susceptibility to Abacavir, Lamivudine, Atazanavir, or Ritonavir
NCT00440947 (26) [back to overview]Number of Confirmed Virologic Failure Participants From Baseline Through Week 36 With Treatment-emergent Reductions in Susceptibility to Abacavir, Lamivudine, Atazanavir, or Ritonavir
NCT00440947 (26) [back to overview]Mean Percent Compliance at Week 36
NCT00440947 (26) [back to overview]Mean Percent Compliance at Week 144
NCT00440947 (26) [back to overview]Percentage of Participants (PAR) Who Achieved Plasma HIV-1 RNA <50 Copies (c) /Milliliter (ml) at the Week 84 Visit
NCT00443703 (23) [back to overview]Number of Patients With Clinical Adverse Experiences (CAEs) Through 24 Weeks
NCT00443703 (23) [back to overview]Mean Percent Change From Baseline in Fasting Serum Cholesterol at Week 12
NCT00443703 (23) [back to overview]Mean Percent Change From Baseline in Fasting Serum Cholesterol at Week 24
NCT00443703 (23) [back to overview]Mean Percent Change From Baseline in Fasting Serum High-Density Lipoprotein Cholesterol (HDL-C) at Week 12
NCT00443703 (23) [back to overview]Mean Percent Change From Baseline in Fasting Serum Low-Density Lipoprotein Cholesterol (LDL-C) at Week 12
NCT00443703 (23) [back to overview]Mean Percent Change From Baseline in Fasting Serum Low-Density Lipoprotein Cholesterol (LDL-C) at Week 24
NCT00443703 (23) [back to overview]Mean Percent Change From Baseline in Non-High-Density Lipoprotein Cholesterol (Non-HDL-C) at Week 12
NCT00443703 (23) [back to overview]Mean Percent Change From Baseline in Non-High-Density Lipoprotein Cholesterol (Non-HDL-C) at Week 24
NCT00443703 (23) [back to overview]Median Percent Change From Baseline in Serum Triglyceride at Week 12
NCT00443703 (23) [back to overview]Median Percent Change From Baseline in Serum Triglyceride at Week 24
NCT00443703 (23) [back to overview]Number of Patients With Plasma Human Immunodeficiency Virus (HIV) RiboNucleic Acid (RNA) <50 Copies/mL at Week 24
NCT00443703 (23) [back to overview]Number of Patients That Died by 24 Week Last Patient Last Visit
NCT00443703 (23) [back to overview]Number of Patients That Discontinued Due to CAEs Through 24 Weeks
NCT00443703 (23) [back to overview]Number of Patients That Discontinued Due to Drug Related CAEs Through 24 Weeks
NCT00443703 (23) [back to overview]Number of Patients That Discontinued Due to LAEs Through 24 Weeks
NCT00443703 (23) [back to overview]Number of Patients That Discontinued With Drug Related LAEs Through 24 Weeks
NCT00443703 (23) [back to overview]Number of Patients With Drug-related CAEs Through 24 Weeks
NCT00443703 (23) [back to overview]Number of Patients With Drug-related Laboratory Adverse Experiences (LAEs) Through 24 Weeks
NCT00443703 (23) [back to overview]Number of Patients With Serious CAEs Through 24 Weeks
NCT00443703 (23) [back to overview]Number of Patients With Laboratory Adverse Experiences (LAEs) Through 24 Weeks
NCT00443703 (23) [back to overview]Number of Patients With Serious LAEs Through 24 Weeks
NCT00443703 (23) [back to overview]Mean Percent Change From Baseline in Fasting Serum High-Density Lipoprotein Cholesterol (HDL-C) at Week 24
NCT00443703 (23) [back to overview]Number of Patients With Serious Drug-related CAEs Through 24 Weeks
NCT00443729 (21) [back to overview]Mean Percent Change From Baseline in Fasting Serum High-density Lipoprotein Cholesterol (HDL-C) at Week 24
NCT00443729 (21) [back to overview]Mean Percent Change From Baseline in Fasting Serum Low-density Lipoprotein Cholesterol (LDL-C) at Week 12
NCT00443729 (21) [back to overview]Mean Percent Change From Baseline in Fasting Serum Low-density Lipoprotein Cholesterol (LDL-C) at Week 24
NCT00443729 (21) [back to overview]Mean Percent Change From Baseline in Non-high-density Lipoprotein Cholesterol (Non-HDL-C) at Week 12
NCT00443729 (21) [back to overview]Mean Percent Change From Baseline in Non-high-density Lipoprotein Cholesterol (Non-HDL-C) at Week 24
NCT00443729 (21) [back to overview]Median Percent Change From Baseline in Serum Triglyceride at Week 12
NCT00443729 (21) [back to overview]Median Percent Change From Baseline in Serum Triglyceride at Week 24
NCT00443729 (21) [back to overview]Number of Patients With Plasma Human Immunodeficiency Virus (HIV) RiboNucleic Acid (RNA) <50 Copies/mL at Week 24
NCT00443729 (21) [back to overview]Number of Patients That Died by 24 Week Last Patient Last Visit
NCT00443729 (21) [back to overview]Number of Patients That Discontinued Due to CAEs Through 24 Weeks
NCT00443729 (21) [back to overview]Number of Patients That Discontinued Due to LAEs Through 24 Weeks
NCT00443729 (21) [back to overview]Number of Patients With Clinical Adverse Experiences (CAEs) Through 24 Weeks
NCT00443729 (21) [back to overview]Number of Patients With Drug-related CAEs Through 24 Weeks
NCT00443729 (21) [back to overview]Number of Patients With Drug-related LAEs Through 24 Weeks
NCT00443729 (21) [back to overview]Number of Patients With Laboratory Adverse Experiences (LAEs) Through 24 Weeks
NCT00443729 (21) [back to overview]Number of Patients With Serious CAEs Through 24 Weeks
NCT00443729 (21) [back to overview]Number of Patients With Serious Drug-related CAEs Through 24 Weeks
NCT00443729 (21) [back to overview]Number of Patients With Serious LAEs Through 24 Weeks
NCT00443729 (21) [back to overview]Mean Percent Change From Baseline in Fasting Serum Cholesterol at Week 12
NCT00443729 (21) [back to overview]Mean Percent Change From Baseline in Fasting Serum Cholesterol at Week 24
NCT00443729 (21) [back to overview]Mean Percent Change From Baseline in Fasting Serum High-density Lipoprotein Cholesterol (HDL-C) at Week 12
NCT00447902 (1) [back to overview]Frequency of Patients (%) With Possible Clinically Significant Abnormalities of Laboratory Measurements
NCT00450580 (7) [back to overview]Steady-state Levels of Amprenavir (APV) and Ritonavir (RTV) Ctau at Weeks 4, 12, and 24
NCT00450580 (7) [back to overview]Number of Participants With HIV-1 RNA <400 Copies/mL (Primary Endpoint) at Week 48 Categorised by Baseline Viral Load, TLOVR Analysis
NCT00450580 (7) [back to overview]Number of Protocol-defined Virological Failures With Genotypic and Phenotypic Resistance Changes
NCT00450580 (7) [back to overview]Percentage of Participants With HIV-1 RNA <400 and >=400 Copies/mL Over 48 Weeks
NCT00450580 (7) [back to overview]Number of Participants With HIV-1 RNA <400 Copies/mL (Primary Endpoint) at Week 48 Categorised by Baseline CD4+ Count, TLOVR Analysis
NCT00450580 (7) [back to overview]Percentage of Participants With HIV-1 RNA <50 and >=50 Copies/mL by Visit Over 48 Weeks
NCT00450580 (7) [back to overview]Change From Baseline in Non-HDL Cholesterol at Week 48
NCT00460746 (10) [back to overview]Median Change From Baseline in LDL Cholesterol at Week 48.
NCT00460746 (10) [back to overview]CD4+ Cell Count (x 10^6 Cell/L): Baseline and Mean Changes From Baseline at 4, 8, 12, 16, 24,36 and 48 Weeks.
NCT00460746 (10) [back to overview]CD4+ Cell Count (x 10^6 Cell/L): Baseline and Median Changes From Baseline at 4, 8, 12, 16, 24, 36 and 48 Weeks.
NCT00460746 (10) [back to overview]Median Change From Baseline in HDL Cholesterol.
NCT00460746 (10) [back to overview]Median Change From Baseline in Total Cholesterol (TC) / High Denisty Lipoprotein (HDL) Ratio at Week 48.
NCT00460746 (10) [back to overview]Median Change From Baseline in Total Cholesterol at Week 48.
NCT00460746 (10) [back to overview]Median Change From Baseline in Triglycerides at Week 48.
NCT00460746 (10) [back to overview]Proportion of Patients Who Have Viral Load Measurements <50 Copies/ml at 2, 4, 8, 12, 16, 24, 36 and 48 Weeks After Switching to DRV/r and ETR, Missing Equals Failure.
NCT00460746 (10) [back to overview]Proportion of Patients Who Maintain Plasma HIV Viral Load Measurements < 400 Copies/ml at 2, 4, 8, 12, 16, 24, 36 and 48 Weeks After Switching to DRV/r and ETR, Missing Equals Failure.
NCT00460746 (10) [back to overview]Median Change From Baseline in Glucose at Week 48.
NCT00474201 (1) [back to overview]Gemfibrozil Area Under the Concentration vs. Time Curve (AUC)
NCT00486330 (1) [back to overview]Area Under the Curve of BUP/NLX With TPV/r (h*ng/mL)
NCT00524368 (12) [back to overview]Time-averaged Difference (DAVG) of log10 Plasma Viral Load Over 48 Weeks
NCT00524368 (12) [back to overview]Virologic Response at Week 48 (Viral Load Less Than 400 Copies/mL)
NCT00524368 (12) [back to overview]Virological Response at Week 48 (Number of Participants With Plasma Viral Load Less Than 50 Copies/mL) - as Defined by the Time to Loss of Virologic Response (TLOVR) Algorithm
NCT00524368 (12) [back to overview]Predose Plasma Concentration (C0h) of DRV and Rtv.
NCT00524368 (12) [back to overview]Percentage of Participants Adherent/Non-adherent to ARV as Determined by Modified Medication Adherence Self Report Inventory (M-MASRI) Questionnaire at Week 48
NCT00524368 (12) [back to overview]Number of Participants Developing Mutations at Endpoint
NCT00524368 (12) [back to overview]Change in log10 Viral Load From Baseline at Week 48
NCT00524368 (12) [back to overview]Time to Loss of Virologic Response
NCT00524368 (12) [back to overview]Area Under the Curve From the Time of Study Medication Administration Upto 24 Hour Postdose (AUC24h) of DRV and Rtv
NCT00524368 (12) [back to overview]Change in CD4+ Cell Count From Baseline
NCT00524368 (12) [back to overview]Time to Reach First Virologic Response
NCT00524368 (12) [back to overview]Change From Baseline in Total Functional Assessment of HIV Infection (FAHI) Score
NCT00525733 (1) [back to overview]The Primary Outcome of This Study is the Proportion of Patients Having Detectable HIV-1 RNA Using the Single Copy Assay After 48 Weeks of Treatment and the Study Hypothesis is That New Treatment is Better Than the Control Group.
NCT00530920 (17) [back to overview]Terminal Half-Life (t1/2) of Tipranavir
NCT00530920 (17) [back to overview]Volume of Distribution (V/F) of Ritonavir
NCT00530920 (17) [back to overview]Volume of Distribution (V/F) of Tipranavir
NCT00530920 (17) [back to overview]Clinical Abnormal Findings in Laboratory and Physical Examination
NCT00530920 (17) [back to overview]Tmax of Ritonavir
NCT00530920 (17) [back to overview]Trough Concentration (Cmin) of Tipranavir
NCT00530920 (17) [back to overview]Viral Load (log10 Copies/mL) Change From Baseline (Last Observation Carried Forward (LOCF))
NCT00530920 (17) [back to overview]Apparent Oral Clearance I(Cl/F) of Ritonavir
NCT00530920 (17) [back to overview]Time to Cmax (Tmax) of Tipranavir
NCT00530920 (17) [back to overview]Apparent Oral Clearance I(Cl/F) of Tipranavir
NCT00530920 (17) [back to overview]Area Under the Curve(AUC) of Tipranavir 24 h for Once Daily (QD) and AUC 12 h for Twice Daily (BID)
NCT00530920 (17) [back to overview]AUC 24 of Ritonavir for QD and AUC 12 of Ritonavir for BID
NCT00530920 (17) [back to overview]Cmax of Ritonavir
NCT00530920 (17) [back to overview]Concentration-24 Hour (hr) Post Dose of Tipranavir - (Cp 24 h for QD and 12 hr Post Dose (CP 12h) for BID
NCT00530920 (17) [back to overview]Cp 24 h of Ritonavir for QD and CP 12 h of Ritonavir for BID
NCT00530920 (17) [back to overview]Maximum Concentration (Cmax) of Tipranavir
NCT00530920 (17) [back to overview]Terminal Half-Life (t1/2) of Ritonavir
NCT00531206 (21) [back to overview]Total Cholesterol Over Time
NCT00531206 (21) [back to overview]Triglycerides Over Time
NCT00531206 (21) [back to overview]Use of Lipid Lowering Agents During the Study
NCT00531206 (21) [back to overview]Creatinine Over Time
NCT00531206 (21) [back to overview]Number of Anti-retroviral Medications Taken in Combination With Tipranavir/Ritonavir
NCT00531206 (21) [back to overview]Low Density Lipoprotein (HDL) Cholesterol Over Time
NCT00531206 (21) [back to overview]High Density Lipoprotein (HDL) Cholesterol Over Time
NCT00531206 (21) [back to overview]Total Bilirubin Over Time
NCT00531206 (21) [back to overview]Adverse Events
NCT00531206 (21) [back to overview]Adverse Events Related to Therapy With Tipranavir/Ritonavir Based on Investigator's Opinion
NCT00531206 (21) [back to overview]Deaths
NCT00531206 (21) [back to overview]Discontinuations Due to an Adverse Event
NCT00531206 (21) [back to overview]Gamma-glutamyl Transpeptidase (GGT) Over Time
NCT00531206 (21) [back to overview]Serious Adverse Events
NCT00531206 (21) [back to overview]Alanine Aminotransferase (ALT) Over Time
NCT00531206 (21) [back to overview]Alkaline Phosphatase Over Time
NCT00531206 (21) [back to overview]Aspartate Aminotransferase (ALT) Over Time
NCT00531206 (21) [back to overview]Body Mass Index Class (Kilograms/Square Meter)
NCT00531206 (21) [back to overview]Subjective Well-being
NCT00531206 (21) [back to overview]CD4+ Cell Count
NCT00531206 (21) [back to overview]Change in Viral Load
NCT00534352 (12) [back to overview]CD4+ Cell Count (Percent): Baseline and Median Changes From Baseline (ITT-Observed Case)
NCT00534352 (12) [back to overview]Number of Participants Contributing to the Pharmacokinetic (PK) Evaluations: Cmin, Cmax, AUC24 & Css,av
NCT00534352 (12) [back to overview]Virologic Response < 50 HIV-1 RNA Copies/mL (ITT-Observed Case)
NCT00534352 (12) [back to overview]Number of Participants With Treatment-Emergent Non-Graded Laboratory Abnormalities(Worst Abnormality): Glucose- Insulin
NCT00534352 (12) [back to overview]Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities(Worst Grade): Lipids- Triglycerides
NCT00534352 (12) [back to overview]Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities(Worst Grade): Lipids- Low-density Lipoprotein (LDL) Direct
NCT00534352 (12) [back to overview]Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities (Worst Grade): Lipids- Total Cholesteral
NCT00534352 (12) [back to overview]Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities (Worst Grade): Glucose-Hyperglycemia
NCT00534352 (12) [back to overview]Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities (Worst Grade): Glucose- Hypoglycemia
NCT00534352 (12) [back to overview]Log10 Viral Load (HIV-1 RNA Copies/mL): Mean Changes From Baseline(ITT-Observed Case)
NCT00534352 (12) [back to overview]CD4+ Cell Count (x 10^6 Cell/L): Baseline and Median Changes From Baseline (ITT-Observed Case)
NCT00534352 (12) [back to overview]Number of Participants With Treatment-Emergent Non-Graded Laboratory Abnormalities(Worst Abnormality): Lipids- High-density Lipoprotein (HDL)
NCT00552240 (46) [back to overview]Change in Fasting Low Density Lipoprotein (LDL)Cholesterol Level
NCT00552240 (46) [back to overview]Change in Fasting Plasma Total Cholesterol Level
NCT00552240 (46) [back to overview]Change in Fasting Plasma Triglycerides Level
NCT00552240 (46) [back to overview]Change in Fasting Total Cholesterol to High Density Lipoprotein (HDL) Ratio
NCT00552240 (46) [back to overview]Change in Framingham Score
NCT00552240 (46) [back to overview]Change in Glomerular Filtration Rate (GFR) From Baseline to Week 48
NCT00552240 (46) [back to overview]Percentage Adherence by Pill Count
NCT00552240 (46) [back to overview]Proportion of Patients Reporting CNS Side Effects of Any Severity
NCT00552240 (46) [back to overview]Proportion of Patients Reporting Hepatic Events of Any Severity
NCT00552240 (46) [back to overview]Proportion of Patients Reporting Rash of Any Severity
NCT00552240 (46) [back to overview]Time to Virologic Response (First Confirmed Viral Load < 50 Copies/ml), All Participants
NCT00552240 (46) [back to overview]Time to Virologic Response (First Confirmed Viral Load < 50 Copies/ml), Only Participants With Confirmed Viral Load < 50 Copies/ml
NCT00552240 (46) [back to overview]Incidence of Patients With AIDS Progression at Each Visit
NCT00552240 (46) [back to overview]Number of Participants With HIV Viral Load < 400 Copies/ml at Week 12 of Treatment
NCT00552240 (46) [back to overview]Number of Participants With HIV Viral Load < 400 Copies/ml at Week 2 of Treatment
NCT00552240 (46) [back to overview]Number of Participants With HIV Viral Load < 400 Copies/ml at Week 24 of Treatment
NCT00552240 (46) [back to overview]Number of Participants With HIV Viral Load < 400 Copies/ml at Week 36 of Treatment
NCT00552240 (46) [back to overview]Number of Participants With HIV Viral Load < 400 Copies/ml at Week 4 of Treatment
NCT00552240 (46) [back to overview]Number of Participants With HIV Viral Load < 400 Copies/ml at Week 48 of Treatment
NCT00552240 (46) [back to overview]Number of Participants With HIV Viral Load < 400 Copies/ml at Week 6 of Treatment
NCT00552240 (46) [back to overview]Number of Participants With HIV Viral Load < 400 Copies/ml at Week 8 of Treatment
NCT00552240 (46) [back to overview]Number of Participants With HIV Viral Load < 50 Copies/ml at Week 12 of Treatment
NCT00552240 (46) [back to overview]Number of Participants With HIV Viral Load < 50 Copies/ml at Week 2 of Treatment
NCT00552240 (46) [back to overview]Number of Participants With HIV Viral Load < 50 Copies/ml at Week 24 of Treatment
NCT00552240 (46) [back to overview]Number of Participants With HIV Viral Load < 50 Copies/ml at Week 36 of Treatment
NCT00552240 (46) [back to overview]Number of Participants With HIV Viral Load < 50 Copies/ml at Week 4 of Treatment
NCT00552240 (46) [back to overview]Number of Participants With HIV Viral Load < 50 Copies/ml at Week 48
NCT00552240 (46) [back to overview]Number of Participants With HIV Viral Load < 50 Copies/ml at Week 48 of Treatment
NCT00552240 (46) [back to overview]Number of Participants With HIV Viral Load < 50 Copies/ml at Week 8 of Treatment
NCT00552240 (46) [back to overview]Number of Participants With Loss of Virologic Response Following Confirmed Virologic Response
NCT00552240 (46) [back to overview]Number of Participants With Virologic Response (VR)
NCT00552240 (46) [back to overview]Number of Participants With Virologic Response According to the Time to Loss of Virologic Response (TLOVR) Algorithm
NCT00552240 (46) [back to overview]Number of Participants With Virologic Success (FDA Definition)
NCT00552240 (46) [back to overview]Number of Patients With Virologic Rebound to >400 Copies/ml
NCT00552240 (46) [back to overview]Proportion of Patients With DAIDS Grade >= 2 Laboratory Abnormalities
NCT00552240 (46) [back to overview]Number of Participants With HIV Viral Load < 50 Copies/ml at Week 6 of Treatment
NCT00552240 (46) [back to overview]AIDS Progression and Death: Number of Patients With a Treatment-emergent AIDS Defining Illness or an AIDS-defining Illness Leading to Death
NCT00552240 (46) [back to overview]Change in CD4+ Cell Count From Baseline to Week 12.
NCT00552240 (46) [back to overview]Change in CD4+ Cell Count From Baseline to Week 2.
NCT00552240 (46) [back to overview]Change in CD4+ Cell Count From Baseline to Week 24.
NCT00552240 (46) [back to overview]Change in CD4+ Cell Count From Baseline to Week 36.
NCT00552240 (46) [back to overview]Change in CD4+ Cell Count From Baseline to Week 4.
NCT00552240 (46) [back to overview]Change in CD4+ Cell Count From Baseline to Week 48.
NCT00552240 (46) [back to overview]Change in CD4+ Cell Count From Baseline to Week 6.
NCT00552240 (46) [back to overview]Change in CD4+ Cell Count From Baseline to Week 8.
NCT00552240 (46) [back to overview]Change in Fasting High Density Lipoprotein (HDL) Cholesterol Level
NCT00571961 (1) [back to overview]Buprenorphine Area Under the Curve With LPV/r (ng/mL*hr)
NCT00608569 (8) [back to overview]Confirmed Virologic Failure at or Prior to Week 24
NCT00608569 (8) [back to overview]Time to First Grade 3 or 4 Lab Event
NCT00608569 (8) [back to overview]Adherence to Second Line HAART Regimen
NCT00608569 (8) [back to overview]Confirmed Virologic Failure at or Prior to Week 48
NCT00608569 (8) [back to overview]CD4 Count at Follow-up Visits
NCT00608569 (8) [back to overview]CD8 Count at Follow-up Visits
NCT00608569 (8) [back to overview]Time to First Grade 3 or 4 Sign or Symptom
NCT00608569 (8) [back to overview]Time to First Grade 3 or 4 Lab or Sign/Symptom Event
NCT00614991 (7) [back to overview]AUC: Steady-state Plasma Amprenavir (APV) Pharmacokinetics (PK) Following Administration of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent Raltegravir (RTG) 400mg BID.
NCT00614991 (7) [back to overview]CL/F: Steady-state Plasma Amprenavir (APV) Pharmacokinetics (PK) Following Administration of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent Raltegravir (RTG) 400mg BID.
NCT00614991 (7) [back to overview]CL/F: Steady-state Plasma Amprenavir (APV) Pharmacokinetics (PK) Following Administration of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent Raltegravir (RTG) 400mg BID.
NCT00614991 (7) [back to overview]Cmin/Cmax: Steady-state Plasma Amprenavir (APV) Pharmacokinetics (PK) Following Admin of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent Raltegravir (RTG) 400mg BID.
NCT00614991 (7) [back to overview]Number of Participants Who Experienced Adverse Events
NCT00614991 (7) [back to overview]Cmin/Cmax: Steady-state Plasma RTG PK Following Admin of FPV 1400mg BID, FPV 700mg/RTV 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent RTG 400mg BID.
NCT00614991 (7) [back to overview]AUC: Steady-state Plasma Amprenavir (APV) Pharmacokinetics (PK) Following Admin of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent Raltegravir (RTG) 400mg BID.
NCT00623597 (21) [back to overview]Change In Hematuria, Glycosuria And Proteinuria From Baseline
NCT00623597 (21) [back to overview]Change In Hemoglobin, Total Protein And Total Albumin From Baseline
NCT00623597 (21) [back to overview]Change In Red Blood Cell (RBC) Counts From Baseline
NCT00623597 (21) [back to overview]Plasma Trough Concentrations (Ctrough) for Ritonavir
NCT00623597 (21) [back to overview]Area Under the Plasma Concentration-time Curve Over the Time Interval From Zero to Twelve Hours (AUC0-12h) for Ritonavir
NCT00623597 (21) [back to overview]Change From Baseline in Cluster Differentiation Antigen 8 (CD8) Lymphocyte Count
NCT00623597 (21) [back to overview]Number of Participants With >1 Log Decrease From Baseline in Human Immunodeficiency Virus (HIV) -Ribonucleic Acid (RNA )
NCT00623597 (21) [back to overview]Maximum Observed Concentration (Cmax) for Saquinavir and Ritonavir
NCT00623597 (21) [back to overview]Incidence of Adverse Events (AE) and Serious Adverse Events (SAE)
NCT00623597 (21) [back to overview]Change In Total Bilirubin, Creatinine, Uric Acid From Baseline
NCT00623597 (21) [back to overview]Change In White Blood Cell (WBC), Platelet, Basophil, Lymphocyte, Monocyte, Neutrophil And Eosinophil Cell Counts From Baseline
NCT00623597 (21) [back to overview]Number of Participants With Human Immunodeficiency Virus (HIV) -Ribonucleic Acid (RNA) <400 Copies/mL
NCT00623597 (21) [back to overview]Number of Participants With Human Immunodeficiency Virus (HIV) -Ribonucleic Acid (RNA) <50 Copies/mL
NCT00623597 (21) [back to overview]Number of Participants With Virological Failure
NCT00623597 (21) [back to overview]Change In Hematocrit From Baseline
NCT00623597 (21) [back to overview]Change In Creatine Kinase (CK), Serum Glutamic Oxaloacetic Transaminase (SGOT), Alkaline Phosphatase (ALP), Serum Glutamic-Pyruvic Transaminase (SGPT), Gamma-Glutamyl Transferase (GGT) Counts From Baseline
NCT00623597 (21) [back to overview]Change In Blood Urea Nitrogen (BUN), Low Density Lipoprotein (LDL) Cholesterol, High Density Lipoprotein (HDL Cholesterol), Triglycerides, Calcium, Potassium, Sodium, Chloride, Phosphate, Fasting Glucose From Baseline
NCT00623597 (21) [back to overview]Change From Baseline in Mean Human Immunodeficiency Virus Viral Load
NCT00623597 (21) [back to overview]Area Under the Plasma Concentration-time Curve Over the Time Interval From Zero to Twelve Hours (AUC0-12h) for Saquinavir
NCT00623597 (21) [back to overview]Change From Baseline in Cluster Differentiation Antigen 4 (CD4) Lymphocyte Count
NCT00623597 (21) [back to overview]Plasma Trough Concentrations (Ctrough) for Saquinavir
NCT00630734 (10) [back to overview]Darunavir Area Under the Plasma Concentration-time Curve (AUC) Over the Dosing Interval
NCT00630734 (10) [back to overview]Darunavir Maximum Plasma Concentration (Cmax)
NCT00630734 (10) [back to overview]Pravastatin + Darunavir/Ritonavir: Pravastatin Area Under the Plasma Concentration-time Curve (AUC) Over the Dosing Interval
NCT00630734 (10) [back to overview]Ritonavir Maximum Plasma Concentration (Cmax)
NCT00630734 (10) [back to overview]Relative Change in Pravastatin Maximum Plasma Concentration (Cmax)
NCT00630734 (10) [back to overview]Pravastatin Alone: Pravastatin Area Under the Plasma Concentration-time Curve (AUC) Over the Dosing Interval
NCT00630734 (10) [back to overview]Ritonavir Area Under the Plasma Concentration-time Curve (AUC) Over the Dosing Interval
NCT00630734 (10) [back to overview]Relative Change in Pravastatin Area Under the Plasma Concentration-time Curve (AUC) Over the Dosing Interval
NCT00630734 (10) [back to overview]Pravastatin + Darunavir/Ritonavir: Pravastatin Maximum Plasma Concentration (Cmax)
NCT00630734 (10) [back to overview]Pravastatin Alone: Pravastatin Maximum Plasma Concentration (Cmax)
NCT00632970 (1) [back to overview]Absolute Change in CD4 Cell Counts
NCT00646776 (26) [back to overview]Cmin of RTV
NCT00646776 (26) [back to overview]Maximum Plasma Concentration (Cmax) of RIB
NCT00646776 (26) [back to overview]Minimum Plasma Concentration (Cmin) of RIB
NCT00646776 (26) [back to overview]Number of Participants With Clinically Significant Vital Signs or Physical Examination Findings
NCT00646776 (26) [back to overview]Number of Participants With Identified Electrocardiogram (ECG) Abnormalities
NCT00646776 (26) [back to overview]T-half of RTV
NCT00646776 (26) [back to overview]Terminal Elimination Half-life (T-half) of ATV
NCT00646776 (26) [back to overview]Number of Participants Who Died, Experienced Other Serious Adverse Events (SAEs), Experienced Adverse Events (AEs) and Experienced Events Leading to Discontinuation.
NCT00646776 (26) [back to overview]Number of Participants With Marked Abnormalities (MAs) in Hematology: Hemoglobin, Hematocrit, Platelet Count and Leukocytes
NCT00646776 (26) [back to overview]Number of Participants With MAs in Hematology: Neutrophils + Bands (Absolute), Lymphocytes (Absolute), Monocytes (Absolute), Basophils (Absolute) and Eosinophils (Absolute)
NCT00646776 (26) [back to overview]Time to Reach Maximum Observed Plasma Concentration (Tmax) of ATV
NCT00646776 (26) [back to overview]Tmax of RTV
NCT00646776 (26) [back to overview]Number of Participants With MAs in Serum Chemistry: Alkaline Phosphatase (ALP),Aspartate Aminotransferase (AST),Alanine Aminotransferase (ALT),Bilirubin (Total),Bilirubin (Direct),Blood Urea Nitrogen (BUN),Creatinine,Sodium (Serum),Potassium (Serum)
NCT00646776 (26) [back to overview]Number of Participants With MAs in Serum Chemistry: Chloride (Serum), Calcium (Total), Protein (Total), Bicarbonate, Phosphorous (Inorganic)
NCT00646776 (26) [back to overview]Number of Participants With MAs in Serum Chemistry: Glucose (Fasting Serum), Albumin, Creatine Kinase, Uric Acid, Lactate Dehydrogenase (LDH)
NCT00646776 (26) [back to overview]Number of Participants With MAs in Urinalysis
NCT00646776 (26) [back to overview]AUC(TAU) for ATV
NCT00646776 (26) [back to overview]AUC(TAU) for RTV
NCT00646776 (26) [back to overview]AUC24avg for 25-O-Desacetyl-RIB
NCT00646776 (26) [back to overview]Average Area Under the Plasma Concentration-time Curve for 24 Hours (AUC24avg) for Rifabutin (RIB)
NCT00646776 (26) [back to overview]Cmax of 25-O-Desacetylrifabutin (25-O-Desacetyl-RIB)
NCT00646776 (26) [back to overview]Cmax of ATV
NCT00646776 (26) [back to overview]Total Area Under the Plasma Concentration-time Curve (AUCtot)
NCT00646776 (26) [back to overview]Cmax of RTV
NCT00646776 (26) [back to overview]Cmin of 25-O-Desacetyl-RIB
NCT00646776 (26) [back to overview]Cmin of ATV
NCT00654147 (6) [back to overview]Weeks to HIV-1 RNA <200 Copies/ml
NCT00654147 (6) [back to overview]Change From Baseline CD4+ and CD8+ Cell Counts
NCT00654147 (6) [back to overview]Time to Virologic Failure
NCT00654147 (6) [back to overview]Time to Confirmed Virologic Failure
NCT00654147 (6) [back to overview]Study Medication Toxicity-related Discontinuation .
NCT00654147 (6) [back to overview]Study Medication Tolerability
NCT00700115 (2) [back to overview]Plasma Viral Loads (HIV-1 RNA PCR)
NCT00700115 (2) [back to overview]To Compare Plasma Triglyceride Levels at 48 Weeks Between LPV/r + RAL and Standard HAART Treated Subjects
NCT00711009 (82) [back to overview]Mean Change From Baseline in Triglycerides (Micromoles/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Total Protein (Grams/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Total Bilirubin (Micromoles/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Temperature (°F)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Soluble Tumor Necrosis Factor Receptor-2 (Picograms/Milliliter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Soluble Tumor Necrosis Factor Receptor-1 (Picograms/Milliliter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Sodium (Micromoles/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Sitting Systolic Blood Pressure (mm Hg)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Sitting Diastolic Blood Pressure (mm Hg)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Red Blood Cell Count (x 10^12/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Potassium (Micromoles/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Platelet Count (x 10^9/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Neutrophils (x 10^9/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Monocytes (x 10^9/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Mid-Thigh Measurement (cm)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Mid-Arm Measurement (cm)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Magnesium (Millimoles/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Lymphocytes (x 10^9/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Low Density Lipoprotein (LDL): High Density Lipoprotein (HDL) Ratio (Ratio)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Low Density Lipoprotein (LDL) (Micromoles/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Lipase (Units/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Leptin (Nanograms/Milliliter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Lactate Dehydrogenase (Units/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Lactate (Millimoles/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Interleukin-6 (Nanograms/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Insulin (Picomoles/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Inorganic Phosphate (Micromoles/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Hips Measurement (cm)
NCT00711009 (82) [back to overview]Mean Change From Baseline in High Density Lipoprotein Cholesterol (HDL) (Micromoles/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Hematocrit (Fraction)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Fasting Glucose (Millimoles/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Eosinophils (x 10^9/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Upper Extremity Total Mass (Grams)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Upper Extremity Lean Mass (Grams)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Upper Extremity Fat (Grams)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Trunk Mass (Grams)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Trunk Lean Mass (Grams)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Trunk Fat (Grams)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Total Body Mass (Grams)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Total Body Lean Mass (Grams)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Lower Extremity Total Mass (Grams)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Lower Extremity Fat (Grams)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Bone Mineral Density (Grams/cm^2)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Bone Mineral Content (Grams)
NCT00711009 (82) [back to overview]Mean Change From Baseline in DEXA Scan of Lower Extremity Lean Mass (Grams)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Creatinine (Micromoles/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Creatine Phosphokinase (Units/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Cholesterol (Micromoles/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Chloride (Micromoles/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Chest Measurement (cm)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Calculated Creatinine Clearance (Milliliters/Second)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Calcium (Micromoles/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Blood Urea Nitrogen (Micromoles/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Bicarbonate (Micromoles/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Basophils (x 10^9/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Aspartate Aminotransferase (Units/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Alkaline Phosphatase (Units/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Albumin (Grams/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Alanine Aminotransferase (Units/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Adiponectin (Micrograms/Milliliter)
NCT00711009 (82) [back to overview]Change From Baseline on Physical Component Score of the Medical Outcomes Study HIV Health Survey
NCT00711009 (82) [back to overview]Change From Baseline on Mental Component of Medical Outcomes Study HIV Health Survey
NCT00711009 (82) [back to overview]Mean Change From Baseline in Uric Acid (Micromoles/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Hemoglobin (Grams/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Sitting Heart Rate (Beats Per Minute)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Total Body Fat (Grams)
NCT00711009 (82) [back to overview]Primary Outcome: Percentage of Participants With Potentially Clinically Significant Laboratory Values
NCT00711009 (82) [back to overview]Percentage of Participants With Moderate or Severe Treatment-emergent, Drug-related Adverse Events
NCT00711009 (82) [back to overview]Percentage of Participants Responding (Plasma HIV-1 RNA Levels Below 40 Copies/Milliliter [mL]) at Each Visit Based on the FDA Time to Loss of Virologic Response (TLOVR) Algorithm
NCT00711009 (82) [back to overview]Number of Participants Who Developed Resistance to Each Drug in the Study Regimen, as Defined by the International AIDS Society-USA (IAS-USA) Panel.
NCT00711009 (82) [back to overview]Mean Change in CD4+ T-Cell Counts From Baseline to Each Visit
NCT00711009 (82) [back to overview]Time to Loss of Virologic Response - Percentage of Participants Still Categorized as Responders at Day 672
NCT00711009 (82) [back to overview]Score on Side Effects Scale of Treatment Satisfaction Questionnaire for Medication
NCT00711009 (82) [back to overview]Score on Global Satisfaction Scale of Treatment Satisfaction Questionnaire for Medication
NCT00711009 (82) [back to overview]Score on Effectiveness Scale of Treatment Satisfaction Questionnaire for Medication (TSQM)
NCT00711009 (82) [back to overview]Percentage of Participants Responding (Plasma HIV-1 Ribonucleic Acid [RNA] Levels Less Than 40 Copies/Milliliter [mL]) at Week 48 Based on the Food and Drug Administration (FDA) Time to Loss of Virologic Response (TLOVR) Algorithm
NCT00711009 (82) [back to overview]Number of Participants Who Developed Resistance, Defined Conservatively, to Lopinavir
NCT00711009 (82) [back to overview]Mean Change From Baseline in White Blood Cell Count (x 10^9/Liter)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Weight (kg)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Waist Measurement (cm)
NCT00711009 (82) [back to overview]Mean Change From Baseline in Urine Specific Gravity
NCT00711009 (82) [back to overview]Mean Change From Baseline in Urine pH
NCT00717067 (13) [back to overview]Half-life (t1/2)
NCT00717067 (13) [back to overview]Hemodialysis Clearance of Maraviroc (MVC) in Subjects With End Stage Renal Disease (ESRD) Undergoing Hemodialysis: CLdD
NCT00717067 (13) [back to overview]Maximum Observed Plasma Concentration (Cmax)
NCT00717067 (13) [back to overview]Renal Clearance (CLR) in Subjects With Normal, Mild, Moderate and Severe Renal Function
NCT00717067 (13) [back to overview]Time of First Occurrence (Tmax)
NCT00717067 (13) [back to overview]Safety and Tolerability of Maraviroc in the Absence and Presence of a Potent CYP3A4 Inhibitor in Subjects With Various Degrees of Renal Impairment or Undergoing Hemodialysis: Number of Subjects With Maximum EGC QTC, QTCB and QTCF Intervals
NCT00717067 (13) [back to overview]Safety and Tolerability of Maraviroc in the Absence and Presence of a Potent CYP3A4 Inhibitor in Subjects With Various Degrees of Renal Impairment or Undergoing Hemodialysis: Number of Subjects With Maximum Increase and Decrease in Supine Blood Pressure
NCT00717067 (13) [back to overview]Area Under the Plasma Concentration Time-curve From Zero to the Last Measured Concentration (AUClast)
NCT00717067 (13) [back to overview]Safety and Tolerability of Maraviroc in the Absence and Presence of a Potent CYP3A4 Inhibitor in Subjects With Various Degrees of Renal Impairment or Undergoing Hemodialysis: Number of Subjects With Pulse Rate < 40 and > 120 Beats Per Minute
NCT00717067 (13) [back to overview]Derivation of Renal Clearance in Subjects With Normal, Mild, Moderate and Severe Renal Function: Ae
NCT00717067 (13) [back to overview]Area Under the Time Curve From 0 to Infinity (AUCinf)
NCT00717067 (13) [back to overview]AUCtau
NCT00717067 (13) [back to overview]Plasma Protein Binding
NCT00752856 (4) [back to overview]Compare Late Activated CD4+ T-cell Recovery Rates Between Treatment Regimens From Baseline to Week 48
NCT00752856 (4) [back to overview]To Compare the Phase 1 Viral Decay Rates Between LPV/r + RAL vs. EFV/TDF/FTC Treatment Combinations.
NCT00752856 (4) [back to overview]Viral Suppression Efficacy at 48 Weeks
NCT00752856 (4) [back to overview]Compare Early Activated CD4+ T-cell Recovery Rates From Baseline to Week 4.
NCT00757783 (16) [back to overview]Change From Baseline in Apolipoprotein B in the LE Set at Week 12 and 48.
NCT00757783 (16) [back to overview]Change From Baseline in CD4 Cell Count at Week 12 and 48, Observed Values.
NCT00757783 (16) [back to overview]Change From Baseline in Cluster of Differentiation (CD) 4 Cell Count at Week 12 and 48, Last Observation Carried Forward (LOCF).
NCT00757783 (16) [back to overview]Change From Baseline in Cluster of Differentiation (CD) 4 Percent at Week 12 and 48, Last Observation Carried Forward (LOCF).
NCT00757783 (16) [back to overview]Change From Baseline in Fasting Triglyceride (TG) Levels in the Lipid Evaluable (LE) Set at Week12
NCT00757783 (16) [back to overview]Change From Baseline in Glucose at Week 12 and 48.
NCT00757783 (16) [back to overview]Change From Baseline in High Density Lipoprotein (HDL) in the LE Set at Week 12 and 48.
NCT00757783 (16) [back to overview]Change From Baseline in HIV-1 RNA Viral Load at Week 12 and 48.
NCT00757783 (16) [back to overview]Change From Baseline in Homeostasis Model Assessment-Insulin Resistance (HOMA-IR) at Week 12 and 48.
NCT00757783 (16) [back to overview]Change From Baseline in Insulin at Week 12 and 48.
NCT00757783 (16) [back to overview]Change From Baseline in TC/HDL Ratio in the LE Set at Week 12 and 48.
NCT00757783 (16) [back to overview]Change From Baseline in Total Cholesterol (TC) Levels in the LE Set at Week 12 and 48
NCT00757783 (16) [back to overview]Number of Participants With Antiviral Activity, HIV-1 RNA, Missing Values as Treatment Failure (M=F)
NCT00757783 (16) [back to overview]Change From Baseline in Low Density Lipoprotein (LDL) Direct in the LE Set at Week 12 and 48.
NCT00757783 (16) [back to overview]Antiviral Activity, Human Immunodeficiency Virus Type 1 (HIV-1) RNA.
NCT00757783 (16) [back to overview]Change From Baseline in Apolipoprotein A1 in the LE Set at Week 12 and 48.
NCT00762320 (10) [back to overview]Lopinavir (Lpv) and Ritonavir (Rtv) Cmax at 4 Weeks
NCT00762320 (10) [back to overview]Symptoms Across All Patients
NCT00762320 (10) [back to overview]Lopinavir and Ritonavir AUC on Low Dose Tablet
NCT00762320 (10) [back to overview]Lopinavir (Lpv) and Ritonavir (Rtv) Maximumu Plasma Concentration (CMax) Liquid
NCT00762320 (10) [back to overview]Lopinavir and Ritonavir Area Under the Curve (AUC) Liquid Kaletra
NCT00762320 (10) [back to overview]Viral Load (VL)
NCT00762320 (10) [back to overview]Patient Satisfaction
NCT00762320 (10) [back to overview]Parent Satisfaction
NCT00762320 (10) [back to overview]Lopinavir AUC Ratio of Baseline:Week 4
NCT00762320 (10) [back to overview]Absolute CD4 and CD4 %
NCT00764465 (5) [back to overview]Number of Participants Who Experienced an Adverse Event
NCT00764465 (5) [back to overview]AUC: Steady-state Plasma MVC PK Following Administration of RTV
NCT00764465 (5) [back to overview]Cmin/Cmax: Steady-state Plasma Amprenavir (APV) Pharmacokinetics ( PK) Following Admin of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent (Maraviroc) MVC 300mg BID.
NCT00764465 (5) [back to overview]Cmin/Cmax: Steady-state Plasma MVC PK Following Administration of RTV
NCT00764465 (5) [back to overview]AUC: Steady-state Plasma Amprenavir (APV) Pharmacokinetics ( PK) Following Admin of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent (Maraviroc) MVC 300mg BID.
NCT00768989 (28) [back to overview]Atazanavir Trough Plasma Concentration (Cmin) 12 Hours Postdose
NCT00768989 (28) [back to overview]Atazanavir Cmin Prior to the Morning Dose
NCT00768989 (28) [back to overview]Atazanavir Area Under the Concentration Curve From Time 0 to 24 Hours (AUC [0-24h]) in 1 Dosing Interval
NCT00768989 (28) [back to overview]Atazanavir Area Under the Concentration Curve From Time 0 to 12 Hours (AUC [0-12h]) in 1 Dosing Interval
NCT00768989 (28) [back to overview]Raltegravir Cmin Prior to the Morning Dose
NCT00768989 (28) [back to overview]Raltegravir Terminal Elimination Half Life
NCT00768989 (28) [back to overview]Raltegravir Tmax
NCT00768989 (28) [back to overview]Baseline and Mean Change From Baseline in Total Cholesterol Levels
NCT00768989 (28) [back to overview]Mean Change From Baseline in Absolute Cluster of Differentiation 4 Cell Count
NCT00768989 (28) [back to overview]Mean Change From Baseline in Electrocardiogram Findings
NCT00768989 (28) [back to overview]Atazanavir Time of Maximum Observed Plasma Concentration (Tmax)
NCT00768989 (28) [back to overview]Number of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs), Death as Outcome, AEs Leading to Discontinuation, SAEs Leading to Discontinuation
NCT00768989 (28) [back to overview]Number of Participants With Blood Chemistry Laboratory Test Results With Worst Toxicity of Grades 1 to 4
NCT00768989 (28) [back to overview]Number of Participants With Blood Chemistry Laboratory Test Results With Worst Toxicity of Grades 1 to 4 (Continued)
NCT00768989 (28) [back to overview]Number of Participants With Enzyme and Urine Laboratory Test Results With Worst Toxicity of Grades 1 to 4
NCT00768989 (28) [back to overview]Number of Participants With Hematology Laboratory Test Results With Worst Toxicity of Grades 1 to 4 Among All Treated Participants
NCT00768989 (28) [back to overview]Atazanavir Terminal Elimination Half Life
NCT00768989 (28) [back to overview]Number of Participants With HIV RNA Levels <400 Copies/mL at Week 24
NCT00768989 (28) [back to overview]Atazanavir Maximum Observed Plasma Concentration (Cmax) in 1 Dosing Interval
NCT00768989 (28) [back to overview]Atazanavir Individual Inhibitory Quotient (IQ)
NCT00768989 (28) [back to overview]Number of Nonresponders at Week 8
NCT00768989 (28) [back to overview]Number of Participants With HIV RNA Levels <400 Copies/mL at Week 48
NCT00768989 (28) [back to overview]Number of Participants With HIV RNA Levels <50 Copies/mL at Weeks 48 and 96
NCT00768989 (28) [back to overview]Raltegravir AUC (0-12h) in 1 Dosing Interval
NCT00768989 (28) [back to overview]Raltegravir Cmax in 1 Dosing Interval
NCT00768989 (28) [back to overview]Raltegravir Cmin 12 Hours Postdose
NCT00768989 (28) [back to overview]Mean Change From Baseline in Total Bilirubin Level
NCT00768989 (28) [back to overview]Number of Participants With Human Immunodeficiency Virus (HIV) Ribonucleic Acid (RNA) Level <50 Copies/mL at Week 24
NCT00775606 (6) [back to overview]CD4+ (Cluster of Differentiation 4) T-cell Apoptosis
NCT00775606 (6) [back to overview]CD4+ T-cell Change
NCT00775606 (6) [back to overview]Activated and Regulatory CD4+ and CD8+ T-cell Frequencies
NCT00775606 (6) [back to overview]Activation and Proliferation of CD4+ and CD8+ T-cell Frequencies
NCT00775606 (6) [back to overview]Naive, Central Memory, Effector Memory, and T Reg CD4+ T-cell Frequency
NCT00775606 (6) [back to overview]Naive, Central Memory, Effector Memory, and T Reg CD4+ T-cell Frequency
NCT00802074 (7) [back to overview]CL/F: Fasting Steady-state Plasma Amprenavir (APV) Pharmacokinetics (PK) Following Administration of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent Raltegravir (RTG) 400mg BID.
NCT00802074 (7) [back to overview]Cmin/Cmax: Steady-state Plasma RTG PK Following Admin of RTG 400mg BID Alone and Following Co-administration With FPV 1400mg BID, FPV 700mg/RTV 100 mg BID, or FPV 1400mg/RTV 100mg QD
NCT00802074 (7) [back to overview]AUC: Steady-state Plasma RTG PK Following Administration of RTG 400mg BID Alone and Following Co-administration With FPV 1400mg BID, FPV 700mg/RTV 100 mg BID, or FPV 1400mg/RTV 100mg QD
NCT00802074 (7) [back to overview]AUC: Fasting Steady-state Plasma Amprenavir (APV) Pharmacokinetics (PK) Following Administration of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent Raltegravir (RTG) 400mg BID.
NCT00802074 (7) [back to overview]Cmin/Cmax: Fasting Steady-state Plasma Amprenavir (APV) Pharmacokinetics (PK) Following Admin of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent Raltegravir (RTG) 400mg BID.
NCT00802074 (7) [back to overview]Number of Participants Who Experienced Adverse Events
NCT00802074 (7) [back to overview]CL/F: Steady-state Plasma RTG PK Following Administration of RTG 400mg BID Alone and Following Co-administration With FPV 1400mg BID, FPV 700mg/RTV 100 mg BID, or FPV 1400mg/RTV 100mg QD
NCT00810108 (1) [back to overview]Lopinavir Area Under the Curve (AUC)
NCT00811954 (19) [back to overview]Change in Waist Circumference From Baseline
NCT00811954 (19) [back to overview]Change in Waist:Height Ratio From Baseline
NCT00811954 (19) [back to overview]Self-reported Adherence
NCT00811954 (19) [back to overview]Incidence of Death or AIDS Defining Events (CDC Category C)
NCT00811954 (19) [back to overview]Incidence of Targeted Serious Non-AIDS Defining Events (Renal Failure, Liver Disease, Serious Metabolic Disorder, and CVD)
NCT00811954 (19) [back to overview]Presence of Mutations Associated With ATV/RTV or DRV/RTV Resistance
NCT00811954 (19) [back to overview]Presence of Mutations Associated With INI Resistance
NCT00811954 (19) [back to overview]Presence of Mutations Associated With NRTI Resistance
NCT00811954 (19) [back to overview]Change in Framingham 10-year Risk of MI or Coronary Death From Baseline
NCT00811954 (19) [back to overview]CD4+ T-cell Count
NCT00811954 (19) [back to overview]CD4+ T-cell Count Changes From Baseline
NCT00811954 (19) [back to overview]Change in Fasting HDL Cholesterol Level From Baseline
NCT00811954 (19) [back to overview]Change in Fasting Plasma Glucose Level From Baseline
NCT00811954 (19) [back to overview]Change in Fasting Total Cholesterol Level From Baseline
NCT00811954 (19) [back to overview]Change in Fasting Triglycerides Level From Baseline
NCT00811954 (19) [back to overview]Cumulative Incidence of Discontinuation of the RAL or PI Component of Randomized Treatment for Toxicity by Week 96
NCT00811954 (19) [back to overview]Cumulative Incidence of First Adverse Event by Week 96
NCT00811954 (19) [back to overview]Cumulative Probability of First Virologic Failure by Week 96
NCT00811954 (19) [back to overview]Cumulative Probability of Time to Loss of Virologic Response (TLOVR) by Week 96
NCT00827112 (16) [back to overview]Time to Loss of Virological Response (TLOVR)
NCT00827112 (16) [back to overview]Change From Baseline in Plasma log10 Viral Load at Weeks 16, 24, 48 and 96
NCT00827112 (16) [back to overview]Number of Participants With HIV-1 RNA Tropism Status Using Trofile Assay
NCT00827112 (16) [back to overview]Percentage of Participants With Less Than 400 Copies/mL of HIV-1 RNA
NCT00827112 (16) [back to overview]Percentage of Participants With Less Than 50 Copies/mL of HIV-1 RNA
NCT00827112 (16) [back to overview]Number of Participants With Phenotypic Resistance
NCT00827112 (16) [back to overview]Time-Averaged Difference (TAD) in log10 Viral Load
NCT00827112 (16) [back to overview]Average Observed Plasma Concentration (Cavg) of Maraviroc
NCT00827112 (16) [back to overview]HIV-1 RNA Levels at Baseline
NCT00827112 (16) [back to overview]Number of Participants With Genotypic Resistance
NCT00827112 (16) [back to overview]Maximum Observed Plasma Concentration (Cmax) of Maraviroc
NCT00827112 (16) [back to overview]Percentage of Participants With Plasma Human Immuno Deficiency Virus-1 Ribonucleic Acid (HIV-1 RNA) Levels Less Than 50 Copies/Milliliter (mL)
NCT00827112 (16) [back to overview]Minimum Observed Plasma Concentration (Cmin) of Maraviroc
NCT00827112 (16) [back to overview]Change From Baseline in Cluster of Differentiation 4+T Lymphocyte (CD4) Cell Counts at Weeks 16, 24, 48 and 96
NCT00827112 (16) [back to overview]Change From Baseline in Cluster of Differentiation 8+T Lymphocyte (CD8) Cell Count at Weeks 16, 24, 48 and 96
NCT00827112 (16) [back to overview]Change From Baseline in HIV-1 RNA Levels of First 15 Participants at Days 4, 7, 10 and 14
NCT00830804 (17) [back to overview]Proportion of Participants With Virologic Failure or Off Study Treatment Regimen or Death at or Prior to Week 24
NCT00830804 (17) [back to overview]Changes in Fasting Total Cholesterol, High-density Lipoprotein and Triglyceride at Week 24
NCT00830804 (17) [back to overview]Changes in Fasting Total Cholesterol, High-density Lipoprotein and Triglyceride at Week 48
NCT00830804 (17) [back to overview]Number of Participants With Pretreatment Drug Resistance
NCT00830804 (17) [back to overview]Proportion of Participants With Plasma HIV-1 RNA <50 Copies/ml or <200 Copies/ml at Week 48
NCT00830804 (17) [back to overview]Proportion of Participants With Plasma HIV-1 RNA < 50 Copies/ml or <200 Copies/ml at Week 24
NCT00830804 (17) [back to overview]Change in CD4 Count at Week 48
NCT00830804 (17) [back to overview]Change in Fasting Low-density Lipoprotein at Week 24
NCT00830804 (17) [back to overview]Change in Fasting Low-density Lipoprotein at Week 48
NCT00830804 (17) [back to overview]Change in Plasma HIV-1 RNA From Baseline to Week 1
NCT00830804 (17) [back to overview]Number of Participants With Integrase Drug Resistance at Virologic Failure
NCT00830804 (17) [back to overview]Number of Participants With Perfect Overall Adherence by Self Report
NCT00830804 (17) [back to overview]Plasma Trough Concentration of Darunavir
NCT00830804 (17) [back to overview]Plasma Trough Concentration of Raltegravir
NCT00830804 (17) [back to overview]Proportion of Participants Who Experienced Signs/Symptoms or Laboratory Toxicities Grade 3 or Higher, or of Any Grade Which Led to a Permanent Change or Discontinuation of Study Treatment
NCT00830804 (17) [back to overview]Proportion of Participants With Virologic Failure After Initiating RAL Plus DRV/RTV at or Prior to Week 24
NCT00830804 (17) [back to overview]Number of Participants With Protease Drug Resistance at Virologic Failure
NCT00833482 (14) [back to overview]AUC(TAU) of Ritonavir, Administered As Atazanavir/Ritonavir With and Without Voriconazole, in EM Participants
NCT00833482 (14) [back to overview]Area Under the Plasma Concentration-time Curve in 1 Dosing Interval [AUC(TAU)] of Atazanavir Administered as Atazanavir/Ritonavir With and Without Voriconazole, in EM Participants
NCT00833482 (14) [back to overview]Tmax of Ritonavir, Administered As Atazanavir/Ritonavir With and Without Voriconazole, in EM Participants
NCT00833482 (14) [back to overview]Number of Participants With Marked Abnormalities in Hematology Laboratory Test and Urinalysis Results
NCT00833482 (14) [back to overview]Number of Participants With Marked Abnormalities in Serum Chemistry Test Results
NCT00833482 (14) [back to overview]Tmax of Voriconazole, Administered With and Without Atazanavir/Ritonavir, in EM Participants
NCT00833482 (14) [back to overview]Time to Maximum Concentration (Tmax) of Atazanavir, Administered as Atazanavir/Ritonavir With and Without Voriconazole, in EM Participants
NCT00833482 (14) [back to overview]AUC(TAU)of Voriconazole, Administered With and Without Atazanavir/Ritonavir, in EM Participants
NCT00833482 (14) [back to overview]Maximum Observed Plasma Concentration (Cmax) and Minimum Observed Plasma Concentration (Cmin) of Atazanavir, Administered as Atazanavir/Ritonavir With and Without Voriconazole, in Participants Who Are Extensive Metabolizers (EM)
NCT00833482 (14) [back to overview]Number of Participants With Abnormalities in Vital Signs
NCT00833482 (14) [back to overview]Number of Participants With Death as Outcome, Serious Adverse Events (SAEs), Adverse Events (AEs) Leading to Discontinuation, and Any AE
NCT00833482 (14) [back to overview]Number of Participants With Investigator-identified Abnormalities in Electrocardiogram Results Not Present Prior to Administration of Study Drug and Considered Not Relevant and Not AEs by Investigator
NCT00833482 (14) [back to overview]Cmax and Cmin of Voriconazole, Administered With and Without Atazanavir/Ritonavir, in EM Participants
NCT00833482 (14) [back to overview]Cmax and Cmin of Ritonavir, Administered As Atazanavir/Ritonavir With and Without Voriconazole, in EM Participants
NCT00838162 (9) [back to overview]Fluctuation Index of TMC310911
NCT00838162 (9) [back to overview]Average Steady-state Plasma Concentration (Css,av) of TMC310911
NCT00838162 (9) [back to overview]Time to Reach the Maximum Plasma Concentration (Tmax) of TMC310911
NCT00838162 (9) [back to overview]Predose Plasma Concentration (C0h) of TMC310911
NCT00838162 (9) [back to overview]Number of Participants With Virologic Response at Any Timepoint During the 14-day Treatment Period
NCT00838162 (9) [back to overview]Mean Changes From Baseline in Plasma log10 Human Immunodeficiency Virus Type 1 Ribonucleic Acid (HIV-1 RNA)
NCT00838162 (9) [back to overview]Mean Changes From Baseline in CD4+ Cell Count
NCT00838162 (9) [back to overview]Maximum Plasma Concentration (Cmax) of TMC310911
NCT00838162 (9) [back to overview]Area Under the Plasma Concentration-time Curve (AUC12) From the Time of Administration of TMC310911 up to 12 Hours After Dosing
NCT00851799 (27) [back to overview]Percent Change in Bone Mineral Density (BMD) of the Hip From Study Entry to Week 96
NCT00851799 (27) [back to overview]Percent Change in Visceral Abdominal Fat (VAT) From Study Entry to Week 96
NCT00851799 (27) [back to overview]Fold Change in High Sensitivity C-reactive Protein (hsCRP) From Study Entry to Weeks 48 and 96
NCT00851799 (27) [back to overview]Fold Change in Interleukin-6 (IL-6) From Study Entry to Weeks 48 and 96
NCT00851799 (27) [back to overview]Fold Change in Percent Expression of CD38+HLADR+ on CD4+ (Percent) From Study Entry to Weeks 24 and 96
NCT00851799 (27) [back to overview]Change in CD4+ T-cell Count From Study Entry to Weeks 24, 48, 96 and 144
NCT00851799 (27) [back to overview]Fold Change in Percent Expression of CD38+HLADR+ on CD8+ (Percent) From Study Entry to Weeks 24 and 96
NCT00851799 (27) [back to overview]Fold Change in Soluble CD14 From Study Entry to Weeks 48 and 96
NCT00851799 (27) [back to overview]Annual Rate of Change in Right Common Carotid Artery Intima-media Thickness (CIMT)
NCT00851799 (27) [back to overview]Change in Brachial Artery (BA) Flow Mediated Dilation (FMD) From Study Entry to Week 24
NCT00851799 (27) [back to overview]Fold Change in Soluble CD163 From Study Entry to Weeks 48 and 96
NCT00851799 (27) [back to overview]Percent Change in Bone Mineral Density (BMD) of the Lumber Spine From Study Entry to Week 96
NCT00851799 (27) [back to overview]Percent Change in Bone Mineral Density (BMD) of the Total Body From Study Entry to Week 96
NCT00851799 (27) [back to overview]Percent Change in Lean Mass From Study Entry to Week 96
NCT00851799 (27) [back to overview]Percent Change in Subcutaneous Abdominal Fat (SAT) From Study Entry to Week 96
NCT00851799 (27) [back to overview]Percent Change in Total Limb Fat From Study Entry to Week 96
NCT00851799 (27) [back to overview]Percent Change in Trunk Fat From Study Entry to Week 96
NCT00851799 (27) [back to overview]Change in Fasting Calculated Low-density Lipoprotein Cholesterol (LDL-C) From Study Entry to Weeks 4, 24, 48 and 96
NCT00851799 (27) [back to overview]CD4+ T-cell Count at Study Entry and Weeks 24, 48, 96 and 144
NCT00851799 (27) [back to overview]Change in Absolute Flow Mediated Dilation (FMD) From Study Entry to Weeks 4, 24 and 48
NCT00851799 (27) [back to overview]Change in Brachial Artery Flow Mediated Dilation (FMD) From Study Entry to Weeks 4 and 48
NCT00851799 (27) [back to overview]Fold Change in D-dimer From Study Entry to Weeks 48 and 96
NCT00851799 (27) [back to overview]Change in Fasting Glucose Level From Study Entry to Weeks 4, 24, 48 and 96
NCT00851799 (27) [back to overview]Change in Fasting High-density Lipoprotein Cholesterol (HDL-C) From Study Entry to Weeks 4, 24, 48 and 96
NCT00851799 (27) [back to overview]Change in Fasting Insulin Level From Study Entry to Weeks 4, 24, 48 and 96
NCT00851799 (27) [back to overview]Change in Fasting Total Cholesterol (TC) From Study Entry to Weeks 4, 24, 48 and 96
NCT00851799 (27) [back to overview]Change in Fasting Triglyceride (TG) From Study Entry to Weeks 4, 24, 48 and 96
NCT00855335 (15) [back to overview]Area Under the Plasma Concentration-Time Curve From Time of Administration to 24 Hours Post-dose (AUC0-24h)
NCT00855335 (15) [back to overview]Maximum Plasma Concentration (Cmax)
NCT00855335 (15) [back to overview]Mean Change From Baseline in CD4+ Cell Count
NCT00855335 (15) [back to overview]Number of Participants With Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA) Plasma Viral Load (<) 50 Copies/Milliliter (mL)
NCT00855335 (15) [back to overview]Predose (Trough) Plasma Concentration (C0h)
NCT00855335 (15) [back to overview]Time to Reach the Maximum Plasma Concentration (Tmax)
NCT00855335 (15) [back to overview]Plasma Concentration of Drug in the Cord Plasma and Maternal Plasma Samples Collected at the Time of Delivery
NCT00855335 (15) [back to overview]Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT00855335 (15) [back to overview]Mean Change From Baseline in CD4+ Cell Count
NCT00855335 (15) [back to overview]Area Under the Plasma Concentration-Time Curve From Time of Administration to 12 Hours Post-dose (AUC0-12h)
NCT00855335 (15) [back to overview]Number of Infants With Human Immunodeficiency Virus (HIV) Positive Test Result
NCT00855335 (15) [back to overview]Mean Change From Baseline in Log10 Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA) Viral Load Value
NCT00855335 (15) [back to overview]Mean Change From Baseline in Log10 Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA) Viral Load Value
NCT00855335 (15) [back to overview]Minimum Plasma Concentration (Cmin)
NCT00855335 (15) [back to overview]Number of Participants With Resistance at Virological Failure
NCT00855413 (38) [back to overview]Adverse Events Possibly or Definitely Related to Study Treatment Through Week 48
NCT00855413 (38) [back to overview]Overall Neurocognitive Impairment Score at Week 2 or 4
NCT00855413 (38) [back to overview]Overall Neurocognitive Impairment at Week 48
NCT00855413 (38) [back to overview]Overall Neurocognitive Impairment at Week 24
NCT00855413 (38) [back to overview]Number of Participants With Virologic Response
NCT00855413 (38) [back to overview]Number of Participants With Virologic Response
NCT00855413 (38) [back to overview]Maximum Ritonavir Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure
NCT00855413 (38) [back to overview]Number of Participants With Neurocognitive Impairment at Week 48
NCT00855413 (38) [back to overview]Number of Participants With Neurocognitive Impairment at Week 24
NCT00855413 (38) [back to overview]Number of Participants With Neurocognitive Impairment at Baseline
NCT00855413 (38) [back to overview]Number of Participants With HIV RNA Measurement Above the Limits of Detection in Cerebrospinal Fluid
NCT00855413 (38) [back to overview]Number of Participants Who Stopped Study Treatment Due to Adverse Event or Intolerance
NCT00855413 (38) [back to overview]Minimum Ritonavir Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure
NCT00855413 (38) [back to overview]Minimum Ritonavir Exposure Range in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture
NCT00855413 (38) [back to overview]Minimum Ritonavir Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen
NCT00855413 (38) [back to overview]Minimum Etravirine Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure
NCT00855413 (38) [back to overview]Minimum Etravirine Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen
NCT00855413 (38) [back to overview]Minimum Etravirine Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture
NCT00855413 (38) [back to overview]Minimum Darunavir Exposure Range in Semen Among Participants Who Consented to an Optional Collection of Semen
NCT00855413 (38) [back to overview]Minimum Darunavir Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure
NCT00855413 (38) [back to overview]Minimum Darunavir Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture
NCT00855413 (38) [back to overview]Median Time to HIV RNA Suppression to <200 Copies/mL
NCT00855413 (38) [back to overview]Median Change in CD4 Cell Count From Week 0 to Week 48.
NCT00855413 (38) [back to overview]Median Change in CD4 Cell Count From Week 0 to Week 24.
NCT00855413 (38) [back to overview]Maximum Ritonavir Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen
NCT00855413 (38) [back to overview]Maximum Ritonavir Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture
NCT00855413 (38) [back to overview]Maximum Etravirine Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen
NCT00855413 (38) [back to overview]Maximum Etravirine Exposure in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure
NCT00855413 (38) [back to overview]Maximum Etravirine Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture
NCT00855413 (38) [back to overview]Maximum Darunavir Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure
NCT00855413 (38) [back to overview]Maximum Darunavir Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen
NCT00855413 (38) [back to overview]Maximum Darunavir Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture
NCT00855413 (38) [back to overview]HIV RNA Levels Immediately Prior to Initiating Study Treatment.
NCT00855413 (38) [back to overview]HIV RNA Detection in Semen
NCT00855413 (38) [back to overview]HIV RNA Detection in Ileal Biopsy Specimens
NCT00855413 (38) [back to overview]Correlation of Time to HIV RNA Levels <200 Copies/mL With Improvement in Neurocognitive Functioning From Baseline to Week 24 and 48
NCT00855413 (38) [back to overview]Correlation of HIV RNA Levels in CSF and Drug Levels With Neurocognitive Functioning
NCT00855413 (38) [back to overview]Change in Overall Neurocognitive Impairment From Baseline to Week 24 or 48
NCT00869960 (8) [back to overview]Ritonavir Systemic Exposure During the Follicular Phase (Days 6-10 After Menses)
NCT00869960 (8) [back to overview]Tenofovir Systemic Exposure During the Luteal Phase (Days 20-25 After Menses)
NCT00869960 (8) [back to overview]Tenofovir Systemic Exposure During the Follicular Phase (Days 6-10 After Menses)
NCT00869960 (8) [back to overview]Ritonavir Systemic Exposure During the Luteal Phase (Days 20-25 After Menses)
NCT00869960 (8) [back to overview]Emtricitabine Systemic Exposure During the Luteal Phase (Days 20-25 After Menses)
NCT00869960 (8) [back to overview]Emtricitabine Systemic Exposure During the Follicular Phase (Days 6-10 After Menses)
NCT00869960 (8) [back to overview]Atazanavir Systemic Exposure During the Luteal Phase (Days 20-25 After Menses)
NCT00869960 (8) [back to overview]Atazanavir Systemic Exposure During the Follicular Phase (Days 6-10 After Menses)
NCT00884793 (4) [back to overview]"Average Change in Activated (CD38+HLADR+) CD8+ T Cells in the Ileum"
NCT00884793 (4) [back to overview]Number of Subjects Who Experienced an Increase in CD4% in the Ileum.
NCT00884793 (4) [back to overview]Number of Subjects Who Experienced an Increase in CD4+ T Cells (as a % of All Cells) in the Ileum.
NCT00884793 (4) [back to overview]Number of Subjects Who Had a Decrease in HIV RNA Per Million CD4+ T Cells in the Ileum
NCT00885482 (1) [back to overview]Number of Patients With Virological Failure (Two Consecutive Measures of HIV-RNA Higher Than 50 Copies/mL or a Single Measure Higher Than 1000 Copies/mL) Within 48 Weeks at intention-to.Treat Analysis
NCT00885495 (4) [back to overview]AUC of Rosuvastatin
NCT00885495 (4) [back to overview]Cmax of Rosuvastatin
NCT00885495 (4) [back to overview]To Compare the Change in Low-density Lipoprotein (LDL) Cholesterol With Rosuvastatin Therapy Alone, Darunavir/Ritonavir Therapy Alone and With the Co-administration of Rosuvastatin and Darunavir/Ritonavir.
NCT00885495 (4) [back to overview]To Investigate the Effect of Rosuvastatin on the Steady State Pharmacokinetics of Darunavir/Ritonavir.
NCT00885664 (11) [back to overview]Symptom Score
NCT00885664 (11) [back to overview]IL-7
NCT00885664 (11) [back to overview]IL-8
NCT00885664 (11) [back to overview]INF Gamma
NCT00885664 (11) [back to overview]SF-12 Mental Capacity Score
NCT00885664 (11) [back to overview]SF-12 Physical Capacity Score
NCT00885664 (11) [back to overview]TNF Alpha
NCT00885664 (11) [back to overview]IL-1 Beta
NCT00885664 (11) [back to overview]IL-10
NCT00885664 (11) [back to overview]IL-6
NCT00885664 (11) [back to overview]IL-4
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Etravirine (ETR) (Results for AUC12hr)
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Low-Dose Ritonavir (Rtv): Treatment A: Atazanavir (ATV)/Rtv 300/100 mg (Results for AUC24hr)
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Low-Dose Ritonavir (Rtv): Treatment B: Atazanavir (ATV)/Rtv 400/100 mg (Results for AUC24hr)
NCT00896051 (18) [back to overview]Change From Pre-Baseline in Log10 Viral Load Over Time
NCT00896051 (18) [back to overview]Change From Prebaseline in CD4+ Cell Count Over Time
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Atazanavir (ATV): Treatment A: ATV/Low-Dose Ritonavir (Rtv) 300/100 mg (Results for C0h, Cmin, and Cmax)
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Atazanavir (ATV): Treatment B: ATV/Low-Dose Ritonavir (Rtv) 400/100 mg (Results for C0h, Cmin, and Cmax)
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Etravirine (ETR) (Results for C0h, Cmin, and Cmax)
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Atazanavir (ATV): Treatment B: ATV/Low-Dose Ritonavir (Rtv) 400/100 mg (Results for AUC24hr)
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Low-Dose Ritonavir (Rtv): Treatment B: Atazanavir (ATV)/Rtv 400/100 mg (Results for C0h, Cmin, and Cmax)
NCT00896051 (18) [back to overview]The Percentage of Participants With a Virologic Response (Plasma Viral Load < 50 Copies/mL) at Week 48 Using the Snapshot Analysis Method
NCT00896051 (18) [back to overview]Percentage of Participants With Undetectable Plasma Viral Load (VL) Values (<50 Copies/mL) at Week 48
NCT00896051 (18) [back to overview]The Percentage of Participants With a Virologic Response Using the Non-Completing = Failure (NC=F) Imputation Method
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Low-Dose Ritonavir (Rtv): Treatment A: Atazanavir (ATV)/Rtv 300/100 mg (Results for C0h, Cmin, and Cmax)
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Atazanavir (ATV): Treatment A: ATV/Low-Dose Ritonavir (Rtv) 300/100 mg (Results for AUC24hr)
NCT00896051 (18) [back to overview]The Percentage of Participants With a Virologic Response Using the Time to Loss of Virologic Response (TLOVR) Imputation Method
NCT00896051 (18) [back to overview]Time to Virologic Failure
NCT00896051 (18) [back to overview]Time to Confirmed Virologic Response
NCT00915655 (2) [back to overview]Virological Response [Viral Load <50 Copies/mL, FDA-SNAPSHOT]
NCT00915655 (2) [back to overview]Virological Response[Viral Load <50 Copies/mL, TLOVR]
NCT00919854 (6) [back to overview]Number of Participants With Virological Response (Viral Load Less Than 50 Copies/mL) at Week 24 - Time to Loss of Virologic Response (TLOVR)
NCT00919854 (6) [back to overview]Mean Change From Baseline to Week 24 and Week 48 in CD4+ Percentage
NCT00919854 (6) [back to overview]Mean Change From Baseline to Week 24 and Week 48 in Plasma log10 Viral Load
NCT00919854 (6) [back to overview]Number of Participants With Virological Response (Viral Load Less Than 50 Copies/mL) at Week 48
NCT00919854 (6) [back to overview]Number of Participants With Virological Response (Viral Load Less Than 400 Copies/mL) at Week 24 and Week 48
NCT00919854 (6) [back to overview]Number of Participants With Less Than or Equal to 1 log10 Decrease in Plasma Viral Load at Week 24 and Week 48
NCT00928187 (13) [back to overview]Number of Patients With WHO Stage 3 and 4 HIV Related Events
NCT00928187 (13) [back to overview]Number of Patients With Resistance Mutations
NCT00928187 (13) [back to overview]Number of Patients With Plasma HIV RNA < 50 Copies/mL
NCT00928187 (13) [back to overview]Number of Patients With HIV Plasma Viral Load < 200 Copies/ml
NCT00928187 (13) [back to overview]Number of Patients With HIV Plasma Viral Load < 50 Copies/ml
NCT00928187 (13) [back to overview]Number of Patients Discontinuing Study Treatment
NCT00928187 (13) [back to overview]Gain in CD4 Cells Between Baseline and W48
NCT00928187 (13) [back to overview]Development of Metabolic Syndrome
NCT00928187 (13) [back to overview]Adherence
NCT00928187 (13) [back to overview]Tolerance: Neuropathies (Grade 1 to 4)
NCT00928187 (13) [back to overview]Tolerance: Gastrointestinal Complains
NCT00928187 (13) [back to overview]Tolerance: Equal or Superior to a 25% Reduction in eGFR (Glomerular Filtration Rate)
NCT00928187 (13) [back to overview]Patients With Plasma HIV RNA < 200 Copies/ml
NCT00931463 (5) [back to overview]Participants With Plasma HIV RNA < 200 Copies/mL 48 Weeks After Randomization, Per-protocol Population, VL Less Than or Equal to 100,000 Copies Per mL
NCT00931463 (5) [back to overview]Participants With Plasma HIV RNA < 200 Copies/mL 48 Weeks After Randomization, Per-protocol Population, Non-completer Classed as Failure
NCT00931463 (5) [back to overview]Participants With Plasma HIV RNA < 200 Copies/mL 48 Weeks After Randomization
NCT00931463 (5) [back to overview]Participants With Plasma HIV RNA < 200 Copies/mL 48 Weeks After Randomization, Per-protocol Population
NCT00931463 (5) [back to overview]Participants With Plasma HIV RNA < 200 Copies/mL 48 Weeks After Randomization, Per-protocol Population, Baseline VL >100,000 Copies Per mL
NCT00945282 (38) [back to overview]Change From Baseline to Nadir in Plasma HIV-1 RNA
NCT00945282 (38) [back to overview]Change From Baseline Through Day 8 in Plasma HIV-1 RNA
NCT00945282 (38) [back to overview]Change From Baseline in Hematology Paramaters- Basophils, Eosinophils, Lymphocytes, Monocytes, White Blood Cell Count
NCT00945282 (38) [back to overview]GSK2248761 PK Parameters Following Dose Administration on Day 1: Apparent Clearance (CL/F)
NCT00945282 (38) [back to overview]GSK2248761 PK Parameters Following Dose Administration on Day 7: AUC(0-τ)
NCT00945282 (38) [back to overview]GSK2248761 PK Parameters Following Dose Administration on Day 7: Tmax
NCT00945282 (38) [back to overview]HIV-1 Rate of Decline by Treatment
NCT00945282 (38) [back to overview]Accumulation Ratio for AUC , Cmax, Cτ, and Time Invariance Ratio Following Repeat Administration
NCT00945282 (38) [back to overview]Assessment of the Achievement of Pre-dose GSK2248761 Steady State Concentration Following Repeat Dose Administration on Day 2 Through 7
NCT00945282 (38) [back to overview]Change From Baseline in CD4+ and CD8+ T-lymphocyte Cell Count at Day1 and Day 8.
NCT00945282 (38) [back to overview]Change From Baseline in Clinical Chemistry Paramaters- Albumin and Total Protein
NCT00945282 (38) [back to overview]Change From Baseline in Clinical Chemistry Paramaters- Alkaline Phosphatase, Alanine Amino Transferase, Aspartate Amino Transferase
NCT00945282 (38) [back to overview]Change From Baseline in Clinical Chemistry Paramaters- Phosphorus
NCT00945282 (38) [back to overview]Change From Baseline in Clinical Chemistry Paramaters- Thyroxine Total, Thyroxine Binding Globulin, Total T3.
NCT00945282 (38) [back to overview]Change From Baseline in Clinical Chemistry Paramaters- Thyroxine, Free
NCT00945282 (38) [back to overview]Change From Baseline in Clinical Chemistry Paramaters- Uric Acid
NCT00945282 (38) [back to overview]Change From Baseline in Clinical Chemistry Paramaters-sodium, Potassium and Carbondioxide or Bicarbonate
NCT00945282 (38) [back to overview]Change From Baseline in Clinical Chemistry Parameters- Blood Urea Nitrogen, Triglycerides, Glucose, Creatinine, Calcium, Cholesterol, Total Bilirubin, and Direct Bilirubin.
NCT00945282 (38) [back to overview]Change From Baseline in Hematology Paramaters- Hematocrit
NCT00945282 (38) [back to overview]Change From Baseline in Hematology Paramaters- Hemoglobin
NCT00945282 (38) [back to overview]Change From Baseline in Hematology Paramaters- Mean Corpuscle Hemoglobin (MCH)
NCT00945282 (38) [back to overview]GSK2248761 PK Parameters Following Dose Administration on Day 1: Time to Maximum Observed Concentration (Tmax), Terminal Half-life (t1/2), Absorption Lag Time (Tlag)
NCT00945282 (38) [back to overview]Change From Baseline in Hematology Paramaters- Mean Corpuscle Volume (MCV)
NCT00945282 (38) [back to overview]Change From Baseline in Hematology Paramaters- Platelet Count
NCT00945282 (38) [back to overview]Change From Baseline in Hematology Paramaters- Red Blood Cell Count
NCT00945282 (38) [back to overview]Change From Baseline in Hematology Paramaters-Mean Corpuscle Hemoglobin Concentration
NCT00945282 (38) [back to overview]Change From Baseline in Hematology Parameters- Total Neutrophil
NCT00945282 (38) [back to overview]Change From Baseline in HR
NCT00945282 (38) [back to overview]Change From Baseline in Vital Signs-systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)
NCT00945282 (38) [back to overview]GSK2248761 Pharmacokinetic (PK) Parameters Following Dose Administration on Day 1: Area Under the Plasma Concentration Time Curve 0 to Infinite (AUC[0-∞]) and Area Under the Plasma Concentration Time Curve (AUC [0-24])
NCT00945282 (38) [back to overview]GSK2248761 PK Parameters Following Dose Administration on Day 1: Maximum Observed Concentration (Cmax) and Concentration at 24 Hours Post Dose (C24)
NCT00945282 (38) [back to overview]GSK2248761 PK Parameters Following Dose Administration on Day 7: t1/2
NCT00945282 (38) [back to overview]GSK2248761 PK Parameters Following Dose Administration on Day 7: Predose Concentration (C0), Concentration at End of Dosing Interval (Cτ), Minimum Observed Concentration During One Dosing Interval (Cmin) and Cmax
NCT00945282 (38) [back to overview]Number of Participants With Abnormal Electrocardiogram (ECG) Findings
NCT00945282 (38) [back to overview]Number of Participants With Serious Adverse Events (SAEs) and Adverse Events (AEs)
NCT00945282 (38) [back to overview]PK Data of Cmax and Ctau at Different Doses for the Assessment of Dose Proportionality
NCT00945282 (38) [back to overview]Percent Change From Baseline in CD4+ and CD8+ T-lymphocyte Cell Count at Day 1 and Day 8
NCT00945282 (38) [back to overview]PK Data of Day 1 AUC(0-inf) and Day 7 AUC(0-tau) at Different Doses for the Assessment of Dose Proportionality
NCT00976950 (3) [back to overview]Virologic Response
NCT00976950 (3) [back to overview]Change in CD4+ Cell Count From Baseline at Week 48
NCT00976950 (3) [back to overview]Number of Patients Reporting Adverse Events (AE)
NCT00978068 (6) [back to overview]Percentage of Uncomplicated Malaria Episodes With Accompanying Adverse Events That Occurred in the 28 Days Following Antimalarial Therapy
NCT00978068 (6) [back to overview]Incidence-density of Malaria Defined as the Number of Incident Episodes of Malaria Per Time at Risk.
NCT00978068 (6) [back to overview]Incidence-density of Malaria Defined as the Number of Incident Episodes of Complicated Malaria Per Time at Risk.
NCT00978068 (6) [back to overview]Estimates of the 6-month Risk of a First Episode of Malaria
NCT00978068 (6) [back to overview]63-day Risk of Recurrent Malaria
NCT00978068 (6) [back to overview]28-day Risk of Recurrent Parasitemia
NCT00983515 (3) [back to overview]Maximum Plasma Concentration (Cmax)
NCT00983515 (3) [back to overview]Area Under the Concentration Versus Time Curve From Time 0 to Time t [AUC(0-t)]
NCT00983515 (3) [back to overview]Area Under the Concentration Versus Time Curve From Time 0 Extrapolated to Infinity [AUC(0-∞)]
NCT00985543 (2) [back to overview]Adverse Events
NCT00985543 (2) [back to overview]Plasma Lopinavir/Ritonavir Concentrations as Measured by the Area Under the Curve (AUC 0-12h).
NCT00993031 (14) [back to overview]Placental Malaria Defined as Positive Placental RDT
NCT00993031 (14) [back to overview]Incidence of Pre-eclampsia Defined by Hypertension > 140/90 on Two Occasions Measured > 6 Hours Apart With ≥1+ Proteinuria on Clean Catch Urine Dipstick
NCT00993031 (14) [back to overview]ART Levels in Hair Samples at Delivery
NCT00993031 (14) [back to overview]Maternal Malaria Defined as the Number of Treatments for New Episodes of Malaria Per Time at Risk After Pregnancy
NCT00993031 (14) [back to overview]Maternal Malaria Defined as the Number of Treatments for New Episodes of Malaria Per Time at Risk During Pregnancy
NCT00993031 (14) [back to overview]Number of Participants With Maternal HIV RNA Suppression of <400 Copies/mL
NCT00993031 (14) [back to overview]Number of Participants With Maternal to Child Transmission of HIV, Measured by Infant HIV DNA PCR
NCT00993031 (14) [back to overview]Number of Participants With Severe Maternal Anemia Defined by Hemoglobin < 8g/dl at Any Point During the Trial in Each Treatment Group
NCT00993031 (14) [back to overview]Prevalence of Composite Clinical Outcome Defined by LBW, Stillbirth(Intrauterine Fetal Demise >20wks GA), Late Spontaneous Abortion(Miscarriage 12-20wks GA), Preterm Delivery(<37wks Gestation), Neonatal Death(Death of Liveborn Infant Within First 28days)
NCT00993031 (14) [back to overview]Prevalence of Malaria Defined as Positive Placental Blood PCR
NCT00993031 (14) [back to overview]Number of Participants With Grade 3 or 4 Toxicity in the Two Treatment Groups in Women
NCT00993031 (14) [back to overview]Placental Malaria Defined Placental Histopathologic Analysis
NCT00993031 (14) [back to overview]Change in Maternal CD4 Cell Counts
NCT00993031 (14) [back to overview]Prevalence of Malaria Defined as Positive Placental Blood Smear
NCT00993148 (9) [back to overview]Drug Adherence, Number of Participants With Missed Doses
NCT00993148 (9) [back to overview]Percentage of Participants With Plasma HIV-1 RNA >50 Copies/mL
NCT00993148 (9) [back to overview]Signs/Symptoms or Laboratory Toxicities of Grade 3 or Higher
NCT00993148 (9) [back to overview]Median CD4 Count Change From Baseline
NCT00993148 (9) [back to overview]Trough Concentrations (Ctrough) of Maraviroc
NCT00993148 (9) [back to overview]Proportion of Participants With Plasma HIV-1 RNA >50 Copies/mL
NCT00993148 (9) [back to overview]Drug Resistance Mutations and Co-receptor Tropism Assessed by Trofile ES
NCT00993148 (9) [back to overview]Percentage of Participants With Virologic Failure or Off Study Treatment Regimen
NCT00993148 (9) [back to overview]Percentage of Participants With Plasma HIV-1 RNA >50
NCT01003990 (1) [back to overview]Number of Participants With Serious Adverse Events (SAEs), Treatment Related SAEs, Treatment Related Adverse Events (AEs), AEs Leading to Discontinuation of Study Therapy, Grade 3 to Grade 4 AEs, Grade 3 to Grade 4 AEs, CDC Class C AIDS Events, or Death
NCT01009814 (36) [back to overview]AUC (0-24) of Ritonavir Following Q12H Dosing
NCT01009814 (36) [back to overview]AUC (0-24) of Ritonavir Following QHS Dosing
NCT01009814 (36) [back to overview]Inhibitory Quotient of BMS-626529 by Ctrough Following Q12H Dosing
NCT01009814 (36) [back to overview]Inhibitory Quotient of BMS-626529 by Ctrough Following QHS Dosing
NCT01009814 (36) [back to overview]Mean Logarithm With Base 10 (Log10) Change From Baseline in Human Immunodeficiency Virus (HIV) Ribonucleic Acid (RNA) at Day 9
NCT01009814 (36) [back to overview]Number of Participants With Any Abnormality in Physical Examination
NCT01009814 (36) [back to overview]Number of Participants With Clinically Significant Abnormalities in Laboratory Parameters
NCT01009814 (36) [back to overview]Area Under the Concentration-time Curve in One Dosing Interval (AUC [Tau]) of BMS-626529 Following Q12H Dosing
NCT01009814 (36) [back to overview]AUC (Tau) of BMS-626529 Following QHS Dosing
NCT01009814 (36) [back to overview]AUC (Tau) of Ritonavir Following Q12H Dosing
NCT01009814 (36) [back to overview]AUC (Tau) of Ritonavir Following Q12H Dosing
NCT01009814 (36) [back to overview]AUC (Tau) of Ritonavir Following QHS Dosing
NCT01009814 (36) [back to overview]Change From Baseline in Cluster of Differentiation 4 Positive (CD4+) Cell Count and Cluster of Differentiation 8 Positive (CD8+) Cell Count
NCT01009814 (36) [back to overview]Change From Baseline in Percent CD4+ Cell Count and Percent CD8+ Cell Count
NCT01009814 (36) [back to overview]Cmax of BMS-626529 Following QHS Dosing
NCT01009814 (36) [back to overview]Cmax of Ritonavir Following Q12H Dosing
NCT01009814 (36) [back to overview]Cmax of Ritonavir Following Q12H Dosing
NCT01009814 (36) [back to overview]Cmax of Ritonavir Following QHS Dosing
NCT01009814 (36) [back to overview]Ctrough of BMS-626529 Following QHS Dosing
NCT01009814 (36) [back to overview]Ctrough of Ritonavir Following Q12H Dosing
NCT01009814 (36) [back to overview]Ctrough of Ritonavir Following Q12H Dosing
NCT01009814 (36) [back to overview]Ctrough of Ritonavir Following QHS Dosing
NCT01009814 (36) [back to overview]Inhibitory Quotient (IQ) of BMS-626529 by Css,Avg and by Lowest Concentration of a Drug During Dosing Interval (Cmin) Following Q12H Dosing
NCT01009814 (36) [back to overview]Inhibitory Quotient (IQ) of BMS-626529 by Css,Avg and by Lowest Concentration of a Drug During Dosing Interval (Cmin) Following QHS Dosing
NCT01009814 (36) [back to overview]Maximum Observed Plasma Concentration (Cmax) of BMS-626529 Following Q12H Dosing
NCT01009814 (36) [back to overview]Number of Participants With Treatment Emergent Non-serious Adverse Event (Non-SAE) and Serious AE (SAE)
NCT01009814 (36) [back to overview]Number of Participants With Worst-case Abnormalities in Diastolic Blood Pressure (DBP) and Systolic Blood Pressure (SBP)
NCT01009814 (36) [back to overview]Number of Participants With Worst-case Abnormalities in Electrocardiogram (ECG) Parameters
NCT01009814 (36) [back to overview]Trough Observed Plasma Concentration (Ctrough) of BMS-626529 Following Q12H Dosing
NCT01009814 (36) [back to overview]Number of Participants With Worst-case Abnormalities in Body Temperature, Respiratory Rate [RR], and Heart Rate [HR]
NCT01009814 (36) [back to overview]Accumulation Index (AI) of BMS-626529 Following Q12H Dosing
NCT01009814 (36) [back to overview]Accumulation Index of BMS-626529 Following QHS Dosing
NCT01009814 (36) [back to overview]Accumulation Index of Ritonavir Following Q12H Dosing
NCT01009814 (36) [back to overview]Accumulation Index of Ritonavir Following QHS Dosing
NCT01009814 (36) [back to overview]Area Under the Concentration-time Curve Over a 24-hour Period (AUC [0-24]) of BMS-626529 Following Q12H Dosing
NCT01009814 (36) [back to overview]AUC (0-24) of BMS-626529 Following QHS Dosing
NCT01010399 (2) [back to overview]Proportion of Subjects With HIV-1 RNA <50 Copies/mL
NCT01010399 (2) [back to overview]Proportion of Subjects With Triglycerides <200 mg/dL
NCT01057433 (1) [back to overview]Area Under the Curve From Time 0 to Tau (AUC 0-τ)
NCT01061151 (28) [back to overview]Maternal Health Component: Incidence of HIV/AIDS-related Event or Death
NCT01061151 (28) [back to overview]Antepartum Component: Probability of Overall and HIV-free Infant Survival Until 104 Weeks of Age, by Antepartum Arm (in Conjunction With Infants in the Postpartum Component)
NCT01061151 (28) [back to overview]Antepartum Component: Number of Mothers With Obstetrical Complications
NCT01061151 (28) [back to overview]Antepartum Component: Number of Mothers With Grade 3 or Higher Toxicities and Selected Grade 2 Hematologic, Renal, and Hepatic Adverse Events
NCT01061151 (28) [back to overview]Antepartum Component: Number of Mothers With Grade 3 or Higher Toxicities and Selected Grade 2 Hematologic, Renal, and Hepatic Adverse Events
NCT01061151 (28) [back to overview]Antepartum Component: Number of Mothers With Adverse Pregnancy Outcomes (e.g.,Stillbirth, Preterm Delivery (< 37 Weeks), Low Birth Weight (< 2,500 Grams), and Congenital Anomalies)
NCT01061151 (28) [back to overview]Antepartum Component: Number of Confirmed Infant HIV Infections
NCT01061151 (28) [back to overview]Antepartum Component: Number of Mothers With Adverse Pregnancy Outcomes (e.g.,Stillbirth, Preterm Delivery (< 37 Weeks), Low Birth Weight (< 2,500 Grams), and Congenital Anomalies)
NCT01061151 (28) [back to overview]Postpartum Component: Proportion of Mother-Infant Pairs With no Death or HIV Diagnosis Through 24 Months Post-delivery
NCT01061151 (28) [back to overview]Postpartum Component: Incidence of Grade 3 or Higher Adverse Events and Selected Grade 2 Hematologic, Renal, and Hepatic Adverse Events
NCT01061151 (28) [back to overview]Antepartum Component: Number of Infant HIV Infections
NCT01061151 (28) [back to overview]Maternal Health Component: Incidence of AIDS-defining Illness
NCT01061151 (28) [back to overview]Maternal Health Component: Incidence of HIV/AIDS-related Events
NCT01061151 (28) [back to overview]Antepartum Component: Number of Mothers With Obstetrical Complications
NCT01061151 (28) [back to overview]Postpartum Component: Adherence to the Maternal and/or Infant ARV Regimens, as Measured by Maternal Report and Hair Measures
NCT01061151 (28) [back to overview]Antepartum Component: Maternal HIV RNA Less Than 400 Copies/mL at Delivery
NCT01061151 (28) [back to overview]Postpartum Component: Proportion of Infants Alive Through 12 and 24 Months Post-delivery
NCT01061151 (28) [back to overview]Antepartum Component: Probability of Overall and HIV-free Infant Survival Until 104 Weeks of Age, by Antepartum Arm (in Conjunction With Infants in the Postpartum Component)
NCT01061151 (28) [back to overview]Postpartum Component: Incidence of Confirmed Infant HIV Infection
NCT01061151 (28) [back to overview]Maternal Health Component: Incidence of Death
NCT01061151 (28) [back to overview]Maternal Health Component: Incidence Rate of Progression to AIDS-defining Illness, Death, or a Serious Non-AIDS Cardiovascular, Hepatic, or Renal Event
NCT01061151 (28) [back to overview]Maternal Health Component: Incidence of HIV/AIDS-related Event or World Health Organization (WHO) Clinical Stage 2 or 3 Events
NCT01061151 (28) [back to overview]Maternal Health Component: Toxicity: Incidence of Grade 3 or Greater Laboratory Results or Signs and Symptoms and Selected Grade 2 Hematologic, Renal, and Hepatic Laboratory Results
NCT01061151 (28) [back to overview]Maternal Health Component: Incidence of Progression to AIDS-defining Illness or Death
NCT01061151 (28) [back to overview]Maternal Health Component: Incidence of Tuberculosis
NCT01061151 (28) [back to overview]Maternal Health Component: Incidence Rate of Cardiovascular or Other Metabolic Events
NCT01061151 (28) [back to overview]Maternal Health Component: Incidence Rate of Death or Any Condition of Particular Concern
NCT01061151 (28) [back to overview]Maternal Health Component: Other Targeted Medical Conditions
NCT01068873 (1) [back to overview]Number of Participants With HIV RNA < 50 and <400 Copies/ml.
NCT01074008 (24) [back to overview]Number of Participants With Resistance-Associated Variants and Phenotypic Resistance to ABT-333 in Non-structural Viral Protein 5B (NS5B)
NCT01074008 (24) [back to overview]Number of Participants With Resistance-Associated Variants and Phenotypic Resistance to ABT-072 in Non-structural Viral Protein 5B (NS5B)
NCT01074008 (24) [back to overview]Time to Maximum Plasma Concentration (Tmax) of Ritonavir
NCT01074008 (24) [back to overview]Time to Maximum Plasma Concentration (Tmax) of ABT-450
NCT01074008 (24) [back to overview]Time to Maximum Plasma Concentration (Tmax) of ABT-333
NCT01074008 (24) [back to overview]Time to Maximum Plasma Concentration (Tmax) of ABT-072
NCT01074008 (24) [back to overview]Percentage of Participants With Rapid Virologic Response (RVR) at Week 4
NCT01074008 (24) [back to overview]Percentage of Participants With Complete Early Virologic Response (cEVR) at Week 12
NCT01074008 (24) [back to overview]Maximum Plasma Concentration (Cmax) of Ritonavir
NCT01074008 (24) [back to overview]Maximum Plasma Concentration (Cmax) of ABT-450
NCT01074008 (24) [back to overview]Maximum Plasma Concentration (Cmax) of ABT-333
NCT01074008 (24) [back to overview]Maximum Plasma Concentration (Cmax) of ABT-072
NCT01074008 (24) [back to overview]Maximal Change From Baseline in Hepatitis C Virus Ribonucleic Acid (HCV RNA) Levels During ABT-450/r, ABT-333, or ABT-072 Monotherapy Treatment
NCT01074008 (24) [back to overview]Change From Baseline in SF-36 Physical Component Summary (PCS)
NCT01074008 (24) [back to overview]Change From Baseline in SF-36 Mental Component Summary (MCS)
NCT01074008 (24) [back to overview]Change From Baseline in Hepatitis C Virus Patient-reported Outcomes (HCV-PRO) Total Score
NCT01074008 (24) [back to overview]Change From Baseline in EQ-5D (3 Level) Health Index Score
NCT01074008 (24) [back to overview]Change From Baseline in ED-5D Visual Analog Scale (ED-5D VAS) Score
NCT01074008 (24) [back to overview]Area Under the Plasma Concentration-time Curve From 0 to 24 Hours (AUC24) Post-dose of Ritonavir
NCT01074008 (24) [back to overview]Area Under the Plasma Concentration-time Curve From 0 to 12 Hours (AUC12) Post-dose of ABT-333
NCT01074008 (24) [back to overview]Area Under the Plasma Concentration-time Curve From 0 to 24 Hours (AUC24) Post-dose of ABT-450
NCT01074008 (24) [back to overview]Area Under the Plasma Concentration-time Curve From 0 to 24 Hours (AUC24) Post-dose of ABT-072
NCT01074008 (24) [back to overview]Percentage of Participants With Partial Early Virologic Response (EVR) at Week 12
NCT01074008 (24) [back to overview]Number of Participants With Resistance-Associated Variants and Phenotypic Resistance to ABT-450 in Non-structural Viral Protein 3 (NS3)
NCT01074931 (6) [back to overview]Evolution of CD4 Count
NCT01074931 (6) [back to overview]Evolution of the HIV Viral Response
NCT01074931 (6) [back to overview]Number of Participants Who Missed Doses, Interrupt or Discontinue Regimen, and Experience Changes in Dosage or of Combination Regimen
NCT01074931 (6) [back to overview]Evolution of the Tolerance Issues
NCT01074931 (6) [back to overview]Adverse Events Observed and Development of Lipodystrophy Lesion and Their Locations
NCT01074931 (6) [back to overview]The Duration on Treatment Until Development of an Adverse Event Leading to Treatment Discontinuation or Until Escape From Treatment
NCT01076972 (4) [back to overview]Mean Number of Human Immunodeficiency Virus (HIV) Ribonucleic Acid (RNA) Copies Per Milliliter (mL) Using a Logarithmic (Base 10) Transformation at Each Visit
NCT01076972 (4) [back to overview]Cluster of Differentiation 4 Lymphocyte Count (CD4)
NCT01076972 (4) [back to overview]Number of Patients Included in Each Center for Disease Control and Prevention (CDC) Classification Category for HIV-infected Adults and Adolescents
NCT01076972 (4) [back to overview]Total Number of Patients With Adverse Drug Reactions
NCT01076985 (1) [back to overview]Number of Patients With Adverse Drug Reactions (ADRs)
NCT01083810 (6) [back to overview]Percentage of Patients With HIV-1 RNA 50 to <200 Copies/ml
NCT01083810 (6) [back to overview]Change in Absolute CD4 Cell Count [CD4+ Cells/µL]
NCT01083810 (6) [back to overview]Percentage of Patients With HIV-1 RNA <50 Copies/ml
NCT01083810 (6) [back to overview]Percentage of Patients With HIV-1 RNA 200 to <500 Copies/ml
NCT01083810 (6) [back to overview]Percentage of Patients With HIV-1 RNA >500 Copies/ml
NCT01083810 (6) [back to overview]Number of Patients With Virus That Develop Mutations Conferring Resistance to Lopinavir/Ritonavir, NRTIs or NNRTIs
NCT01095094 (2) [back to overview]Grade 3-5 Toxicity as Assessed by NCI CTC v3.0
NCT01095094 (2) [back to overview]Progression-free Survival
NCT01099579 (18) [back to overview]Time to Maximum Observed Concentration (Tmax) of Atazanavir and Ritonavir
NCT01099579 (18) [back to overview]Maximum Observed Concentration (Cmax) and Minimum Observed Concentration (Cmin) of Atazanavir and Ritonavir
NCT01099579 (18) [back to overview]Mean Change From Baseline in HIV RNA Levels at Week 48 by Prior Antiretroviral (ARV) Treatment Status
NCT01099579 (18) [back to overview]Electrocardiogram Changes From Baseline in PR Interval, QTC Bazett, and QTC Fridericia at Week 48
NCT01099579 (18) [back to overview]Percentage of Participants With HIV RNA Levels <50 c/mL and <400 c/mL at Week 48 by Treatment/Weight
NCT01099579 (18) [back to overview]Percentage of Participants With HIV RNA Levels <50 c/mL and <400 c/mL at Week 48 by Prior Antiretroviral (ARV) Treatment Status
NCT01099579 (18) [back to overview]Number of Participants With Laboratory Test Results With Worst Toxicity of Grade 3-4
NCT01099579 (18) [back to overview]Number of Participants Who Acquired Phenotypic Resistance to Atazanavir or Atazanovir/Ritonavir
NCT01099579 (18) [back to overview]CD4 Cell Count Changes From Baseline at Week 48 by Prior Antiretroviral (ARV) Treatment Status
NCT01099579 (18) [back to overview]CD4 Cell Count Changes From Baseline at Week 48 by Treatment/Weight
NCT01099579 (18) [back to overview]Mean CD4 Percent Changes From Baseline at Week 48 by Antiretroviral (ARV) Treatment Status
NCT01099579 (18) [back to overview]Mean CD4 Percent Changes From Baseline at Week 48 by Treatment/Weight
NCT01099579 (18) [back to overview]Mean Change From Baseline in HIV RNA Levels at Week 48 by Treatment/Weight
NCT01099579 (18) [back to overview]Number of Participants With Centers for Disease Control (CDC) Class C AIDS Events
NCT01099579 (18) [back to overview]Apparent Total Body Clearance (CLT/F) of Atazanavir and Ritonavir
NCT01099579 (18) [back to overview]Number of Participants With Death as Outcome, Serious Adverse Events (SAEs), Adverse Events (AEs) Leading to Discontinuation
NCT01099579 (18) [back to overview]Apparent Total Body Clearance Per Body Weight (CLT/F) Per Kilogram of Atazanavir and Ritonavir
NCT01099579 (18) [back to overview]Area Under the Concentration Curve (in 1 Dosing Interval From Time 0 to 24 Hours Post Observed Dose) (AUC[TAU])of Atazanavir and Ritonavir
NCT01106586 (7) [back to overview]The Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 48
NCT01106586 (7) [back to overview]The Percentage of Participants With Virologic Success Using the Food and Drug Administration (FDA)-Defined Snapshot Analysis as Determined by the Achievement of HIV-1 Ribonucleic Acid (RNA) < 50 Copies/mL at Week 48
NCT01106586 (7) [back to overview]The Percentage of Participants Achieving and Maintaining Confirmed HIV-1 RNA < 50 Copies/mL at Week 48 Using the FDA-defined Time to Loss of Virologic Response (TLOVR) Algorithm
NCT01106586 (7) [back to overview]The Percentage of Participants With Virologic Success Using the FDA-Defined Snapshot Analysis as Determined by the Achievement of HIV-1 RNA < 50 Copies/mL at Week 96
NCT01106586 (7) [back to overview]The Percentage of Participants With Virologic Success Using the FDA-Defined Snapshot Analysis as Determined by the Achievement of HIV-1 RNA < 50 Copies/mL at Week 192
NCT01106586 (7) [back to overview]The Percentage of Participants With Virologic Success Using the FDA-Defined Snapshot Analysis as Determined by the Achievement of HIV-1 RNA < 50 Copies/mL at Week 144
NCT01106586 (7) [back to overview]The Change From Baseline in Cluster Determinant 4 (CD4) Cell Count at Weeks 48, 96, 144, and 192
NCT01146873 (5) [back to overview]Viral Failure
NCT01146873 (5) [back to overview]Viral Rebound
NCT01146873 (5) [back to overview]CD4 Cell Percentage at 48 Weeks After Randomization
NCT01146873 (5) [back to overview]Highest Grade ALT After Randomization
NCT01146873 (5) [back to overview]Percentage of Participants With Elevated Total Cholesterol, Elevated LDL, Abnormal HDL, or Abnormal Triglycerides at 40 Weeks After Randomization
NCT01153269 (42) [back to overview]Viral Load
NCT01153269 (42) [back to overview]Viral Load
NCT01153269 (42) [back to overview]Viral Load
NCT01153269 (42) [back to overview]Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters
NCT01153269 (42) [back to overview]Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters
NCT01153269 (42) [back to overview]Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters
NCT01153269 (42) [back to overview]Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters
NCT01153269 (42) [back to overview]CD4 Cell Count
NCT01153269 (42) [back to overview]CD4 Cell Count
NCT01153269 (42) [back to overview]CD4 Cell Count
NCT01153269 (42) [back to overview]CD4 Cell Count
NCT01153269 (42) [back to overview]Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters
NCT01153269 (42) [back to overview]Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters
NCT01153269 (42) [back to overview]Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters
NCT01153269 (42) [back to overview]Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters
NCT01153269 (42) [back to overview]Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters
NCT01153269 (42) [back to overview]Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters
NCT01153269 (42) [back to overview]Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters
NCT01153269 (42) [back to overview]Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters
NCT01153269 (42) [back to overview]Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters
NCT01153269 (42) [back to overview]Viral Load
NCT01153269 (42) [back to overview]Viral Load
NCT01153269 (42) [back to overview]Viral Load
NCT01153269 (42) [back to overview]Viral Load
NCT01153269 (42) [back to overview]Viral Load
NCT01153269 (42) [back to overview]Viral Load
NCT01153269 (42) [back to overview]Viral Load
NCT01153269 (42) [back to overview]Viral Load
NCT01153269 (42) [back to overview]Viral Load
NCT01153269 (42) [back to overview]Viral Load
NCT01153269 (42) [back to overview]CD4 Cell Count
NCT01153269 (42) [back to overview]CD4 Cell Count
NCT01153269 (42) [back to overview]CD4 Cell Count
NCT01153269 (42) [back to overview]Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters
NCT01153269 (42) [back to overview]CD4 Cell Count
NCT01153269 (42) [back to overview]CD4 Cell Count
NCT01153269 (42) [back to overview]CD4 Cell Count
NCT01153269 (42) [back to overview]CD4 Cell Count
NCT01153269 (42) [back to overview]CD4 Cell Count
NCT01153269 (42) [back to overview]CD4 Cell Count
NCT01153269 (42) [back to overview]CD4 Cell Count
NCT01153269 (42) [back to overview]Viral Load
NCT01154673 (1) [back to overview]Change in Proviral HIV-1 DNA in Total CD4+ T-cells From Baseline to Week 48 in Participants Randomized to the Intensified Arm Versus the Control Arm Who Received Placebo in Addition to Standard HAART.
NCT01156389 (4) [back to overview]Pharmacokinetics Analysis: Cmax
NCT01156389 (4) [back to overview]Summary of Treatment Emergent Adverse Events
NCT01156389 (4) [back to overview]Pharmacokinetics Analysis: Half-life, Tmax
NCT01156389 (4) [back to overview]Pharmacokinetics Analysis: AUC0-tau, AUC0-∞
NCT01172535 (10) [back to overview]Adherence
NCT01172535 (10) [back to overview]Minimum Concentration of Lopinavir/Ritonavir (Cmin)
NCT01172535 (10) [back to overview]Clearance of Lopinavir/Ritonavir (CL/F)
NCT01172535 (10) [back to overview]Lopinovir/Ritonavir Area Under the Concentration-time Curve (AUC0-24)
NCT01172535 (10) [back to overview]Maximum Concentration of Lopinavir/Ritonavir (Cmax)
NCT01172535 (10) [back to overview]Treatment Efficacy (HIV Viral Load)
NCT01172535 (10) [back to overview]Treatment Efficacy (CD4%)
NCT01172535 (10) [back to overview]Proportion of Participants Tolerating LPV/r
NCT01172535 (10) [back to overview]Proportion of Participants With an AUC of Less Than 10% of Adults
NCT01172535 (10) [back to overview]Number of Participants Experiencing Adverse Events of Grade 3 or 4
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 20
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 8
NCT01199939 (14) [back to overview]Change From Baseline in Cluster of Differentiation 4 (CD4+) and Cluster of Differentiation 8 (CD8+) Cell Counts at Week 48
NCT01199939 (14) [back to overview]Time to Reach First Confirmed Virologic Response
NCT01199939 (14) [back to overview]Number of Participants With Virologic Failure
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 12
NCT01199939 (14) [back to overview]Number of Participants With Confirmed Virologic Response (CVR) at Week 48
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 16
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 48
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 42
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 4
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 36
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 30
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 24
NCT01221298 (7) [back to overview]Percentage of Participants With Sustained Virologic Response 24 Weeks (SVR24) Post-Treatment
NCT01221298 (7) [back to overview]Percentage of Participants With Sustained Virologic Response 12 Weeks (SVR12) Post-treatment
NCT01221298 (7) [back to overview]Percentage of Participants With Hepatitis C Virus (HCV) Ribonucleic Acid (RNA) Suppressed Below the Lower Limit of Quantitation (LLOQ) From Week 4 Through Week 12
NCT01221298 (7) [back to overview]Percentage of Participants With Hepatitis C Virus (HCV) Ribonucleic Acid (RNA) Below the Lower Limit of Quantitation (LLOQ) at Week 4
NCT01221298 (7) [back to overview]Time to Virologic Relapse Through 24 Weeks Post-treatment
NCT01221298 (7) [back to overview]Percentage of Participants With Hepatitis C Virus (HCV) Ribonucleic Acid (RNA) < 1000 International Units Per Milliliter (IU/mL)
NCT01221298 (7) [back to overview]Time to Failure to Suppress or Rebound During Treatment
NCT01232127 (7) [back to overview]Number of Participants With Death as Outcome, Serious Adverse Events (SAEs), Adverse Events (AEs) Leading to Discontinuation, AES, and AEs of Clinical Interest
NCT01232127 (7) [back to overview]Time of Maximum Observed Plasma Concentration (Tmax) for Atazanavir and Ritonavir
NCT01232127 (7) [back to overview]Maximum Observed Plasma Concentration (Cmax) and Trough Observed Plasma Concentration (Ctrough) for Atazanavir and Ritonavir
NCT01232127 (7) [back to overview]Number of Participants With Abnormalities in Electrocardiogram (ECG) Findings
NCT01232127 (7) [back to overview]Area Under the Plasma Concentration-time Curve in 1 Dosing Interval (Time 0 to 24 Hours Postdose) (AUC[TAU]) for Atazanavir and Ritonavir
NCT01232127 (7) [back to overview]Number of Participants With Abnormalities in Laboratory Test Results
NCT01232127 (7) [back to overview]Number of Participants With Abnormalities in Vital Signs
NCT01281813 (3) [back to overview]Number of Participants With Adverse Events Leading to Study Drug Discontinuation
NCT01281813 (3) [back to overview]Number of Participants With Adverse Events Possibly Related to Darunavir/Ritonavir (DRV/Rtv) Treatment
NCT01281813 (3) [back to overview]Number of Participants With Serious Adverse Events
NCT01306617 (12) [back to overview]Percentage of Participants With Sustained Virologic Response 24 Weeks (SVR24) Post-Treatment
NCT01306617 (12) [back to overview]Pharmacokinetics (C Trough) of ABT 450 in HCV Infected Participants
NCT01306617 (12) [back to overview]Pharmacokinetics (C Trough) of ABT-333 in HCV Infected Participants
NCT01306617 (12) [back to overview]Pharmacokinetics (C Trough) of Ribavirin in HCV Infected Participants
NCT01306617 (12) [back to overview]Pharmacokinetics (C Trough) of Ritonavir in HCV Infected Participants
NCT01306617 (12) [back to overview]Time to Failure to Suppress or Rebound During Treatment
NCT01306617 (12) [back to overview]Time to Virologic Relapse Post-treatment
NCT01306617 (12) [back to overview]Resistance-Associated Variants and Phenotypic Resistance
NCT01306617 (12) [back to overview]Percentage of Participants With HCV RNA < 1000 International Units Per Milliliter (IU/mL)
NCT01306617 (12) [back to overview]Percentage of Participants With HCV RNA Below the Lower Limit of Quantitation (LLOQ; <25 IU/mL) at Week 4
NCT01306617 (12) [back to overview]Percentage of Participants With Hepatitis C Virus (HCV) Ribonucleic Acid (RNA) Suppressed Below the Lower Limit of Detection (LLOD) From Week 4 Through Week 12
NCT01306617 (12) [back to overview]Percentage of Participants With Sustained Virologic Response 12 Weeks (SVR12) Post-treatment
NCT01332227 (7) [back to overview]Number of Participants With Genotypable/Phenotypable Isolates, Emergent Genotypic Substitutions in Patients With Genotypable Isolates, and Phenotypic Resistance in Patients With Phenotypable Isolates at Week 24
NCT01332227 (7) [back to overview]Number of Participants With Genotypable/Phenotypable Isolates, Emergent Genotypic Substitutions in Patients With Genotypable Isolates, and Phenotypic Resistance in Patients With Phenotypable Isolates at Week 48
NCT01332227 (7) [back to overview]Number of Patients With Death as Outcome, Serious Adverse Events (SAEs), Treatment-related SAEs, Treatment-emergent Adverse Events (AEs) Leading to Discontinuation, and Treatment-emergent AEs
NCT01332227 (7) [back to overview]Number of Participants With Virologic Rebound at Weeks 24 and 48
NCT01332227 (7) [back to overview]Mean Changes in Fasting Lipid Levels From Baseline to Week 48
NCT01332227 (7) [back to overview]Percentage of Participants With HIV-1 RNA Level <40 c/mL at Week 24
NCT01332227 (7) [back to overview]Percentage of Participants With HIV-1 RNA Level <40 c/mL at Week 48
NCT01335698 (14) [back to overview]Number of Participants Who Died and With Adverse Events (AEs) Leading to Discontinuation, Hyperbilirubinemia, Jaundice, First-degree Arterioventricular Block, Tachycardia, and Rash on ATV Powder
NCT01335698 (14) [back to overview]Minimum Plasma Concentration (Cmin)
NCT01335698 (14) [back to overview]Number of Participants Who Experienced a SAE on ATV Powder
NCT01335698 (14) [back to overview]CD4 Cell Count Changes From Baseline on ATV Powder
NCT01335698 (14) [back to overview]CD4 Cell Count Changes From Baseline on ATV Powder
NCT01335698 (14) [back to overview]Mean Change From Baseline in CD4 Percent on ATV Powder
NCT01335698 (14) [back to overview]Mean Change From Baseline in CD4 Percent on ATV Powder
NCT01335698 (14) [back to overview]Mean Change From Baseline in HIV RNA on ATV Powder
NCT01335698 (14) [back to overview]Number of Participants With A Center of Disease Control and Prevention (CDC) Class C AIDS Event on ATV Powder
NCT01335698 (14) [back to overview]Number of Participants With Emergent Genotypic Substitutions on ATV Powder Through Week 48
NCT01335698 (14) [back to overview]Number of Participants With HIV RNA <50 Copies/mL and <400 Copies/mL in the Week 24 Atazanavir Powder Cohort and the Eligible Week 48 Atazanavir Powder Cohort
NCT01335698 (14) [back to overview]Number of Participants With Laboratory Test Results Meeting the Criteria for Grade 3-4 Abnormality on ATV Powder
NCT01335698 (14) [back to overview]Area Under the Concentration-Time Curve [AUC(TAU)]
NCT01335698 (14) [back to overview]Maximum Observed Plasma Concentration (Cmax)
NCT01345630 (25) [back to overview]Changes in Bone Mineral Density (Using DEXA Scan and Serum Markers) From Baseline and at Week 48 - AP Lumbar Spine (L1 - L4) BMD
NCT01345630 (25) [back to overview]Change in Bone Turnover Markers From Baseline and at Week 48 - Blood Osteocalcin
NCT01345630 (25) [back to overview]Change in Bone Turnover Markers From Baseline and at Week 48 - Type 1 Collagen Peptide (CTX-1)
NCT01345630 (25) [back to overview]Frequency of Adverse Events (AE).
NCT01345630 (25) [back to overview]Changes in Trunk to Limb Fat Distribution Using DEXA Scan From Baseline and at Week 48
NCT01345630 (25) [back to overview]Changes in Peripheral Fat Distribution Using Dual Energy X-ray Absorptiometry [DEXA] Scan From Baseline and at Week 48.
NCT01345630 (25) [back to overview]Number of Treatment-related AEs
NCT01345630 (25) [back to overview]Changes in Bone Mineral Density (Using DEXA Scan and Serum Markers) From Baseline and at Week 48 - Femoral Neck BMD
NCT01345630 (25) [back to overview]Changes in Bone Mineral Density (Using DEXA Scan and Serum Markers) From Baseline and at Week 48 - Total Hip BMD
NCT01345630 (25) [back to overview]Percent Change From Baseline in Immune Cell Function at Week 48: Lymphocyte Activation Marker CD4 (%)
NCT01345630 (25) [back to overview]Virologic Outcomes at Week 48 Using Protocol-Defined Treatment Failure (PDTF).
NCT01345630 (25) [back to overview]Tropism Change Between Screening or Baseline and PDTF
NCT01345630 (25) [back to overview]Severity of Abnormal Laboratory Values
NCT01345630 (25) [back to overview]Percent Change From Baseline in Immune Cell Function at Week 48: Lymphocyte Activation Marker CD8 (%)
NCT01345630 (25) [back to overview]Number of Participants With Viral Resistance to Maraviroc (Maraviroc Treated Participants Only) in Participants Meeting PDTF Criteria.
NCT01345630 (25) [back to overview]Number of Participants With Resistance to Nucleoside/Nucleotide Reverse Transcriptase Inhibitors (NRTI), Non-nucleoside Reverse Transcriptase Inhibitors (NNRTI), and Protease Inhibitors (PI) in Participants Meeting PDTF Criteria
NCT01345630 (25) [back to overview]Number of Participants With Abnormal Laboratory Values
NCT01345630 (25) [back to overview]Absolute Change in CD4+/CD8+ Ratio From Baseline to Week 48
NCT01345630 (25) [back to overview]Absolute Change From Baseline in Immune Cell Function at Week 48: Lymphocyte Marker Cluster of Differentiation 8 (CD8, Cell/mm^3)
NCT01345630 (25) [back to overview]Absolute Change From Baseline in Immune Cell Function at Week 48: Lymphocyte Marker Cluster of Differentiation 4 (CD4, Cell/mm^3)
NCT01345630 (25) [back to overview]The Relationship Between the Proportion of Participants With Plasma HIV-1 RNA <50 Copies/mL at the Week 48 and the Screening Tropism Test (Genotype Test or ESTA).
NCT01345630 (25) [back to overview]Percentage of Participants With Plasma HIV-1 RNA <50 Copies/mL.
NCT01345630 (25) [back to overview]Number of Participants With Treatment-emergent Serious Adverse Events
NCT01345630 (25) [back to overview]Number of Participants With Grade 3 or 4 AEs
NCT01345630 (25) [back to overview]Number of Participants Who Discontinued Due to AEs
NCT01351740 (2) [back to overview]Proportions of Subjects Experiencing Virologic Failure by Randomized Treatment Arm
NCT01351740 (2) [back to overview]Proportions of Subjects in Each Randomized Treatment Arm With Atazanavir Trough Levels Below 150ng/mL
NCT01352715 (9) [back to overview]Number of Participants With HIV-1 Drug Resistance Mutations in Protease, Reverse Transcriptase, and Integrase in Participants With Virologic Failure at Baseline and at Time of Virologic Failure
NCT01352715 (9) [back to overview]Changes in Fasting Total Cholesterol, High-density Lipoprotein (HDL) Cholesterol, Low-density Lipoprotein (LDL) Cholesterol, Triglycerides, and Glucose From Baseline
NCT01352715 (9) [back to overview]Percentage of Time Spent in Hospital
NCT01352715 (9) [back to overview]Number of Participants With a Targeted Serious Non-AIDS-defining Event or Death
NCT01352715 (9) [back to overview]Change in CD4+ Cell Count From Baseline to Week 48
NCT01352715 (9) [back to overview]Cumulative Probability of Virologic Failure by Week 48
NCT01352715 (9) [back to overview]Number of Participants Discontinuing Randomized Treatment for Toxicity
NCT01352715 (9) [back to overview]Number of Participants With a New AIDS-defining Events or Death
NCT01352715 (9) [back to overview]Number of Participants With Grade 3 or Higher Adverse Event (AE) at Least One Grade Higher Than Baseline
NCT01374802 (4) [back to overview]Cτ,ss of Darunavir
NCT01374802 (4) [back to overview]Cmax,ss of Darunavir
NCT01374802 (4) [back to overview]Tmax,ss of Darunavir
NCT01374802 (4) [back to overview]AUCτ,ss of Darunavir
NCT01384734 (17) [back to overview]Change From Monotherapy Baseline in Cluster of Differentiation (CD)4+ and CD8+ T-cell Counts During Monotherapy
NCT01384734 (17) [back to overview]Change From Monotherapy Baseline in log10 HIV RNA of the Monotherapy Period
NCT01384734 (17) [back to overview]Number of Participants With Newly-emergent Genotypic Substitutions at Week 24
NCT01384734 (17) [back to overview]Number of Participants With Newly-emergent Genotypic Substitutions at Week 48
NCT01384734 (17) [back to overview]Number of Participants With Newly-emergent Genotypic Substitutions at Week 96
NCT01384734 (17) [back to overview]Number of Participants With SAE and Discontinuation Due to AEs During Monotherapy Period
NCT01384734 (17) [back to overview]Number of Participants With SAE and Discontinuation Due to AEs During Primary Study
NCT01384734 (17) [back to overview]Number of Participants With Serious Adverse Events (SAE) and Discontinuation Due to AEs up to Week 24
NCT01384734 (17) [back to overview]Percentage of Participants With Plasma HIV-1 RNA < 50 c/mL at Primary Study
NCT01384734 (17) [back to overview]Percentage of Participants With Plasma HIV-1 RNA < 50 c/mL at Day 8 of the Monotherapy Period
NCT01384734 (17) [back to overview]Change From Baseline in IC50 Fold Change Among Participants With VF at Week 48
NCT01384734 (17) [back to overview]Change From Baseline in IC50 Fold Change Among Participants With VF at Week 96
NCT01384734 (17) [back to overview]Maximum Change From Baseline in Inhibitory Concentration at 50% (IC50) Fold Change Among Participants With VF at Week 24
NCT01384734 (17) [back to overview]Maximum Decrease From Monotherapy Baseline in log10 Plasma HIV-1 RNA
NCT01384734 (17) [back to overview]Percentage of Participants With Plasma HIV-1 Ribonucleic Acid (RNA) < 50 Copies Per Milliliter (c/mL) at Week 24
NCT01384734 (17) [back to overview]Change From Baseline in CD4+ T-cell Count
NCT01384734 (17) [back to overview]Change From Monotherapy Baseline in CD4+ and CD8+ T-cell Proportion During Monotherapy
NCT01391013 (17) [back to overview]Change From Baseline in Mean Framingham Risk Score at Week 24 and Week 48: Medican Change in Framingham Risk Score
NCT01391013 (17) [back to overview]Change From Baseline in Cluster of Differentiation 4 (CD4) Count Over Week 48
NCT01391013 (17) [back to overview]Change From Baseline in Insulin Sensitivity at Week 24 and Week 48: Median Change in Homeostasis Model Assessment of Insulin Resistance (HOMA-IR)
NCT01391013 (17) [back to overview]Change From Baseline to Week 48 in Circulating Endothelial Cells
NCT01391013 (17) [back to overview]Change From Baseline to Week 24 in Brachial Artery Flow Mediated Vasodilatation (FMD): Median Change in FMD (%)
NCT01391013 (17) [back to overview]Change From Baseline to Week 48 in Lumbar Z Score: Median Change in Lumbar Z Score
NCT01391013 (17) [back to overview]Change From Baseline in Mean Triglycerides at Week 24 and Week 48: Median Change in Triglycerides
NCT01391013 (17) [back to overview]Change From Baseline in Mean Low-density Lipoprotein (LDL) Cholesterol at Week 24 and Week 48: Median Change in LDL
NCT01391013 (17) [back to overview]Change From Baseline in Mean High-density Lipoprotein (HDL) Cholesterol at Week 24 and Week 48: Median Change in HDL
NCT01391013 (17) [back to overview]Change From Baseline to Week 48 in Lumbar T Score: Median Change in Lumbar T Score
NCT01391013 (17) [back to overview]Change From Baseline to Week 48 in Precursors of Circulating Endothelial Cells
NCT01391013 (17) [back to overview]Number of Participants With a Human Immunodeficiency Virus- Ribonucleic Acid (HIV-RNA) Greater Than or Equal to 50 Copies/mL
NCT01391013 (17) [back to overview]Change From Baseline to Week 48 in Leg Fat Content: Median Change in Leg Fat (Total)
NCT01391013 (17) [back to overview]Change From Baseline to Week 48 in Femoral Neck Z Score: Median Change in Femoral Neck Z Score
NCT01391013 (17) [back to overview]Change From Baseline to Week 48 in Femoral Neck T Score: Median Change in Femoral Neck T Score
NCT01391013 (17) [back to overview]Change From Baseline to Week 48 in Brachial Artery FMD: Median Change in FMD (%)
NCT01391013 (17) [back to overview]Change From Baseline to Week 48 in Visceral Fat Content in Abdomen: Median Change in Visceral Abdominal Tissue (VAT)
NCT01400412 (20) [back to overview]Change in Levels of D-dimer From Baseline
NCT01400412 (20) [back to overview]Change in CD4 Count From Baseline to Week 24
NCT01400412 (20) [back to overview]Percent Change in Lumbar Spine Bone Mineral Density (BMD)
NCT01400412 (20) [back to overview]Percent Change in Expression of RANKL+ on CD8+ T Cells From Baseline to Week 48
NCT01400412 (20) [back to overview]Change in CD4 Count From Baseline to Week 48
NCT01400412 (20) [back to overview]Number of Participants Who Experienced Bone Fractures
NCT01400412 (20) [back to overview]Percent Change in Expression of CD57+ on CD8+ T Cells From Baseline to Week 48
NCT01400412 (20) [back to overview]Cumulative Probability of Virologic Failure by Week 48
NCT01400412 (20) [back to overview]Percent Change From Baseline in Total Hip Bone Mineral Density (BMD)
NCT01400412 (20) [back to overview]Percentage Change in Expression of CD38+/HLA-DR+ on CD8+ T Cells From Baseline to Week 48
NCT01400412 (20) [back to overview]Percentage Change in Expression of CD38+/HLA-DR+ on CD4+ T Cells From Baseline to Week 48
NCT01400412 (20) [back to overview]Percent Change in Expression of CD28+/CD57+ on CD8+ T Cells From Baseline to Week 48
NCT01400412 (20) [back to overview]Percent Change in Expression of CD28+ on CD8+ T Cells From Baseline to Week 48
NCT01400412 (20) [back to overview]CD8+ T-cell Change From Baseline to Week 48
NCT01400412 (20) [back to overview]Change in Levels of sCD163 From Baseline to Week 48
NCT01400412 (20) [back to overview]Change in Levels of sCD14 From Baseline
NCT01400412 (20) [back to overview]Change in Level of IP-10 From Baseline to Week 48
NCT01400412 (20) [back to overview]Number of Participants Who Developed Grade 3 or 4 Primary Adverse Events
NCT01400412 (20) [back to overview]Number of Participants Who Died During the Study
NCT01400412 (20) [back to overview]Change in Levels of IL-6 From Baseline to Week 48
NCT01422369 (2) [back to overview]NK-104 AUC
NCT01422369 (2) [back to overview]Number of Participants With at Least One Adverse Event.
NCT01423812 (7) [back to overview]Number of Participants With Greater Than 95% Adherence at 48 Weeks
NCT01423812 (7) [back to overview]Secondary Efficacy Endpoints
NCT01423812 (7) [back to overview]Secondary Efficacy Endpoints
NCT01423812 (7) [back to overview]Primary Efficacy Endpoint for Virologic Suppression in HIV-infected Subjects
NCT01423812 (7) [back to overview]Change in Total Cholesterol From Baseline to 48 Weeks
NCT01423812 (7) [back to overview]Absolute Value Change in CD4+ From Baseline to Week 48
NCT01423812 (7) [back to overview]Assessment of Virologic Failure
NCT01426958 (3) [back to overview]Area Under Curve From 0 to ∞ Hours (AUC0-∞)
NCT01426958 (3) [back to overview]Area Under Curve From 0 to tz (AUC0-tz)
NCT01426958 (3) [back to overview]Maximum Concentration (Cmax)
NCT01448707 (7) [back to overview]Change From Baseline in Global Neurocognitive Performance z-Score
NCT01448707 (7) [back to overview]Virologic Response (Food Drug and Administration [FDA] Snapshot, Switch = Failure)
NCT01448707 (7) [back to overview]Virologic Response (Food Drug and Administration [FDA] Snapshot, Switch = Failure)
NCT01448707 (7) [back to overview]Virologic Response (FDA Snapshot, Switch Included)
NCT01448707 (7) [back to overview]Time to Loss of Virologic Response
NCT01448707 (7) [back to overview]Number of Participants Reporting Resistance Mutations With Confirmed Virologic Failure Who Have HIV RNA >400 Copies/mL and Genotype Resistance Results
NCT01448707 (7) [back to overview]Number of Participants Reporting Treatment-Emergent Phenotypic Drug Resistance
NCT01449929 (16) [back to overview]Time to Virologic Suppression (<50 Copies/mL) Through Week 48
NCT01449929 (16) [back to overview]Change From Baseline in Fasting Low-density Lipoprotein (LDL) Cholesterol Through Week 48
NCT01449929 (16) [back to overview]Number of Participants (Par.) With Detectable Virus That Has Genotypic or Phenotypic Evidence of Treatment-emergent Resistance to DTG, DRV+RTV and Other On-study ART at Time of Protocol Defined Virology Failure (PDVF)
NCT01449929 (16) [back to overview]Percentage of Participants With Grade 2 or Higher Abnormalities in Fasting LDL Cholesterol Through Week 48
NCT01449929 (16) [back to overview]Percentage of Participants With Plasma HIV-1 RNA <400 c/mL at Week 48
NCT01449929 (16) [back to overview]Percentage of Participants With Plasma Human Immunodeficiency Virus-1 (HIV-1) Ribonucleic Acid (RNA) <50 Copies/Milliliter (c/mL) at Week 48
NCT01449929 (16) [back to overview]Change From Baseline in Acquired Immune Deficiency Syndrome (AIDS) Clinical Trials Group (ACTG) Symptom Distress Module (SDM) Bother Score at Week 4, Week 24, and Week 48
NCT01449929 (16) [back to overview]Change From Baseline in CD4+ and CD8+ Cell Counts
NCT01449929 (16) [back to overview]Change From Baseline in EQ-5D Thermometer Scores at Week 24 and Week 48
NCT01449929 (16) [back to overview]Change From Baseline in European Quality of Life -5 Dimensions (EQ-5D) Utility Scores at Week 24 and Week 48
NCT01449929 (16) [back to overview]Number of Participants With HIV-1 Associated Disease Progression With the Indicated Shift to CDC Class C, or New CDC Class C or Death at Week 48
NCT01449929 (16) [back to overview]Human Immunodeficiency Virus Treatment Satisfaction Questionnaire (HIVTSQ) Convenience Score at Week 4, Week 24, and Week 48
NCT01449929 (16) [back to overview]Human Immunodeficiency Virus Treatment Satisfaction Questionnaire (HIVTSQ) Lifestyle/Ease Sub Score at Week 4, Week 24, and Week 48
NCT01449929 (16) [back to overview]Human Immunodeficiency Virus Treatment Satisfaction Questionnaire (HIVTSQ) Total Score at Week 4, Week 24, and Week 48
NCT01449929 (16) [back to overview]Number of Participants With the Indicated Grade 3 and Grade 4 Maximum Post-Baseline Chemistry and Hematology Laboratory Toxicities
NCT01449929 (16) [back to overview]Change From Baseline in Plasma HIV-1 RNA (log10 c/mL) at Weeks 4, 8, 12, 16, 24, 36 and 48
NCT01458535 (7) [back to overview]Percentage of Participants With Sustained Virologic Response 12 Weeks (SVR12) Post-treatment
NCT01458535 (7) [back to overview]Percentage of Participants With Hepatitis C Virus (HCV) Ribonucleic Acid (RNA) Below the Lower Limit of Quantitation (LLOQ) at Week 4 Rapid Virologic Response (RVR)
NCT01458535 (7) [back to overview]Percentage of Participants With Hepatitis C Virus (HCV) Ribonucleic Acid (RNA) < 1000 International Units Per Milliliter (IU/mL)
NCT01458535 (7) [back to overview]Percentage of Participants Who Experienced Virologic Relapse Through End of Post Treatment Period (up to 48 Weeks)
NCT01458535 (7) [back to overview]Percentage of Participants With Hepatitis C Virus (HCV) Ribonucleic Acid (RNA) Suppressed Below the Lower Limit of Quantitation (LLOQ) From Week 4 Through Week 12 [(Extended Rapid Virologic Response (eRVR)]
NCT01458535 (7) [back to overview]Percentage of Participants With Virologic Failure During Treatment
NCT01458535 (7) [back to overview]Percentage of Participants With Sustained Virologic Response 24 Weeks (SVR24) Post-Treatment
NCT01464827 (6) [back to overview]Percentage of Participants With Sustained Virologic Response 24 Weeks Post-dose for 8 Weeks Versus 12 Weeks of Treatment With 3 DAAs and Ribavirin
NCT01464827 (6) [back to overview]Percentage of Participants With Sustained Virologic Response 24 Weeks Post-dose in Treatment-naïve Versus Null-responders
NCT01464827 (6) [back to overview]Number of Participants With Adverse Events (AEs)
NCT01464827 (6) [back to overview]Percentage of Participants With Sustained Virologic Response 24 Weeks Post-dose Following Treatment of Different Durations With 3 Direct-acting Antiviral Agents (DAAs) and Ribavirin
NCT01464827 (6) [back to overview]Percentage of Participants With Sustained Virologic Response 24 Weeks Post-dose Following Treatment for 12 Weeks With 3 DAAs With Versus Without Ribavirin
NCT01464827 (6) [back to overview]Percentage of Participants With Sustained Virologic Response 24 Weeks Post-dose Following Treatment for 12 Weeks With 2 DAAs and Ribavirin Versus 3 DAAs and Ribavirin
NCT01511809 (1) [back to overview]Proportion of Patients With Treatment Failure (TF)
NCT01513122 (6) [back to overview]Mean Bone Mineral Density Changes From Baseline to 48 Weeks as Measured by DXA Scan
NCT01513122 (6) [back to overview]Mean Glucose Changes From Baseline to 48 Weeks
NCT01513122 (6) [back to overview]Mean Limbs Fat Changes From Baseline to 48 Weeks as Measured by DXA Scan
NCT01513122 (6) [back to overview]Mean Total Body Fat Changes From Baseline to 48 Weeks as Measured by DXA Scan
NCT01513122 (6) [back to overview]Mean Total Cholesterol Changes From Baseline to 48 Weeks
NCT01513122 (6) [back to overview]Mean Triglycerides Changes From Baseline to 48 Weeks
NCT01516970 (4) [back to overview]Worst Sheehan Disability Scale (SDS) Score for the Safety Population
NCT01516970 (4) [back to overview]Percentage of Participants Who Developed Detectable HIV Antibodies
NCT01516970 (4) [back to overview]Number of Participants With Early Discontinuation From Randomized Human Immunodeficiency Virus Postexposure Prophylaxis (HIV PEP)
NCT01516970 (4) [back to overview]Number of Participants With Treatment-Emergent Adverse Events (TEAEs)
NCT01563536 (12) [back to overview]Mean Maximal Decrease From Baseline in Hepatitis C Virus (HCV) Ribonucleic Acid (RNA) During ABT-267 Monotherapy
NCT01563536 (12) [back to overview]Percentage of Participants With End-of-Treatment Response
NCT01563536 (12) [back to overview]Percentage of Participants With Rapid Virologic Response
NCT01563536 (12) [back to overview]Time of Maximum Plasma Concentration (Tmax) of ABT-267 Following Monotherapy on Day 1
NCT01563536 (12) [back to overview]Mean Change in Viral Load From Baseline to Pre-dose on Day 2 and Day 3 of ABT-267 Monotherapy
NCT01563536 (12) [back to overview]Percentage of Participants With Extended Rapid Virologic Response
NCT01563536 (12) [back to overview]Resistance-Associated Variants and Phenotypic Resistance
NCT01563536 (12) [back to overview]Number of Participants With Adverse Events (AEs)
NCT01563536 (12) [back to overview]Percentage of Participants With Sustained Virologic Response 12 Weeks and 24 Weeks After Combination Therapy
NCT01563536 (12) [back to overview]Plasma Concentration of ABT-267 Pre-dose (Ctrough) on Day 2 and Day 3
NCT01563536 (12) [back to overview]Maximum Plasma Concentration (Cmax) of ABT-267 Following Monotherapy on Day 1
NCT01563536 (12) [back to overview]Area Under the Plasma Concentration-time Curve From Time 0 to 24 Hours (AUC[24]) of ABT-267 Following Monotherapy on Day 1
NCT01565889 (8) [back to overview]Part A: Plasma Pharmacokinetics of SOF, EFV, TFV, and FTC: Cmax at Day 7
NCT01565889 (8) [back to overview]Incidence of Adverse Events Leading to Permanent Discontinuation of Study Drug(s)
NCT01565889 (8) [back to overview]Part B: Percentage of Participants With Sustained Virologic Response (SVR) at 12 Weeks After Discontinuation of Therapy (SVR12)
NCT01565889 (8) [back to overview]Part A: Plasma Pharmacokinetics of SOF, EFV, Tenofovir (TFV), and FTC: AUCtau at Day 7
NCT01565889 (8) [back to overview]Part B: On-treatment HCV RNA
NCT01565889 (8) [back to overview]Part B: On-treatment HIV RNA
NCT01565889 (8) [back to overview]Part B: Percentage of Participants Experiencing Viral Breakthrough or Viral Relapse
NCT01565889 (8) [back to overview]Part B: Percentage of Participants With Sustained Virologic Response at 4 and 24 Weeks After Discontinuation of Therapy (SVR4 and SVR24)
NCT01601626 (26) [back to overview]Percent of Participants Whose HIV Viral Load Was Less Than 400 Copies/mL at Week 48.
NCT01601626 (26) [back to overview]CD4 Count Change From Baseline to Week 24
NCT01601626 (26) [back to overview]RAL Cmax and Cmin in Participants Enrolled in Arm C
NCT01601626 (26) [back to overview]LPV Cmax and Cmin in Participants Enrolled in Arms A, B, and C
NCT01601626 (26) [back to overview]RBT AUC in Participants Enrolled in Arms A and C
NCT01601626 (26) [back to overview]RAL AUC in Participants Enrolled in Arm C
NCT01601626 (26) [back to overview]Percent of Participants Who Experienced a New AIDS-defining Illness or Died
NCT01601626 (26) [back to overview]RBT Cmax and Cmin in Participants Enrolled in Arms A and C
NCT01601626 (26) [back to overview]Percent of Participants Who Experienced Sputum Conversion at Week 8.
NCT01601626 (26) [back to overview]Percent of Participants Who Experienced HIV Virologic Failure
NCT01601626 (26) [back to overview]Percent of Participants Whose HIV Viral Load Was Less Than 50 Copies/mL at Week 48
NCT01601626 (26) [back to overview]Percent of Participants Who Experienced a New AIDS-defining Illness
NCT01601626 (26) [back to overview]Percent of Participants Who Interrupted or Discontinued at Least One TB Drug Due to Toxicity
NCT01601626 (26) [back to overview]Percent of Participants Who Experienced TB Treatment Failure
NCT01601626 (26) [back to overview]Percent of Participants Who Interrupted or Discontinued at Least One HIV Drug Due to Toxicity
NCT01601626 (26) [back to overview]Percent of Participants Who Died
NCT01601626 (26) [back to overview]Percent of Participants Who Experienced TB Relapse/Recurrence
NCT01601626 (26) [back to overview]Number of Participants Reporting a Grade 3 or 4 Sign or Symptom
NCT01601626 (26) [back to overview]Number of Participants Reporting a Grade 3 or 4 Laboratory Abnormality
NCT01601626 (26) [back to overview]LPV AUC in Participants Enrolled in Arms A, B, and C
NCT01601626 (26) [back to overview]Cumulative Probability of HIV Virologic Failure at Week 72
NCT01601626 (26) [back to overview]CD4 Count Change From Baseline to Week 8
NCT01601626 (26) [back to overview]CD4 Count Change From Baseline to Week 72
NCT01601626 (26) [back to overview]CD4 Count Change From Baseline to Week 48
NCT01601626 (26) [back to overview]Percent of Participants Who Experienced TB Relapse/Recurrence and Who Had TB Drug Resistance
NCT01601626 (26) [back to overview]Number of Participants Who Experienced MTB IRIS
NCT01609933 (4) [back to overview]Percentage of Participants Achieving Virologic Response 24 Weeks Post Treatment (SVR24)
NCT01609933 (4) [back to overview]Percentage of Participants Achieving Sustained Virologic Response 12 Weeks Post Treatment (SVR12)
NCT01609933 (4) [back to overview]Number of Participants With Adverse Events
NCT01609933 (4) [back to overview]Percentage of Participants With Extended Rapid Virologic Response (eRVR)
NCT01632891 (7) [back to overview]Change in log10(Pf Gametocyte Density) From Entry to Day 30
NCT01632891 (7) [back to overview]Change in log10(Pf Parasite Density) From Entry to Day 30
NCT01632891 (7) [back to overview]Number of Participants With Uncomplicated Clinical Malaria
NCT01632891 (7) [back to overview]Proportion of Participants With Plasmodium Falciparum (Pf) Subclinical Parasitemia (SCP) Clearance
NCT01632891 (7) [back to overview]Time to First Pf SCP Clearance
NCT01632891 (7) [back to overview]Log10(Pf Parasite Density)
NCT01632891 (7) [back to overview]Number of Participants With Detectable Pf Gametocyte Density
NCT01662336 (20) [back to overview]Change From Baseline in Health-related Quality of Life Pain Domain Score
NCT01662336 (20) [back to overview]Change From Baseline in Health-related Quality of Life Role Functioning Domain Score
NCT01662336 (20) [back to overview]Change From Baseline in Health-related Quality of Life Social Functioning Domain Score
NCT01662336 (20) [back to overview]Change From Baseline in Patient Perception of Stress
NCT01662336 (20) [back to overview]Change From Baseline in Psychological Well-being
NCT01662336 (20) [back to overview]Cluster of Differentiation 4 (CD4) Positive Cell Counts at Each Visit
NCT01662336 (20) [back to overview]Health Resource Utilization
NCT01662336 (20) [back to overview]Healthcare Provider Satisfaction
NCT01662336 (20) [back to overview]Viral Load at Each Visit
NCT01662336 (20) [back to overview]Change From Baseline in Health-related Quality of Life Physical Functioning Domain Score
NCT01662336 (20) [back to overview]Percentage of Participants Adherent to Treatment at Month 12
NCT01662336 (20) [back to overview]Percentage of Participants Adherent to Treatment at Month 6
NCT01662336 (20) [back to overview]Change From Baseline in Adherence Integration Subscale Score at Months 6 and 12
NCT01662336 (20) [back to overview]Change From Baseline in Adherence Perseverance Subscale Score at Months 6 and 12
NCT01662336 (20) [back to overview]Change From Baseline in Adherence Summative Score at Months 6 and 12
NCT01662336 (20) [back to overview]Change From Baseline in Coping Self-Efficacy
NCT01662336 (20) [back to overview]Change From Baseline in Health-related Quality of Life Cognitive Functioning Domain Score
NCT01662336 (20) [back to overview]Change From Baseline in Health-related Quality of Life Energy/ Fatigue Domain Score
NCT01662336 (20) [back to overview]Change From Baseline in Health-related Quality of Life General Health Perception Domain Score
NCT01662336 (20) [back to overview]Change From Baseline in Health-related Quality of Life Mental Health Domain Score
NCT01667978 (1) [back to overview]AUC Norethindrone
NCT01672983 (4) [back to overview]Percentage of Participants With End of Treatment (EOT) Response
NCT01672983 (4) [back to overview]Percentage of Participants With Sustained Virologic Response 12 Weeks After Treatment (SVR12)
NCT01672983 (4) [back to overview]Percentage of Participants With Sustained Virologic Response 24 Weeks After Treatment (SVR24)
NCT01672983 (4) [back to overview]Number of Participants With Adverse Events (AEs)
NCT01691794 (3) [back to overview]Number of Participants Who Died and With Serious Adverse Events (SAEs), Adverse Events (AEs) Leading to Discontinuation, Grade 2-4 Related AEs, Grade 3-4 AEs, and Centers for Disease Control (CDC) Class C AIDS Events
NCT01691794 (3) [back to overview]Number of Participants With Laboratory Test Results Meeting the Criteria for Abnormal, Grades 1-4
NCT01691794 (3) [back to overview]Number of Participants With Laboratory Test Results Meeting the Criteria for Abnormal, Grades 1-4 (Continued)
NCT01695954 (3) [back to overview]AUC
NCT01695954 (3) [back to overview]GMR of Cmax of Pitavastatin When Coadministered With Efavirenz or With Darunavir/Ritonavir
NCT01695954 (3) [back to overview]GMR of 24- Hour AUC of Pitavastatin When Coadministered With Efavirenz or With Darunavir/Ritonavir Over 24 Hour AUC of Pitavastatin
NCT01773070 (5) [back to overview]Percentage of Participants Who Experienced Relapse˅Overall Without and With New HCV Infection
NCT01773070 (5) [back to overview]Percentage of Participants Who Experienced Relapse12 Without and With New HCV Infection
NCT01773070 (5) [back to overview]Percentage of Participants Who Experienced Relapse12overall With and Without New HCV Infection
NCT01773070 (5) [back to overview]Percentage of Participants Who Experienced Relapse24 Without and With New HCV Infection
NCT01773070 (5) [back to overview]Number of HCV Genotype (GT)1a-Infected Participants With Persistence of Treatment-Emergent Substitutions in NS3, NS5A, or NS5B
NCT01782495 (4) [back to overview]Percentage of Participants With Sustained Virologic Response 24 Weeks Post-treatment (SVR24)
NCT01782495 (4) [back to overview]Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT01782495 (4) [back to overview]Percentage of Participants With On-treatment Virologic Failure
NCT01782495 (4) [back to overview]Percentage of Participants With Post-treatment Relapse
NCT01803074 (28) [back to overview]Change in Plasma Log10 HIV-1 Ribonucleic Acid (RNA) Levels From Baseline to Day 11
NCT01803074 (28) [back to overview]Area Under The Plasma Concentration - Time Curve Over the Dosing Interval (AUC([Tau]) - Part A and C
NCT01803074 (28) [back to overview]Maximum Decline From Baseline in Log10 HIV-1 RNA - Part B
NCT01803074 (28) [back to overview]Maximum Decline From Baseline in Log10 HIV-1 RNA - Part A and C
NCT01803074 (28) [back to overview]Number of Participants With Clinically Significant Changes in Heart Rate
NCT01803074 (28) [back to overview]Plasma Half-life: Part A and C
NCT01803074 (28) [back to overview]Time to Reach Maximum Plasma Concentration (Tmax) - Part A and C
NCT01803074 (28) [back to overview]Plasma Concentration 24 Hours Post-Dose (C24) - Part B
NCT01803074 (28) [back to overview]Plasma Concentration 24 Hours Post-Dose (C24) - Part A and C
NCT01803074 (28) [back to overview]Percent Change From Baseline in Cluster of Differentiation (CD) 4+ and CD8+ Lymphocyte Percent - Part B
NCT01803074 (28) [back to overview]Area Under The Plasma Concentration - Time Curve Over the Dosing Interval (AUC[Tau]) - Part B
NCT01803074 (28) [back to overview]Percent Change From Baseline in Cluster of Differentiation (CD) 4+ and CD8+ Lymphocyte Percent - Part A and C
NCT01803074 (28) [back to overview]Degree of Fluctuation (DF): Part A and C
NCT01803074 (28) [back to overview]Number of Participants With Death as Outcome, Serious Adverse Events (SAEs), Related SAEs, Discontinuations Due to SAEs, Adverse Events (AEs), and Discontinuations Due to AEs During the Study
NCT01803074 (28) [back to overview]Number of Participants With Clinically Significant Grade 3/4 Laboratory Abnormalities From Baseline
NCT01803074 (28) [back to overview]Time to Maximum Decline in Log 10 HIV-1 RNA - Part A and C
NCT01803074 (28) [back to overview]Number of Participants With Clinically Significant Changes in Electrocardiogram (ECG)
NCT01803074 (28) [back to overview]Number of Participants With Abnormal Changes in Physical Examination
NCT01803074 (28) [back to overview]Maximum Observed Plasma Concentrations (Cmax) - Part B
NCT01803074 (28) [back to overview]Time to Maximum Decline in Log 10 HIV-1 RNA - Part B
NCT01803074 (28) [back to overview]Maximum Observed Plasma Concentrations (Cmax) - Part A and C
NCT01803074 (28) [back to overview]Number of Participants With Clinically Significant Changes in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)
NCT01803074 (28) [back to overview]Change From Baseline in Cluster of Differentiation (CD) 4+ and CD8+ Lymphocyte Counts - Part A and C
NCT01803074 (28) [back to overview]Apparent Total Body Clearance: Part A and C
NCT01803074 (28) [back to overview]Average Observed Plasma Concentration at Steady State (Css-avg): Part A and C
NCT01803074 (28) [back to overview]Accumulation Index (AI): Part A and C
NCT01803074 (28) [back to overview]Time to Reach Maximum Plasma Concentration (Tmax) - Part B
NCT01803074 (28) [back to overview]Change From Baseline in Cluster of Differentiation (CD) 4+ and CD8+ Lymphocyte Counts - Part B
NCT01818258 (17) [back to overview]Change in CD4 Percent
NCT01818258 (17) [back to overview]Change in HIV Viral Load From Baseline
NCT01818258 (17) [back to overview]Change in Mid-upper Arm Circumference
NCT01818258 (17) [back to overview]Change in WHO Weight-for-height Z-score
NCT01818258 (17) [back to overview]Free Fraction of LPV at Hour 2 Post Dose
NCT01818258 (17) [back to overview]Minimum Trough Concentration (Ctrough) of Lopinavir
NCT01818258 (17) [back to overview]Plasma Clearance of Lamivudine
NCT01818258 (17) [back to overview]Plasma Clearance of Lopinavir
NCT01818258 (17) [back to overview]Plasma Clearance of Ritonavir
NCT01818258 (17) [back to overview]Plasma Clearance of Zidovudine
NCT01818258 (17) [back to overview]Steady-state Lamivudine Area Under the Curve
NCT01818258 (17) [back to overview]Steady-state Ritonavir Area Under the Curve
NCT01818258 (17) [back to overview]Steady-state Zidovudine Area Under the Curve
NCT01818258 (17) [back to overview]HIV Viral Load <400 Copies/mL
NCT01818258 (17) [back to overview]Grade 3 or Higher Adverse Events Related to Study Drugs Through Week 24
NCT01818258 (17) [back to overview]Grade 3 or Higher Adverse Events Through 24 Weeks
NCT01818258 (17) [back to overview]Steady-state Lopinavir Area Under the Curve
NCT01869634 (4) [back to overview]Change in Percentage of Total Artery Diameter
NCT01869634 (4) [back to overview]Change in Systemic Immune Activation
NCT01869634 (4) [back to overview]Number of CD4+ T-cells in the Lamina Propria/mm2 Before and After 12 Months of Therapy Compared to Age-matched Control Volunteers Without HIV
NCT01869634 (4) [back to overview]Number of CD4+ T-cells in the Lamina Propria/mm2 Before and After 12 Months of Therapy Compared to Age-matched Control Volunteers Without HIV
NCT01903031 (21) [back to overview]RTV PK Parameter CLss/F Determined Based on RTV Levels From Individual Participants Enrolled in Arm C
NCT01903031 (21) [back to overview]Ritonavir (RTV) PK Parameter AUC(0-24h) Calculated Based on Intensive RTV PK Samples Obtained From Individual Participants Enrolled in Arm C
NCT01903031 (21) [back to overview]EFV PK Parameter Maximum Plasma Concentration (Cmax) Determined Based on EFV Levels From Individual Participants Enrolled in Arm B
NCT01903031 (21) [back to overview]Proportion of Participants With Progesterone Levels Greater Than 5 ng/mL.
NCT01903031 (21) [back to overview]EFV PK Parameter Clearance (CLss/F) Determined Based on EFV Levels From Individual Participants Enrolled in Arm B
NCT01903031 (21) [back to overview]EFV PK Parameter Area Under the Concentration-Time Curve (AUC0-24hours) Calculated Based on Intensive EFV PK Samples Obtained From Individual Participants Enrolled in Arm B
NCT01903031 (21) [back to overview]ATV PK Parameter Time to Cmax (Tmax) Determined Based on ATV Levels From Individual Participants Enrolled in Arm C
NCT01903031 (21) [back to overview]ATV PK Parameter Cmin Determined Based on ATV Levels From Individual Participants Enrolled in Arm C
NCT01903031 (21) [back to overview]Etonogestrel Concentrations Obtained on Study Days 7 and 14
NCT01903031 (21) [back to overview]ATV PK Parameter CLss/F Determined Based on ATV Levels From Individual Participants Enrolled in Arm C
NCT01903031 (21) [back to overview]Ethinyl Estradiol Concentrations Obtained on Study Days 7 and 14.
NCT01903031 (21) [back to overview]EFV PK Parameter Minimum Plasma Concentration (Cmin) Determined Based on EFV Levels From Individual Participants Enrolled in Arm B
NCT01903031 (21) [back to overview]ATV PK Parameter Cmax Determined Based on ATV Levels From Individual Participants Enrolled in Arm C
NCT01903031 (21) [back to overview]Proportion of Participants With Plasma HIV-1 RNA Levels <40 Copies/mL
NCT01903031 (21) [back to overview]RTV PK Parameter Tmax Determined Based on RTV Levels From Individual Participants Enrolled in Arm C
NCT01903031 (21) [back to overview]RTV PK Parameter Cmin Determined Based on RTV Levels From Individual Participants Enrolled in Arm C
NCT01903031 (21) [back to overview]Ethinyl Estradiol Concentrations at Study Day 21
NCT01903031 (21) [back to overview]Etonogestrel Concentrations at Study Day 21
NCT01903031 (21) [back to overview]Percentage of Participants With Signs and Symptoms of Grade 2 or Higher Deemed Possibly, Probably or Definitely Related to Study Treatment
NCT01903031 (21) [back to overview]ATV PK Parameter AUC(0-24h) Calculated Based on Intensive Atazanavir (ATV) PK Samples Obtained From Individual Participants Enrolled in Arm C
NCT01903031 (21) [back to overview]RTV PK Parameter Cmax Determined Based on RTV Levels From Individual Participants Enrolled in Arm C
NCT01910402 (48) [back to overview]Number of Participants With Post-Baseline HIV-1 Disease Progression-Randomized Phase
NCT01910402 (48) [back to overview]Number of Participants With Treatment Emergent Resistances for ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200mg QD (Randomized Phase)
NCT01910402 (48) [back to overview]Number of Participants With Treatment Emergent Resistances for DTG 50 mg/ABC 600 mg/3TC 300 mg QD (Randomized + Continuation Phase)
NCT01910402 (48) [back to overview]Percentage of Participants With Plasma HIV-1 RNA <50 and <400 c/mL Over Time-Randomized Phase
NCT01910402 (48) [back to overview]Percentage of Participants With Plasma HIV-1 RNA <50 c/mL in Continuation Phase
NCT01910402 (48) [back to overview]Percentage of Participants With Plasma HIV-1 RNA <50 Copies/mL at Week 48 by Subgroups
NCT01910402 (48) [back to overview]Percentage of Participants With Plasma HIV-1 RNA <50 Copies/mL at Week 48 by Subgroups
NCT01910402 (48) [back to overview]Change From Baseline in Plasma HIV-1 RNA at Indicated Time Points-Continuation Phase
NCT01910402 (48) [back to overview]Change From Baseline in TC/HDL Ratio at Week 48
NCT01910402 (48) [back to overview]Change From Baseline in Triglycerides at Week 48
NCT01910402 (48) [back to overview]Number of Participants Who Withdrew From Treatment Due to AEs-Continuation Phase
NCT01910402 (48) [back to overview]Number of Participants Who Withdrew From Treatment Due to AEs-Randomized Phase
NCT01910402 (48) [back to overview]Percentage of Participants With Plasma HIV-1 RNA <50 Copies/mL at Week 48
NCT01910402 (48) [back to overview]Absolute Values in CD4+ Cell Count at Indicated Timepoints-Continuation Phase
NCT01910402 (48) [back to overview]Absolute Values in CD4+ Cell Count at Indicated Timepoints-Randomized Phase
NCT01910402 (48) [back to overview]Absolute Values in Plasma HIV-1 RNA at Indicated Time Points-Continuation Phase
NCT01910402 (48) [back to overview]Absolute Values in Plasma HIV-1 RNA at Indicated Time Points-Randomized Phase
NCT01910402 (48) [back to overview]Bone Specific Alkaline Phosphatase, Osteocalcin, Procollagen 1 N-terminal Propeptide, Type 1 Collagen C-Telopeptide, Vitamin D Ratio of Week 48 Results Over Baseline
NCT01910402 (48) [back to overview]Change From Baseline at Week 48 in SF-12 Total Score, MCS and PCS
NCT01910402 (48) [back to overview]Change From Baseline in Alanine Aminotransferase, Alkaline Phosphatase, Aspartate Aminotransferase, Creatine Kinase at Indicated Time Points
NCT01910402 (48) [back to overview]Change From Baseline in Albumin at Indicated Timepoints
NCT01910402 (48) [back to overview]Change From Baseline in Basophils, Eosinophils, Lymphocytes, Monocytes at Indicated Time Points
NCT01910402 (48) [back to overview]Change From Baseline in Bilirubin and Creatinine at Indicated Timepoints
NCT01910402 (48) [back to overview]Change From Baseline in Bone Specific Alkaline Phosphatase, Osteocalcin and Procollagen 1 N-terminal Propeptide at Indicated Timepoints
NCT01910402 (48) [back to overview]Change From Baseline in Carbon Dioxide, Electrolytes, Lipids, Glucose, Urea at Indicated Time Points
NCT01910402 (48) [back to overview]Change From Baseline in Carbon Dioxide, Electrolytes, Lipids, Glucose, Urea at Indicated Time Points
NCT01910402 (48) [back to overview]Change From Baseline in CD4+ Cell Count at Indicated Timepoints-Continuation Phase
NCT01910402 (48) [back to overview]Change From Baseline in CD4+ Cell Count at Indicated Timepoints-Randomized Phase
NCT01910402 (48) [back to overview]Change From Baseline in Creatinine Clearance at Indicated Time Points
NCT01910402 (48) [back to overview]Change From Baseline in Erythrocyte Mean Corpuscular Volume at Indicated Time Points
NCT01910402 (48) [back to overview]Change From Baseline in Erythrocytes at Indicated Time Points
NCT01910402 (48) [back to overview]Change From Baseline in Hematocrit Count at Indicated Time Points
NCT01910402 (48) [back to overview]Change From Baseline in Lipase at Indicated Timepoints
NCT01910402 (48) [back to overview]Change From Baseline in Plasma HIV-1 RNA at Indicated Time Points-Randomized Phase
NCT01910402 (48) [back to overview]Change From Baseline in Total CHLS/HDL CHLS Ratio at Indicated Timepoints
NCT01910402 (48) [back to overview]Change From Baseline in Type I Collagen C-telopeptides at Indicated Timepoints
NCT01910402 (48) [back to overview]Change From Baseline in Urine Albumin Creatinine Ratio at Indicated Time Points
NCT01910402 (48) [back to overview]Change From Baseline in Vitamin D, Vitamin D2 and Vitamin D3 at Week 24 and Week 48
NCT01910402 (48) [back to overview]HIVTSQs Total Score at Indicated Timepoints
NCT01910402 (48) [back to overview]Number of Participants With AEs by Maximum Toxicity-Continuation Phase
NCT01910402 (48) [back to overview]Number of Participants With AEs by Maximum Toxicity-Randomized Phase
NCT01910402 (48) [back to overview]Number of Participants With Any Adverse Events (AEs), and Serious Adverse Events (SAEs)-Randomized Phase
NCT01910402 (48) [back to overview]Number of Participants With Any AEs, and SAEs in Continuation Phase
NCT01910402 (48) [back to overview]Number of Participants With Maximum Post-Baseline Emergent Chemistry Toxicities-Continuation Phase
NCT01910402 (48) [back to overview]Number of Participants With Maximum Post-Baseline Emergent Chemistry Toxicities-Randomized Phase
NCT01910402 (48) [back to overview]Number of Participants With Maximum Post-Baseline Emergent Hematology Toxicities-Continuation Phase
NCT01910402 (48) [back to overview]Number of Participants With Maximum Post-Baseline Emergent Hematology Toxicities-Randomized Phase
NCT01910402 (48) [back to overview]Number of Participants With Post-Baseline HIV-1 Disease Progression for DTG 50 mg/ABC 600 mg/3TC 300 mg QD (Randomized + Continuation Phase)
NCT01939197 (14) [back to overview]Percentage of Participants in Part 1a With Plasma HIV-1 RNA Suppression at End of Treatment and 12 Weeks Post-Treatment
NCT01939197 (14) [back to overview]Percentage of Participants in Part 1b With On-Treatment HCV Virologic Failure During the Treatment Period
NCT01939197 (14) [back to overview]Percentage of Participants in Part 1b Achieving SVR12
NCT01939197 (14) [back to overview]Percentage of Participants in Part 1a With Relapse12
NCT01939197 (14) [back to overview]Percentage of Participants in Part 1b With Relapse12 for Each Arm and Overall
NCT01939197 (14) [back to overview]Percentage of Participants in Part 2 With Relapse12
NCT01939197 (14) [back to overview]Percentage of Participants in Arm F and Arm G of Part 2 Achieving SVR12
NCT01939197 (14) [back to overview]Percentage of Participants in Part 1a With On-Treatment HCV Virologic Failure During the Treatment Period
NCT01939197 (14) [back to overview]Percentage of Participants in GT1 Analysis Group 1 in Part 2 Achieving Sustained Virologic Response 12 Weeks Post-Treatment (SVR12)
NCT01939197 (14) [back to overview]Percentage of Participants in Part 2 With Plasma HIV-1 RNA Suppression at End of Treatment and 12 Weeks Post-Treatment
NCT01939197 (14) [back to overview]Percentage of Participants in Part 1b With Plasma HIV-1 RNA Suppression at End of Treatment and 12 Weeks Post-Treatment
NCT01939197 (14) [back to overview]Percentage of Participants With GT4 HCV in Part 2 Achieving SVR12, by Arm and Overall
NCT01939197 (14) [back to overview]Percentage of Participants in Part 1a Achieving SVR12
NCT01939197 (14) [back to overview]Percentage of Participants in Part 2 With On-Treatment HCV Virologic Failure During the Treatment Period
NCT02023099 (7) [back to overview]Percentage of Non-cirrhotic Treatment-Naïve Participants Who Are Eligible for Interferon (IFN)-Based Therapy and Who Have High Viral Load in the DB Active Treatment Group With a Sustained Virologic Response 12 Weeks Post-treatment
NCT02023099 (7) [back to overview]Percentage of Participants in the Active Treatment Group With On-treatment Virologic Failure During Treatment
NCT02023099 (7) [back to overview]Percentage of Participants in the Active Treatment Group With Sustained Virologic Response 12 Weeks Post-treatment
NCT02023099 (7) [back to overview]Percentage of Participants in Substudy 1 Arm A Active Treatment Group With Post-treatment Relapse, by Subpopulation
NCT02023099 (7) [back to overview]Percentage of Participants in Substudy 1 Arm A Active Treatment Group With Sustained Virologic Response 12 Weeks Post-Treatment, by Subpopulation
NCT02023099 (7) [back to overview]Percentage of Participants in the Substudy 1 Arm A Active Treatment Group With On-treatment Virologic Failure During Treatment, by Subpopulation
NCT02023099 (7) [back to overview]Percentage of Participants in the Active Treatment Group With Post-treatment Relapse
NCT02023112 (6) [back to overview]Percentage of Participants With SVR12 Weeks Post-treatment for Each Treatment Arm Within Different Subpopulations
NCT02023112 (6) [back to overview]Percentage of Participants in Each Treatment Arm With On-treatment Virologic Failure During the Treatment Period
NCT02023112 (6) [back to overview]Percentage of Participants With Post-treatment Relapse
NCT02023112 (6) [back to overview]Percentage of Non-cirrhotic, Treatment-naive Participants in Each Treatment Group With a Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02023112 (6) [back to overview]Percentage of Participants in Each Treatment Arm With On-treatment Virologic Failure During the Treatment Period for Each Treatment Arm Within Different Subpopulations
NCT02023112 (6) [back to overview]Percentage of Participants With Post-treatment Relapse Within Different Subpopulations
NCT02068222 (4) [back to overview]The Percentage of Subjects With Post-Treatment Relapse
NCT02068222 (4) [back to overview]The Percentage of Subjects With Virologic Failure During Treatment
NCT02068222 (4) [back to overview]The Percentage of Subjects Who Achieve 24-week Sustained Virologic Response (SVR24)
NCT02068222 (4) [back to overview]The Percentage of Subjects Who Achieve 12-week Sustained Virologic Response (SVR12)
NCT02116660 (1) [back to overview]Change From Baseline in Estimated Glomerular Filtration Rate (eGFR)
NCT02155101 (3) [back to overview]HIV-1 RNA Viral Load
NCT02155101 (3) [back to overview]HIV-1 RNA Viral Load
NCT02155101 (3) [back to overview]HIV-1 RNA Viral Load
NCT02159352 (11) [back to overview]Plasma Concentration Observed at 24 Hours Postdose (C24) of Daclatasvir
NCT02159352 (11) [back to overview]Time of Maximum Observed Plasma Concentration (Tmax) of Daclatasvir
NCT02159352 (11) [back to overview]Dose-normalized Maximum Observed Plasma Concentration (Cmax/D) and Dose-normalized Plasma Concentration Observed at 24 Hours Postdose (C24/D) of Daclatasvir
NCT02159352 (11) [back to overview]Number of Participants With Abnormalities in Electrocardiogram (ECG) Findings
NCT02159352 (11) [back to overview]Number of Participants With Serious Adverse Events (SAEs) and Discontinuations Due to Adverse Events (AEs) and Who Died
NCT02159352 (11) [back to overview]Dose-normalized Area Under the Concentration-Time Curve in 1 Dosing Interval (AUC[TAU]/D) of Daclatasvir
NCT02159352 (11) [back to overview]Maximum Observed Plasma Concentration (Cmax) for Daclatasvir
NCT02159352 (11) [back to overview]Number of Participants With Abnormalities in Vital Sign Measurements
NCT02159352 (11) [back to overview]Number of Participants With Marked Abnormalities in Hematology Laboratory Test Results
NCT02159352 (11) [back to overview]Area Under the Concentration-Time Curve in 1 Dosing Interval (AUC[TAU]) for Daclatasvir
NCT02159352 (11) [back to overview]Number of Participants With Abnormalities in Urinalysis and Other Chemistry Testing Results
NCT02167945 (11) [back to overview]Mean Change From Baseline in Short Form 36 Version 2.0 (SF-36 V2) Mental Component Summary (MCS) Score at Post-Treatment Week 12 and Post-Treatment Week 24
NCT02167945 (11) [back to overview]Mean Change From Baseline in Short Form 36 Version 2.0 (SF-36 V2) Physical Component Summary (PCS) Score at Post-Treatment Week 12 and Post-Treatment Week 24
NCT02167945 (11) [back to overview]Liver Decompensation: Time to Event
NCT02167945 (11) [back to overview]Liver Transplantation: Time to Event
NCT02167945 (11) [back to overview]Liver-Related Death: Time to Event
NCT02167945 (11) [back to overview]All-Cause Death: Time to Event
NCT02167945 (11) [back to overview]Hepatocellular Carcinoma: Time to Event
NCT02167945 (11) [back to overview]Treatment Compliance: Percentage of Tablets Taken Relative to the Total Tablets
NCT02167945 (11) [back to overview]Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02167945 (11) [back to overview]Mean Change From Baseline in Functional Assessment of Chronic Illness Therapy - Fatigue (FACIT-F) Total Score at Post-Treatment Week 12 and Post-Treatment Week 24
NCT02167945 (11) [back to overview]All-Cause Death, Liver-Related Death, Liver Decompensation, Liver Transplantation, Hepatocellular Carcinoma: Time to Event
NCT02194998 (13) [back to overview]Number of Participants With Selected HIV-1 Resistance Mutations Among Participants Who Experience HIV-1 Virologic Failure (VF)
NCT02194998 (13) [back to overview]Percentage of Participants With Sustained Virologic Response at 12 Weeks After HCV Treatment Discontinuation (SVR12)
NCT02194998 (13) [back to overview]Percentage of Participants With Sustained Virologic Response at 24 Weeks After HCV Treatment Discontinuation (SVR24)
NCT02194998 (13) [back to overview]Change in IP-10 Concentration.
NCT02194998 (13) [back to overview]Change in Soluble CD14 (sCD14)
NCT02194998 (13) [back to overview]Levels of IP-10 Concentration.
NCT02194998 (13) [back to overview]Levels of Soluble CD14 (sCD14)
NCT02194998 (13) [back to overview]Number of Participants Who Experienced HIV-1 Virologic Failure (VF)
NCT02194998 (13) [back to overview]Number of Participants Who Prematurely Discontinued HCV Study Treatment for Any Reason Other Than HCV Virologic Failure (VF)
NCT02194998 (13) [back to overview]Number of Participants With an Occurrence of Diagnoses Leading to Premature HCV Study Treatment or HIV-1 Antiretroviral (ARV) Discontinuation.
NCT02194998 (13) [back to overview]Number of Participants With an Occurrence of Laboratory Abnormality Grade 3 or Higher.
NCT02194998 (13) [back to overview]Number of Participants With an Occurrence of Serious Adverse Events (SAEs) as Defined by International Conference on Harmonisation (ICH) Criteria
NCT02194998 (13) [back to overview]Number of Participants With an Occurrence of Signs/Symptoms Grade 3 or Higher
NCT02207088 (3) [back to overview]Percentage of Participants With Sustained Virologic Response 12 (SVR12) Weeks Post-treatment
NCT02207088 (3) [back to overview]Percentage of Participants With On-treatment Virologic Failure
NCT02207088 (3) [back to overview]Percentage of Participants With Post-Treatment Relapse
NCT02216422 (3) [back to overview]Percentage of Participants With Sustained Virologic Response 12 Weeks (SVR12) Post-treatment
NCT02216422 (3) [back to overview]Percentage of Participants With Post-Treatment Relapse
NCT02216422 (3) [back to overview]Percentage of Participants With On-Treatment Virologic Failure
NCT02219477 (9) [back to overview]Percentage of Participants With Improvement From Baseline to Post-Treatment Week 12 in Hepatic Function Tests
NCT02219477 (9) [back to overview]Percentage of Participants With Improvement From Baseline to Post-Treatment Week 12 in Hepatic Function Tests
NCT02219477 (9) [back to overview]Percentage of Participants With SVR12 in Group 3
NCT02219477 (9) [back to overview]Percentage of Participants With Improvement From Baseline to Post-Treatment Week 12 in FibroTest
NCT02219477 (9) [back to overview]Percentage of Participants With Improvement From Baseline to Post-Treatment Week 12 in Model for End-Stage Liver Disease (MELD) Score
NCT02219477 (9) [back to overview]Percentage of Participants With SVR12 Non-Response Due to Experiencing On-Treatment Virologic Failure
NCT02219477 (9) [back to overview]Percentage of Participants With SVR12 Non-Response Due to Experiencing Relapse˅12
NCT02219477 (9) [back to overview]Percentages of Participants With Sustained Virologic Response 12 Weeks Post-Treatment (SVR12) in Group 1 and in Group 2
NCT02219477 (9) [back to overview]Percentage of Participants With Improvement From Baseline to Post-Treatment Week 12 in Chld-Pugh Score
NCT02219490 (8) [back to overview]Liver Decompensation: Time to Event
NCT02219490 (8) [back to overview]All-Cause Death, Liver-Related Death, Liver Decompensation, Liver Transplantation, Hepatocellular Carcinoma: Time to Event
NCT02219490 (8) [back to overview]Liver Transplantation: Time to Event
NCT02219490 (8) [back to overview]Hepatocellular Carcinoma: Time to Event
NCT02219490 (8) [back to overview]Liver-Related Death: Time to Event
NCT02219490 (8) [back to overview]Change From Baseline in FibroScan Score by SVR12 Status
NCT02219490 (8) [back to overview]All-Cause Death: Time to Event
NCT02219490 (8) [back to overview]Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02219503 (3) [back to overview]Percentage of Participants With Sustained Virologic Response 12 Weeks (SVR12) Post-treatment
NCT02219503 (3) [back to overview]Percentage of Participants With On-Treatment Virologic Failure
NCT02219503 (3) [back to overview]Percentage of Participants With Post-Treatment Relapse
NCT02223351 (13) [back to overview]Half-Life (t1/2) of ASP2151
NCT02223351 (13) [back to overview]Apparent Total Body Clearance (CL/F) From Plasma of ASP1955888-00
NCT02223351 (13) [back to overview]Apparent Total Body Clearance (CL/F) of ASP2151 From Plasma
NCT02223351 (13) [back to overview]Apparent Volume of Distribution (Vd/F) of ASP1955888-00
NCT02223351 (13) [back to overview]Area Under the Curve (AUC) of ASP1955888-00
NCT02223351 (13) [back to overview]Area Under the Curve (AUC) of ASP2151
NCT02223351 (13) [back to overview]Half-life (t1/2) of ASP1955888-00
NCT02223351 (13) [back to overview]Peak Plasma Concentration (Cmax) of ASP1955888-00
NCT02223351 (13) [back to overview]Peak Plasma Concentration (Cmax) of ASP2151
NCT02223351 (13) [back to overview]Time of Peak Concentration (Tmax) of ASP1955888-00
NCT02223351 (13) [back to overview]Time of Peak Concentration (Tmax) of ASP2151
NCT02223351 (13) [back to overview]Number of Participants With Serious and Non-Serious Adverse Events
NCT02223351 (13) [back to overview]Apparent Volume of Distribution (Vd/F) of ASP2151
NCT02265237 (5) [back to overview]Percentage of Participants in Arms A, B and C With Post-treatment Relapse
NCT02265237 (5) [back to overview]Percentage of Participants in Arms A, B and C With On-treatment Virologic Failure
NCT02265237 (5) [back to overview]Percentage of Participants With SVR12 in Participants Receiving 16 Weeks (Arm B) of Treatment Compared to Participants Receiving 24 Weeks of Treatment (Arm C)
NCT02265237 (5) [back to overview]Percentage of Participants With SVR12 in Participants Receiving 12 Weeks (Arm A) of Treatment Compared to Participants Receiving 16 Weeks of Treatment (Arm B)
NCT02265237 (5) [back to overview]Percentage of Participants in Arms A, B and C With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02275780 (16) [back to overview]Change From Baseline in Mean CD4+ T-cell Count at Week 48
NCT02275780 (16) [back to overview]Mean Change From Baseline in Fasting Low Density Lipoprotein Cholesterol (LDL-C) at Week 48
NCT02275780 (16) [back to overview]Mean Change From Baseline in Fasting Triglyceride at Week 48
NCT02275780 (16) [back to overview]Mean Change From Baseline in Fasting Total Cholesterol at Week 48
NCT02275780 (16) [back to overview]Percentage of Participants Achieving Plasma HIV-1 RNA <40 Copies/mL at Week 96
NCT02275780 (16) [back to overview]Percentage of Participants Achieving Plasma HIV-1 RNA <40 Copies/mL at Week 48
NCT02275780 (16) [back to overview]Mean Change From Baseline in Fasting Non-High Density Lipoprotein Cholesterol (Non-HDL-C) at Week 48
NCT02275780 (16) [back to overview]Percentage of Participants Achieving Plasma HIV-1 RNA <50 Copies/mL at Week 48
NCT02275780 (16) [back to overview]Change From Baseline in Mean CD4+ T-cell Count at Week 96
NCT02275780 (16) [back to overview]Mean Change From Baseline in Fasting High Density Lipoprotein Cholesterol (HDL-C) at Week 48
NCT02275780 (16) [back to overview]Percentage of Participants With Any Serious Adverse Event
NCT02275780 (16) [back to overview]Percentage of Participants With Any Drug-related Serious Adverse Event
NCT02275780 (16) [back to overview]Percentage of Participants With Any Drug-related Adverse Event
NCT02275780 (16) [back to overview]Percentage of Participants With Any Adverse Event
NCT02275780 (16) [back to overview]Percentage of Participants Who Discontinued Study Treatment Due to an Adverse Event
NCT02275780 (16) [back to overview]Percentage of Participants Achieving Plasma HIV-1 RNA <50 Copies/mL at Week 96
NCT02292719 (3) [back to overview]Percentage of Participants With Post-treatment Relapse
NCT02292719 (3) [back to overview]Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02292719 (3) [back to overview]Percentage of Participants With On-treatment Virologic Failure
NCT02356562 (4) [back to overview]Percentage of Participants With Post-Treatment Relapse
NCT02356562 (4) [back to overview]Percentage of Participants With On-treatment Virologic Failure
NCT02356562 (4) [back to overview]Percentage of Part 2 Participants With Sustained Virologic Response 12 (SVR12) Weeks Post-treatment
NCT02356562 (4) [back to overview]Percentage of Part 1 Participants With Sustained Virologic Response 12 (SVR12) Weeks Posttreatment
NCT02386098 (8) [back to overview]Percentage of Participants With Plasma HIV-1 RNA <40 c/mL at Weeks 48 and 96-Stage 1
NCT02386098 (8) [back to overview]Percentage of Participants With HIV-1 RNA <200 c/mL at Weeks 24, 48 and 96-Stage 1
NCT02386098 (8) [back to overview]Change From Baseline in Percentage of CD4+ Cells Over Time-Stage 1
NCT02386098 (8) [back to overview]Change From Baseline in Logarithm to the Base 10 (log10) HIV-1 RNA Over Time-Stage 1
NCT02386098 (8) [back to overview]Change From Baseline in Cluster of Differentiation 4+ (CD4+) Cell Count Over Time-Stage 1
NCT02386098 (8) [back to overview]Number of Participants With Serious Adverse Events (SAEs) and Adverse Events (AEs) Leading to Discontinuation (AELD)-Stage 1
NCT02386098 (8) [back to overview]Number of Participants With Occurrence of New Acquired Immunodeficiency Syndrome (AIDS) Defining Events-Stage 1
NCT02386098 (8) [back to overview]Percentage of Participants With Plasma Human Immunodeficiency Virus-1 (HIV-1) Ribonucleic Acid (RNA) <40 Copies Per Milliliter (c/mL) at Week 24-Stage 1
NCT02397096 (12) [back to overview]Mean Change From Baseline in Cluster of Differentiation (CD4) Cell Counts
NCT02397096 (12) [back to overview]Mean Change From Baseline in Fasting Low-density Lipoprotein Cholesterol (LDL-C)
NCT02397096 (12) [back to overview]Mean Change From Baseline in Fasting Non-high-density Lipoprotein Cholesterol (Non-HDL-C)
NCT02397096 (12) [back to overview]Mean Change From Baseline in Cluster of Differentiation (CD4) Cell Counts
NCT02397096 (12) [back to overview]Percentage of Participants Discontinuing From Study Medication Due to an AE(s)
NCT02397096 (12) [back to overview]Percentage of Participants Experiencing ≥1 Adverse Event (AE)
NCT02397096 (12) [back to overview]Percentage of Participants Experiencing ≥1 Serious Adverse Event (SAE)
NCT02397096 (12) [back to overview]Percentage of Participants Maintaining HIV-1 RNA <40 Copies/mL
NCT02397096 (12) [back to overview]Percentage of Participants Maintaining HIV-1 RNA <40 Copies/mL
NCT02397096 (12) [back to overview]Percentage of Participants Maintaining HIV-1 RNA <50 Copies/mL
NCT02397096 (12) [back to overview]Percentage of Participants Maintaining Human Immunodeficiency Virus-1 Ribonucleic Acid (HIV-1 RNA) <50 Copies/mL
NCT02397096 (12) [back to overview]Percentage of Participants With HIV-1 RNA >=50 Copies/mL
NCT02399345 (3) [back to overview]Percentage of Participants With Sustained Virologic Response 12 Weeks (SVR12) Post-treatment
NCT02399345 (3) [back to overview]Percentage of Subjects With On-treatment Virologic Failure
NCT02399345 (3) [back to overview]Percentage of Subjects With Post-treatment Relapse
NCT02430181 (1) [back to overview]Improvement in Quantitative Serum HDV RNA Levels After 4-12 Weeks of Lonafarnib-based Therapy
NCT02430194 (4) [back to overview]ALT Normalization at End of Treatment
NCT02430194 (4) [back to overview]Mean HDV RNA Decline
NCT02430194 (4) [back to overview]≥2 log10 Decline of HDV RNA From Baseline at End of Treatment (EOT)
NCT02430194 (4) [back to overview]< LLOQ in HDV RNA at End of Treatment (EOT)
NCT02442271 (7) [back to overview]Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02442271 (7) [back to overview]Hepatitis C Virus Patient-Reported Outcomes Instrument (HCV-PRO) Total Score: Change From Baseline to 12 Weeks After the Last Dose of Study Drug
NCT02442271 (7) [back to overview]Percentage of Participants With SVR12 by Fibrosis Stage
NCT02442271 (7) [back to overview]Percentage of Participants With SVR12 by Participant Eligibility for Treatment With Interferon (IFN) at Screening
NCT02442271 (7) [back to overview]Percentage of Participants With SVR12 by Participant Prior HCV Treatment Experience
NCT02442271 (7) [back to overview](SF-36v2) Mental Component Summary (MCS) Scores: Change From Baseline to 12 Weeks After the Last Dose of Study Drug
NCT02442271 (7) [back to overview]Short-Form 36 Version 2 Health Survey (SF-36v2) Physical Component Summary (PCS) Scores: Change From Baseline to 12 Weeks After the Last Dose of Study Drug
NCT02442284 (4) [back to overview]Percentage of Participants With Post-treatment Relapse
NCT02442284 (4) [back to overview]Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02442284 (4) [back to overview]Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12) Among Participants With Ongoing Psychiatric Disorders
NCT02442284 (4) [back to overview]Percentage of Participants With Virologic Failure During Treatment
NCT02461745 (2) [back to overview]Sustained Virological Response (SVR) at Week 12
NCT02461745 (2) [back to overview]Sustained Virological Response (SVR) at Week 4
NCT02476617 (1) [back to overview]Change in Interferon (IFN)-Stimulated Genes (ISG) Expression in Peripheral Blood Mononucleated Cells (PBMCs) for Participants Achieving SVR12
NCT02486406 (16) [back to overview]Part 1: Maximum Plasma Concentration (Cmax) of Ombitasvir (OBV)
NCT02486406 (16) [back to overview]Parts 1 and 2: Percentage of Participants With Alanine Aminotransferase (ALT) Normalization During Treatment by Formulation, Age and Weight Group, Across All Subjects, and Across All Subjects on the Adult Formulations
NCT02486406 (16) [back to overview]Parts 1 & 2: Percentage of Participants With Sustained Virologic Response 24 Weeks After the Last Actual Dose of Study Drug (SVR24), Summarized by Formulation, Age and Weight Group, Across All Subjects, and Across All Subjects on the Adult Formulations
NCT02486406 (16) [back to overview]Part 1: Concentration of Drug in Blood Plasma Against Time [Area Under the Curve (AUC)] of Dasabuvir (DSV)
NCT02486406 (16) [back to overview]Part 1: Maximum Plasma Concentration (Cmax) of Ritonavir (RTV)
NCT02486406 (16) [back to overview]Part 1: Concentration of Drug in Blood Plasma Over Time [Area Under the Curve (AUC)] of Ritonavir (RTV)
NCT02486406 (16) [back to overview]Parts 1 and 2: Percentage of Participants With Sustained Virologic Response 12 Weeks After the Last Actual Dose of Study Drug (SVR12)
NCT02486406 (16) [back to overview]Part 1: Lowest Plasma Concentration (Ctrough) of Dasabuvir (DSV)
NCT02486406 (16) [back to overview]Part 1: Lowest Plasma Concentration (Ctrough) of Ombitasvir (OBV)
NCT02486406 (16) [back to overview]Part 1: Lowest Plasma Concentration (Ctrough) of Paritaprevir (PTV)
NCT02486406 (16) [back to overview]Part 1: Lowest Plasma Concentration (Ctrough) of Ritonavir (RTV)
NCT02486406 (16) [back to overview]Part 1: Maximum Plasma Concentration (Cmax) of Paritaprevir (PTV)
NCT02486406 (16) [back to overview]Parts 1 and 2: Percentage of Participants With Sustained Virologic Response 12 Weeks After the Last Actual Dose of Study Drug (SVR12) Summarized by Formulation, Age and Weight Group, and Across All Subjects on the Adult Formulations
NCT02486406 (16) [back to overview]Part 1: Maximum Plasma Concentration (Cmax) of Dasabuvir (DSV)
NCT02486406 (16) [back to overview]Part 1: Concentration of Drug in Blood Plasma Over Time [Area Under the Curve (AUC)] of Paritaprevir (PTV)
NCT02486406 (16) [back to overview]Part 1: Concentration of Drug in Blood Plasma Over Time [Area Under the Curve (AUC)] of Ombitasvir (OBV)
NCT02487199 (4) [back to overview]Percentage of Participants With On-treatment Virologic Failure
NCT02487199 (4) [back to overview]Percentage of Participants With Post-treatment Relapse
NCT02487199 (4) [back to overview]Number of Participants With Adverse Events
NCT02487199 (4) [back to overview]Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02493855 (1) [back to overview]Slope of the Second Phase Decline in Plasma HCV Ribonucleic Acid (RNA) Levels During Treatment
NCT02511431 (2) [back to overview]Number of Participants With Decline of HDV RNA Quantitative Measurement of > 2 Logs From Baseline at 24 Weeks of Treatment
NCT02511431 (2) [back to overview]Number of Participants With Decline of Hepatitis Delta Virus (HDV) RNA Quantitative Measurements of >2 Logs From Baseline at 12 Weeks of Treatment
NCT02517515 (5) [back to overview]Percentage of Participants With Post-treatment Relapse by Post-treatment Week 12
NCT02517515 (5) [back to overview]Percentage of Participants With Post-treatment Relapse by Post-treatment Week 24
NCT02517515 (5) [back to overview]Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02517515 (5) [back to overview]Percentage of Participants With Sustained Virologic Response 24 Weeks Post-treatment (SVR24)
NCT02517515 (5) [back to overview]Percentage of Participants With On-treatment Virologic Failure
NCT02517528 (5) [back to overview]Percentage of Participants Achieving Sustained Virologic Response 24 Weeks Post-Treatment (SVR24)
NCT02517528 (5) [back to overview]Percentage of Participants With On Treatment Virologic Failure
NCT02517528 (5) [back to overview]Percentage of Participants With Virologic Relapse
NCT02517528 (5) [back to overview]Percentage of Participants Achieving Sustained Virologic Response 12 Weeks Post-Treatment (SVR12)
NCT02517528 (5) [back to overview]Percentage of Participants With Virologic Relapse by Post-Treatment Week 24
NCT02527707 (2) [back to overview]Number of Patients With 1 Log Reduction From Baseline by Timepoint
NCT02527707 (2) [back to overview]Change From Baseline to Week 24 in Mean Hepatitis D Virus (HDV) Ribonucleic Acid (RNA) Titer
NCT02581163 (8) [back to overview]Percentage of Participants With Relapse
NCT02581163 (8) [back to overview]Percentage of Participants Achieving Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02581163 (8) [back to overview]Percentage of Participants With Virologic Response at End of Treatment (EoT)
NCT02581163 (8) [back to overview]Percentage of Participants With On-treatment Virologic Failure
NCT02581163 (8) [back to overview]Percentage of Participants With Viral Breakthrough
NCT02581163 (8) [back to overview]Percentage of Participants With Missing Sustained Virologic Response 12 Weeks Post-Treatment (SVR12) Data and/or Nonresponders Who Did Not Meet Specific SVR12 Nonresponder Criteria
NCT02581163 (8) [back to overview]Percentage of Participants Meeting Relapse Criteria
NCT02581163 (8) [back to overview]Percentage of Participants Meeting Premature Study Drug Discontinuation Criteria
NCT02581189 (8) [back to overview]Percentage of Participants Meeting Premature Study Drug Discontinuation Criteria
NCT02581189 (8) [back to overview]Percentage of Participants With On-treatment Virologic Failure
NCT02581189 (8) [back to overview]Percentage of Participants With Relapse
NCT02581189 (8) [back to overview]Percentage of Participants With Viral Breakthrough
NCT02581189 (8) [back to overview]Percentage of Participants With Virologic Response at End of Treatment (EoT)
NCT02581189 (8) [back to overview]Percentage of Participants Achieving Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02581189 (8) [back to overview]Percentage of Participants Meeting Relapse Criteria
NCT02581189 (8) [back to overview]Percentage of Participants With Missing Sustained Virologic Response 12 Weeks Post-Treatment (SVR12) Data and/or Nonresponders Who Did Not Meet Specific SVR12 Nonresponder Criteria
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: Glucose
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: HDL
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: Insulin
NCT02581202 (36) [back to overview]Number of Participants With Adverse Events
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: Serum Creatinine
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: Total Cholesterol
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: Triglycerides
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: LDL
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: Glucose
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: HDL
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: Insulin
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: LDL
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: Serum Creatinine
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: Total Cholesterol
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: Triglycerides
NCT02581202 (36) [back to overview]Number of Participants Who Developed Resistance to Nucleoside Reverse Transcriptase Inhibitors (NRTIs), Non-nucleoside Reverse Transcriptase Inhibitors (NNRTIs), and Protease Inhibitors (PIs)
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in CD4+ T-cell Counts (Cells/mm^3) at Week 24
NCT02581202 (36) [back to overview]Percentage of Participants on Dual Therapy (LPV/r + 3TC) With Undetectable HIV-1 RNA Level at Week 24
NCT02581202 (36) [back to overview]Percentage of Participants on Dual Therapy (LPV/r + 3TC) With Undetectable HIV-1 RNA Level at Week 48
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline (BL) in HIV-1- RNA Viral Load at Week 24 (Base-10 Logarithm Transformed Data)
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline (BL) in HIV-1- RNA Viral Load at Week 24 (Untransformed Data)
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Anthropometric Measurements At Week 24
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Anthropometric Measurements At Week 48
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in CD4+ T-cell Counts (%) at Week 24
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in CD4+ T-cell Counts (%) at Week 48
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: Alanine Aminotransferase (µmol/L)
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in CD4+ T-cell Counts (Cells/mm^3) at Week 48
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in HIV-1- RNA Viral Load at Week 48 (Base-10 Logarithm Transformed Data)
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in HIV-1- RNA Viral Load at Week 48 (Untransformed Data)
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: Alanine Aminotransferase (U/L)
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: Alanine Aminotransferase (U/L)
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: Alanine Aminotransferase (µmol/L)
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: Aspartate Aminotransferase (U/L)
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: Aspartate Aminotransferase (µmol/L)
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: Aspartate Aminotransferase (U/L)
NCT02581202 (36) [back to overview]Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: Aspartate Aminotransferase (µmol/L)
NCT02582632 (10) [back to overview]Percentage of Female Participants Responding With SVR12
NCT02582632 (10) [back to overview]Percentage of Female Participants Responding With SVR12: mITT-GT Population
NCT02582632 (10) [back to overview]Percentage of Participants Who Achieve Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02582632 (10) [back to overview]Percentage of Participants With Post-Treatment Relapse12: mITT-GT Population
NCT02582632 (10) [back to overview]Percentage of Participants With Post-Treatment Relapse12
NCT02582632 (10) [back to overview]Percentage of Participants With On-Treatment Virologic Failure During Treatment Period: mITT-GT Population
NCT02582632 (10) [back to overview]Percentage of Participants With On-Treatment Virologic Failure During Treatment Period
NCT02582632 (10) [back to overview]Percentage of Participants With Baseline HCV RNA < 6,000,000 IU/mL Responding With SVR12: mITT-GT Population
NCT02582632 (10) [back to overview]Percentage of Participants With Baseline HCV RNA < 6,000,000 IU/mL Responding With SVR12
NCT02582632 (10) [back to overview]Percentage of Participants Who Achieve SVR12: mITT-GT Population
NCT02582671 (8) [back to overview]Percentage of Participants Achieving Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02582671 (8) [back to overview]Percentage of Participants With Virologic Response at End of Treatment (EOT)
NCT02582671 (8) [back to overview]Percentage of Participants With Relapse
NCT02582671 (8) [back to overview]Percentage of Participants With On-treatment Virologic Failure
NCT02582671 (8) [back to overview]Percentage of Participants With Missing Sustained Virologic Response 12 Weeks Post-Treatment (SVR12) Data and/or Nonresponders Who Did Not Meet Specific SVR12 Nonresponder Criteria
NCT02582671 (8) [back to overview]Percentage of Participants Meeting Relapse Criteria
NCT02582671 (8) [back to overview]Percentage of Participants Meeting Premature Study Drug Discontinuation Criteria
NCT02582671 (8) [back to overview]Percentage of Participants With Viral Breakthrough
NCT02609659 (5) [back to overview]Percentage of Participants With Hemoglobin < 10 g/dL During Treatment
NCT02609659 (5) [back to overview]Percentage of Participants With On-treatment Virologic Failure
NCT02609659 (5) [back to overview]Percentage of Participants With Post-treatment Relapse
NCT02609659 (5) [back to overview]Mean Change in Hemoglobin Values From Baseline to End of Treatment
NCT02609659 (5) [back to overview]Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02707952 (6) [back to overview]Percentage of Participants in Arm A With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02707952 (6) [back to overview]Percentage of Participants in Arms A and B With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02707952 (6) [back to overview]Percentage of Participants With On-treatment Virologic Failure
NCT02707952 (6) [back to overview]Percentage of Participants With Post-Treatment Relapse
NCT02707952 (6) [back to overview]Percentage of Participants for Each Sub-Population in Arms C and D With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02707952 (6) [back to overview]Percentage of Participants for Each Sub-Population in Arms C and D With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02725866 (8) [back to overview]Percentage of Participants With On-treatment Virologic Failure
NCT02725866 (8) [back to overview]Percentage of Participants With Viral Breakthrough
NCT02725866 (8) [back to overview]Percentage of Participants With Missing Sustained Virologic Response 12 Weeks Post-Treatment (SVR12) Data and/or Nonresponders Who Did Not Meet Specific SVR12 Nonresponder Criteria
NCT02725866 (8) [back to overview]Percentage of Participants Achieving Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02725866 (8) [back to overview]Percentage of Participants With Relapse
NCT02725866 (8) [back to overview]Percentage of Participants Meeting Relapse Criteria
NCT02725866 (8) [back to overview]Percentage of Participants Meeting Premature Study Drug Discontinuation Criteria
NCT02725866 (8) [back to overview]Percentage of Participants With Virologic Response at End of Treatment (EoT)
NCT02786537 (28) [back to overview]Mean Change in Fatigue PRO Score -Phase 1
NCT02786537 (28) [back to overview]Mean Change in HCV- PRO- Phase 1
NCT02786537 (28) [back to overview]Mean Change in HCV-PRO (Functional Well-being) EBR/GZR vs. SOF/LDV Score-Phase 2
NCT02786537 (28) [back to overview]Mean Change in Headache-EBR/GZR and SOF/LDV
NCT02786537 (28) [back to overview]Mean Change in Headache-PRO Scores -Phase 1
NCT02786537 (28) [back to overview]Mean Change in Nausea/Vomiting PRO Score -Phase 1
NCT02786537 (28) [back to overview]Mean Change in Nausea/Vomiting PROMIS Score -EBR/GZR vs. LDV/SOF
NCT02786537 (28) [back to overview]Median Change in Fatigue -Phase 1
NCT02786537 (28) [back to overview]Median Change in Fatigue-Phase 2
NCT02786537 (28) [back to overview]Median Change in HCV-PRO (Overall Well Being) -Phase 1
NCT02786537 (28) [back to overview]Median Change in Headache -Phase 1
NCT02786537 (28) [back to overview]Median Change in Headache-Phase 2
NCT02786537 (28) [back to overview]Median Change in Nausea PRO Score -Phase 1
NCT02786537 (28) [back to overview]Median Change in Nausea PROMIS Score-EBR/GZR SOF/LDV
NCT02786537 (28) [back to overview]Number of Participants With Adverse Events That Caused Treatment Discontinuation-EBR/GZR vs. LDV/SOF
NCT02786537 (28) [back to overview]Phase 1 Number of Participants With Sustained Virologic Response (SVR12-mITT Without Imputation)
NCT02786537 (28) [back to overview]Phase 1-Sustained Virologic Response (SVR12) mITT With Imputation
NCT02786537 (28) [back to overview]Phase 1/2 Number of Participants With Sustained Virologic Response (SVR12-mITT Without Imputation)
NCT02786537 (28) [back to overview]Post-treatment Progression/Regression of Liver Disease-Fib-4
NCT02786537 (28) [back to overview]Sustained Virologic Response (SVR12) mITT With Imputation-Phase 1 and 2 EBR/GZR, SOF/LDV
NCT02786537 (28) [back to overview]Treatment Non-Adherence Probability Estimates
NCT02786537 (28) [back to overview]Change in Functional Status (HCV-PRO) Within Treatment
NCT02786537 (28) [back to overview]Change in HCV-associated Symptoms (PROMIS Measures) After HCV Treatment Initiation
NCT02786537 (28) [back to overview]Impact of Baseline NS5A RASs on Treatment Outcomes-Phase 2
NCT02786537 (28) [back to overview]16 Wk EBR/GZR With RBV Efficacy on Patients With HCV RASs
NCT02786537 (28) [back to overview]HCV SVR Durability -No Cirrhosis
NCT02786537 (28) [back to overview]HCV SVR Durability-Patients With Cirrhosis
NCT02786537 (28) [back to overview]Mean Change in Fatigue PRO -EBR/GZR vs SOF/LDV
NCT02803138 (9) [back to overview]Percentage of Participants With Viral Breakthrough
NCT02803138 (9) [back to overview]Percentage of Participants With Relapse
NCT02803138 (9) [back to overview]Percentage of Participants With Sufficient Follow-up Who Achieved Sustained Virological Response 12 Weeks Posttreatment
NCT02803138 (9) [back to overview]Percentage of Participants With Virologic Response at End of Treatment (EoT)
NCT02803138 (9) [back to overview]Percentage of Participants Achieving Sustained Virologic Response 12 Weeks Post-treatment (SVR12)
NCT02803138 (9) [back to overview]Percentage of Participants Meeting Premature Study Drug Discontinuation Criteria
NCT02803138 (9) [back to overview]Percentage of Participants Meeting Relapse Criteria
NCT02803138 (9) [back to overview]Percentage of Participants With Missing Sustained Virologic Response 12 Weeks Post-Treatment (SVR12) Data and/or Nonresponders Who Did Not Meet Specific SVR12 Nonresponder Criteria
NCT02803138 (9) [back to overview]Percentage of Participants With On-treatment Virologic Failure
NCT03020004 (1) [back to overview]Percentage of Subjects With Sustained Virologic Response (SVR12) 12 Weeks Post-treatment
NCT03020082 (1) [back to overview]Proportion of Participants With Sustained Virologic Response 12 Weeks After Discontinuation of Therapy (SVR12)
NCT03020095 (1) [back to overview]Percentage of Subjects With Sustained Virologic Response (SVR12) 12 Weeks Post-treatment
NCT03070470 (3) [back to overview]"Change From Baseline J-Tpeakc With Balanced Ion Channel Drugs (Ranolazine, Verapamil, Lopinavir / Ritonavir)"
NCT03070470 (3) [back to overview]"Change From Baseline QTc With Predominant hERG Blocking Drug (Chloroquine)"
NCT03070470 (3) [back to overview]QTc Shortening From Calcium Block (Diltiazem) in the Presence of hERG Block (Dofetilide)
NCT03234036 (45) [back to overview]Part 1: AUC (0-infinity) Following Administration of GSK2838232 Tablet in Fasted and Fed State
NCT03234036 (45) [back to overview]Part 1: Area Under the Concentration-time Curve From Time Zero (Pre-dose) Extrapolated to Infinite Time (AUC [0-infinity]) Following Administration of GSK2838232 Tablet and Capsule Formulation in Fed State
NCT03234036 (45) [back to overview]Part 1: Change From Baseline in Blood Pressure
NCT03234036 (45) [back to overview]Part 1: Blood Pressure at Indicated Time Points
NCT03234036 (45) [back to overview]Part 2: Observed Concentration at the End of the Dosing Interval (Ctau) Following Administration of Non-boosted Once-daily Doses of GSK2838232 Tablet in Fed State
NCT03234036 (45) [back to overview]Part 2: Time of Last Quantifiable Concentration (Tlast) Following Administration of Non-boosted Once-daily Doses of GSK2838232 Tablet in Fed State on Day 11
NCT03234036 (45) [back to overview]Part 2: Lag-time (Tlag) Following Administration of Non-boosted Once-daily Doses of GSK2838232 Tablet in Fed State on Day 1
NCT03234036 (45) [back to overview]Part 2: Pulse Rate at Indicated Time Points
NCT03234036 (45) [back to overview]Part 2: AUC(0-infinity) Following Administration of Non-boosted Once-daily Doses of GSK2838232 Tablet in Fed State on Day 11
NCT03234036 (45) [back to overview]Part 1: Plasma Drug Concentration at 24 Hours (C24) Following Administration of GSK2838232 Tablet and Capsule Formulation in Fed State and Tablet Formulation in Fasted State
NCT03234036 (45) [back to overview]Part 2: Accumulation Ratio Calculated From Ctau Following Administration of GSK2838232 as Non-boosted Once-daily Doses of a Tablet Formulation
NCT03234036 (45) [back to overview]Part 2: Accumulation Ratio Calculated From Cmax Following Administration of GSK2838232 as Non-boosted Once-daily Doses of a Tablet Formulation
NCT03234036 (45) [back to overview]Part 2: Accumulation Ratio Calculated From AUC(0-tau) Following Administration of GSK2838232 as Non-boosted Once-daily Doses of a Tablet Formulation
NCT03234036 (45) [back to overview]Part 2: Slope of Pre-dose Concentration of GSK2838232 Administered as Non-boosted Once-daily Doses of a Tablet Formulation to Assess Achievement of Steady State
NCT03234036 (45) [back to overview]Part 2: Tmax Following Administration of Non-boosted Once-daily Doses of GSK2838232 Tablet in Fed State
NCT03234036 (45) [back to overview]Part 1: Maximum Observed Concentration (Cmax) Following Administration of GSK2838232 Tablet and Capsule Formulation in Fed State
NCT03234036 (45) [back to overview]Part 2: Number of Participants With Worst Case Urinalysis Results Relative to Normal Range Criteria
NCT03234036 (45) [back to overview]Part 2: Apparent Terminal Phase Half-life (T1/2) Following Administration of Non-boosted Once-daily Doses of GSK2838232 Tablet in Fed State on Day 11
NCT03234036 (45) [back to overview]Part 1: Cmax Following Administration of GSK2838232 Tablet in Fasted and Fed State
NCT03234036 (45) [back to overview]Part 1: AUC(0-t) Following Administration of GSK2838232 Tablet and Capsule Formulation in Fed State and Tablet Formulation in Fasted State
NCT03234036 (45) [back to overview]Part 2: Number of Participants With Abnormal Electrocardiogram (ECG) Findings
NCT03234036 (45) [back to overview]Part 1: T1/2 Following Administration of GSK2838232 Tablet and Capsule Formulation in Fed State and Tablet Formulation in Fasted State
NCT03234036 (45) [back to overview]Part 1: Time of Occurrence of Cmax (Tmax) Following Administration of GSK2838232 Tablet in Fasted and Fed State
NCT03234036 (45) [back to overview]Part 1: Tlag Following Administration of GSK2838232 Tablet and Capsule Formulation in Fed State and Tablet Formulation in Fasted State
NCT03234036 (45) [back to overview]Part 1: Tlast Following Administration of GSK2838232 Tablet and Capsule Formulation in Fed State and Tablet Formulation in Fasted State
NCT03234036 (45) [back to overview]Part 1: Tmax Following Administration of GSK2838232 Tablet and Capsule Formulation in Fed State and Tablet Formulation in Fasted State
NCT03234036 (45) [back to overview]Part 1: Change From Baseline in Mean Heart Rate Values as ECG Parameter
NCT03234036 (45) [back to overview]Part 1: Change From Baseline in PR Interval, QRS, QT Interval, and QTcF Interval as ECG Parameters
NCT03234036 (45) [back to overview]Part 1: Change From Baseline in Pulse Rate
NCT03234036 (45) [back to overview]Part 1: Number of Participants With Abnormal ECG Findings
NCT03234036 (45) [back to overview]Part 1: Number of Participants With SAEs and Non-SAEs
NCT03234036 (45) [back to overview]Part 1: Number of Participants With Worst Case Clinical Chemistry Results to PCI Criteria
NCT03234036 (45) [back to overview]Part 1: Number of Participants With Worst Case Hematology Results to PCI Criteria
NCT03234036 (45) [back to overview]Part 1: Number of Participants With Worst Case Urinalysis Results Relative to Normal Range Criteria
NCT03234036 (45) [back to overview]Part 1: Pulse Rate at Indicated Time Points
NCT03234036 (45) [back to overview]Part 2: Area Under the Plasma Drug Concentration Time Curve From Pre-dose to the End of the Dosing Interval at Steady State (AUC[0-tau]) Following Administration of Non-boosted Once-daily Doses of GSK2838232 Tablet in Fed State
NCT03234036 (45) [back to overview]Part 2: Blood Pressure at Indicated Time Points
NCT03234036 (45) [back to overview]Part 2: Change From Baseline in Blood Pressure
NCT03234036 (45) [back to overview]Part 2: Change From Baseline in Mean Heart Rate Values as ECG Parameter
NCT03234036 (45) [back to overview]Part 2: Change From Baseline in PR Interval, QRS, QT Interval, and QTcF Interval as ECG Parameters
NCT03234036 (45) [back to overview]Part 2: Change From Baseline in Pulse Rate
NCT03234036 (45) [back to overview]Part 2: Cmax Following Administration of Non-boosted Once-daily Doses of GSK2838232 Tablet in Fed State
NCT03234036 (45) [back to overview]Part 2: Number of Participants With Serious Adverse Events (SAEs) and Non-SAEs
NCT03234036 (45) [back to overview]Part 2: Number of Participants With Worst Case Clinical Chemistry Results to PCI Criteria
NCT03234036 (45) [back to overview]Part 2: Number of Participants With Worst Case Hematology Results to Potential Clinical Importance (PCI) Criteria
NCT03383692 (11) [back to overview]Best Objective Response as Confirmed By The Investigator's Assessment in Participants With HER2-Expressing Advanced Solid Malignant Tumors
NCT03383692 (11) [back to overview]Pharmacokinetic Parameter of Area Under the Serum Concentration-time Curve Following Treatment With DS-8201a and Itraconazole - Cohort 2
NCT03383692 (11) [back to overview]Pharmacokinetic Parameter of Area Under the Serum Concentration-time Curve Following Treatment With DS-8201a and Ritonavir - Cohort 1
NCT03383692 (11) [back to overview]Pharmacokinetic Parameter of Area Under the Serum Concentration-time Curve of MAAA-1181a Following Treatment With DS-8201a and Itraconazole - Cohort 2
NCT03383692 (11) [back to overview]Pharmacokinetic Parameter of Maximum (Peak) Observed Serum Concentration (Cmax) Following Treatment With DS-8201a and Itraconazole - Cohort 2
NCT03383692 (11) [back to overview]Pharmacokinetic Parameter of Maximum (Peak) Observed Serum Concentration (Cmax) Following Treatment With DS-8201a and Ritonavir - Cohort 1
NCT03383692 (11) [back to overview]Pharmacokinetic Parameter of Maximum (Peak) Observed Serum Concentration (Cmax) For MAAA-1181a Following Treatment With DS-8201a and Ritonavir - Cohort 1
NCT03383692 (11) [back to overview]Pharmacokinetic Parameter of Maximum (Peak) Observed Serum Concentration (Cmax) of MAAA-1181a Following Treatment With DS-8201a and Itraconazole - Cohort 2
NCT03383692 (11) [back to overview]Pharmacokinetic Parameter of Area Under the Serum Concentration-time Curve of MAAA-1181a Following Treatment With DS-8201a and Ritonavir - Cohort 1
NCT03383692 (11) [back to overview]Objective Response Rate as Confirmed By The Investigator's Assessment in Participants With HER2-Expressing Advanced Solid Malignant Tumors
NCT03383692 (11) [back to overview]Objective Response Ratio (ORR) as Confirmed By The Investigator's Assessment in Participants With HER2-Expressing Advanced Solid Malignant Tumors
NCT03537404 (10) [back to overview]AUCtau of Raltegravir
NCT03537404 (10) [back to overview]Number of Patients With Abnormal ECG Changes
NCT03537404 (10) [back to overview]Number of Patients With Abnormal Laboratory Values
NCT03537404 (10) [back to overview]AUCtau of Tenofovir
NCT03537404 (10) [back to overview]Cmax of Narlaprevir
NCT03537404 (10) [back to overview]Cmax of Tenofovir
NCT03537404 (10) [back to overview]Number of Patients With Adverse Events
NCT03537404 (10) [back to overview]Cmax of Raltegravir
NCT03537404 (10) [back to overview]AUCtau of Narlaprevir
NCT03537404 (10) [back to overview]Number of Patients With Changes in Vital Signs
NCT03600714 (12) [back to overview]Number of Participants With Reduction of Hepatic Venous Pressure Gradient (HVPG)
NCT03600714 (12) [back to overview]Number of Participants With Sustained Virologic Response
NCT03600714 (12) [back to overview]Number of Participants With Loss of HBsAg at Week 24
NCT03600714 (12) [back to overview]Number of Participants With Loss of HBsAg at Week 12 Weeks After Completing Therapy
NCT03600714 (12) [back to overview]Number of Participants With Loss of HBsAg at 24 Weeks After Completing Therapy
NCT03600714 (12) [back to overview]Number of Participants With Decline of Hepatitis D Virus (HDV) RNA Viral Titer of >2 Logs
NCT03600714 (12) [back to overview]Number of Participants Who Discontinue Medication
NCT03600714 (12) [back to overview]Change in Quantitative Log HBsAg Levels From Baseline to Week 24
NCT03600714 (12) [back to overview]Change in Quantitative Log HBsAg Levels From Baseline 24 Weeks After Completing Therapy
NCT03600714 (12) [back to overview]Number of Participants With Normalization of Serum ALT
NCT03600714 (12) [back to overview]Number of Participants With Reduction in Fibroscan Transient Elastography Values
NCT03600714 (12) [back to overview]Number of Participants With Reduction in Histologic Inflammatory Scores (Modified HAI)
NCT04354428 (6) [back to overview]Number of Participants With Hospitalization or Mortality
NCT04354428 (6) [back to overview]Number of Participants With Serious Adverse Events and Adverse Events Resulting in Treatment Discontinuation
NCT04354428 (6) [back to overview]Number of Persons With Lower Respiratory Tract Infection (LRTI), Defined as Resting Blood Oxygen Saturation (SpO2<93%) Level Sustained for 2 Readings 2 Hours Apart and Presence of Subjective Dyspnea or Cough
NCT04354428 (6) [back to overview]Time to Clearance of Nasal SARS-CoV-2
NCT04354428 (6) [back to overview]Time to Resolution of COVID-19 Symptom Resolution in Days
NCT04354428 (6) [back to overview]COVID-19-related Hospitalization Days
NCT04372628 (8) [back to overview]Hospital-free Days: Day 1 to Day 29 (Group 2 and Placebo Control Group)
NCT04372628 (8) [back to overview]ICU-free Days: Day 1 to Day 29 (Group 2 and Placebo Control Group)
NCT04372628 (8) [back to overview]Oxygen-free Days: Day 1 to Day 29 (Group 2 and Placebo Control Group)
NCT04372628 (8) [back to overview]Time to Hospitalization Day 1 to Day 29 (Group 2 and Placebo Control Group)
NCT04372628 (8) [back to overview]Time to Symptom Resolution: Day 1 to Day 29 (Group 2 and Placebo Control Group)
NCT04372628 (8) [back to overview]Vasopressor-free Days Through Study Day 29 (Group 2 and Placebo Control Group)
NCT04372628 (8) [back to overview]Fever-free Days: Day 1 to Day 29 (Group 2 and Placebo Control Group)
NCT04372628 (8) [back to overview]Ventilator-free Days: Day 1 to Day 29 (Group 2 and Placebo Control Group)
NCT04630002 (54) [back to overview]Cohort 1: AUC(0-tau) of DRV
NCT04630002 (54) [back to overview]Cohort 1: AUC(0-tau) of RTV
NCT04630002 (54) [back to overview]Cohort 1: Cmax of DRV
NCT04630002 (54) [back to overview]Cohort 1: Cmax of RTV
NCT04630002 (54) [back to overview]Cohort 1: Ctau of GSK3640254
NCT04630002 (54) [back to overview]Cohort 1: Ctau of RTV
NCT04630002 (54) [back to overview]Cohort 1: Maximum Observed Concentration (Cmax) of GSK3640254
NCT04630002 (54) [back to overview]Cohort 1: Plasma Concentration at the End of the Dosing Interval (Ctau) of DRV
NCT04630002 (54) [back to overview]Cohort 1: Time of Maximum Observed Concentration (Tmax) of DRV
NCT04630002 (54) [back to overview]Cohort 1: Tmax of RTV
NCT04630002 (54) [back to overview]Cohort 2: AUC(0-tau) of ETR
NCT04630002 (54) [back to overview]Cohort 2: AUC(0-tau) of GSK3640254
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Urinalysis Parameters
NCT04630002 (54) [back to overview]Cohort 1: Area Under the Plasma Concentration-time Curve From Time Zero to the End of the Dosing Interval at Steady State (AUC[0-tau]) of GSK3640254
NCT04630002 (54) [back to overview]Cohort 3: AUC(0-tau) of GSK3640254
NCT04630002 (54) [back to overview]Cohort 2: Tmax of GSK3640254
NCT04630002 (54) [back to overview]Cohort 2: Tmax of ETR
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Clinically Significant Abnormal ECG Findings
NCT04630002 (54) [back to overview]Cohort 2: Ctau of GSK3640254
NCT04630002 (54) [back to overview]Cohort 2: Ctau of ETR
NCT04630002 (54) [back to overview]Cohort 2: Cmax of GSK3640254
NCT04630002 (54) [back to overview]Cohort 2: Cmax of ETR
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Vital Sign Values of Potential Clinical Importance (PCI) Criteria
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With AEs Leading to Discontinuations and Deaths
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Clinically Significant Abnormal Electrocardiogram (ECG) Findings
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Clinically Significant Abnormal ECG Findings
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Clinically Significant Abnormal Electrocardiogram (ECG) Findings
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With AEs Leading to Discontinuations and Deaths
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Calcium, Creatine Kinase, Creatinine, Phosphate, Potassium and Sodium
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Alanine Aminotransferase, Albumin, Alkaline Phosphatase, Amylase, Aspartate Aminotransferase, Bilirubin and Direct Bilirubin
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Clinically Significant Abnormal ECG Findings
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Glucose, Triglycerides, Lipase, Urate and Cholesterol
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Hematology Parameters
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Urinalysis Parameters
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With SAEs and Non-SAEs
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Vital Sign Values of PCI Criteria
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With AEs Leading to Discontinuations and Deaths
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With Clinically Significant Abnormal ECG Findings
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Alanine Aminotransferase, Albumin, Alkaline Phosphatase, Amylase, Aspartate Aminotransferase, Bilirubin and Direct Bilirubin
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Calcium, Creatine Kinase, Creatinine, Phosphate, Potassium and Sodium
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Glucose, Triglycerides, Lipase, Urate and Cholesterol
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Hematology Parameters
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With Clinically Significant Abnormal ECG Findings
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With SAEs and Non-SAEs
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With Vital Sign Values of PCI Criteria
NCT04630002 (54) [back to overview]Cohort 3: Cmax of GSK3640254
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Serious Adverse Events (SAEs) and Non-serious Adverse Events (Non-SAEs)
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Urinalysis Parameters
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Hematology Parameters
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Glucose, Triglycerides, Lipase, Urate and Cholesterol
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Calcium, Creatine Kinase, Creatinine, Phosphate, Potassium and Sodium
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Alanine Aminotransferase, Albumin, Alkaline Phosphatase, Amylase, Aspartate Aminotransferase, Bilirubin and Direct Bilirubin
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Clinically Significant Abnormal Electrocardiogram (ECG) Findings
NCT04630002 (54) [back to overview]Cohort 1: Tmax of GSK3640254
NCT04909853 (16) [back to overview]Number of Participants With Clinical Laboratory Abnormalities
NCT04909853 (16) [back to overview]Number of Participants With Clinically Significant Findings in Physical Examination
NCT04909853 (16) [back to overview]Number of Participants With Clinically Significant Vital Signs Abnormalities
NCT04909853 (16) [back to overview]Plasma Concentration of PF-07321332 at 12 Hours Post Dose (C12)
NCT04909853 (16) [back to overview]Plasma Concentration of PF-07321332 at 24 Hours Post Dose (C24)
NCT04909853 (16) [back to overview]Renal Clearance (CLr) of PF-07321332
NCT04909853 (16) [back to overview]Terminal Elimination Plasma Half-life (t1/2) of PF-07321332
NCT04909853 (16) [back to overview]Time to Reach Maximum Observed Plasma Concentration (Tmax) of PF-07321332
NCT04909853 (16) [back to overview]Number of Participants With Treatment-Emergent Adverse Events (TEAEs), Treatment Emergent Serious Adverse Events (SAEs), Treatment Emergent Treatment Related AEs and Treatment Emergent Treatment Related SAEs
NCT04909853 (16) [back to overview]Number of Participants With Clinically Significant 12-Lead Electrocardiogram (ECG) Abnormalities
NCT04909853 (16) [back to overview]Maximum Observed Plasma Concentration (Cmax) of PF-07321332
NCT04909853 (16) [back to overview]Area Under the Plasma Concentration-time Profile From Time Zero (0) to Extrapolated Infinite Time (AUCinf) of PF-07321332
NCT04909853 (16) [back to overview]Area Under the Plasma Concentration-time Profile From Time Zero (0) to the Time of the Last Quantifiable Concentration (AUClast) of PF-07321332
NCT04909853 (16) [back to overview]Apparent Volume of Distribution (Vz/F) of PF-07321332
NCT04909853 (16) [back to overview]Apparent Clearance (CL/F) of PF-07321332 From Plasma
NCT04909853 (16) [back to overview]Amount of PF-07321332 Excreted Unchanged in Urine Over 48 Hours (Ae48)
NCT04960202 (38) [back to overview]Percentage of Participants With Severe Covid-19 Signs and Symptoms Through Day 28- mITT1 Population
NCT04960202 (38) [back to overview]Percentage of Participants With Severe Covid-19 Signs and Symptoms Through Day 28- mITT Population
NCT04960202 (38) [back to overview]Percentage of Participants With Covid-19 Related Hospitalization or Death From Any Cause Through Day 28- Modified Intent-To-Treat 1 (mITT1) Population
NCT04960202 (38) [back to overview]Percentage of Participants With Covid-19 Related Hospitalization or Death From Any Cause Through Day 28- Modified Intent-To-Treat (mITT) Population
NCT04960202 (38) [back to overview]Percentage of Participants Who Died Through Week 24- mITT2 Population
NCT04960202 (38) [back to overview]Percentage of Participants Who Died Through Week 24- mITT1 Population
NCT04960202 (38) [back to overview]Time to Sustained Alleviation of All Targeted COVID-19 Signs and Symptoms Through Day 28- Modified Intent-to-Treat 2 (mITT2) Population
NCT04960202 (38) [back to overview]Percentage of Participants Who Died Through Week 24- mITT Population
NCT04960202 (38) [back to overview]Number of Participants With Treatment Emergent Adverse Events (TEAEs)
NCT04960202 (38) [back to overview]Number of Participants With Progression to a Worsening Status in 1 or More Self-reported COVID-19 Associated Symptoms Through Day 28-mITT2 Population
NCT04960202 (38) [back to overview]Number of Participants With Progression to a Worsening Status in 1 or More Self-reported COVID-19 Associated Symptoms Through Day 28-mITT1 Population
NCT04960202 (38) [back to overview]Number of Participants With Progression to a Worsening Status in 1 or More Self-reported COVID-19 Associated Symptoms Through Day 28-mITT Population
NCT04960202 (38) [back to overview]Time to Sustained Resolution of Each Targeted COVID-19 Signs and Symptoms- mITT1 Population
NCT04960202 (38) [back to overview]Time to Sustained Resolution of Each Targeted COVID-19 Signs and Symptoms- mITT Population
NCT04960202 (38) [back to overview]Time to Sustained Alleviation of Each Targeted COVID-19 Signs and Symptoms- mITT2 Population
NCT04960202 (38) [back to overview]Time to Sustained Alleviation of Each Targeted COVID-19 Signs and Symptoms- mITT1 Population
NCT04960202 (38) [back to overview]Time to Sustained Alleviation of Each Targeted COVID-19 Signs and Symptoms- mITT Population
NCT04960202 (38) [back to overview]Plasma Concentration Versus Time Summary of PF-07321332
NCT04960202 (38) [back to overview]Percentage of Participants With a Resting Peripheral Oxygen Saturation >=95% at Days 1 and 5- mITT2 Population
NCT04960202 (38) [back to overview]Percentage of Participants With a Resting Peripheral Oxygen Saturation >=95% at Days 1 and 5- mITT1 Population
NCT04960202 (38) [back to overview]Percentage of Participants With a Resting Peripheral Oxygen Saturation >=95% at Days 1 and 5- mITT Population
NCT04960202 (38) [back to overview]Number of Participants With AEs Leading to Discontinuation and Serious Adverse Events (SAEs)
NCT04960202 (38) [back to overview]Number of Days in Hospital and ICU for the Treatment of COVID-19- mITT2 Population
NCT04960202 (38) [back to overview]Number of Days in Hospital and ICU for the Treatment of COVID-19- mITT1 Population
NCT04960202 (38) [back to overview]Number of Days in Hospital and ICU for the Treatment of COVID-19- mITT Population
NCT04960202 (38) [back to overview]Change From Baseline in Logarithm to Base10 (Log10) Transformed Viral Load at Day 3, 5, 10 and 14- mITT Population
NCT04960202 (38) [back to overview]Change From Baseline in Log10 Transformed Viral Load at Day 3, 5, 10 and 14- mITT2 Population
NCT04960202 (38) [back to overview]Change From Baseline in Log10 Transformed Viral Load at Day 3, 5, 10 and 14- mITT1 Population
NCT04960202 (38) [back to overview]Time to Sustained Resolution of All Targeted COVID-19 Signs and Symptoms Through Day 28- mITT2 Population
NCT04960202 (38) [back to overview]Number of COVID-19 Related Medical Visits- mITT1 Population
NCT04960202 (38) [back to overview]Percentage of Participants With Severe Covid-19 Signs and Symptoms Through Day 28- mITT2 Population
NCT04960202 (38) [back to overview]Time to Sustained Resolution of All Targeted COVID-19 Signs and Symptoms Through Day 28- mITT1 Population
NCT04960202 (38) [back to overview]Time to Sustained Resolution of All Targeted COVID-19 Signs and Symptoms Through Day 28- mITT Population
NCT04960202 (38) [back to overview]Number of COVID-19 Related Medical Visits- mITT2 Population
NCT04960202 (38) [back to overview]Time to Sustained Alleviation of All Targeted COVID-19 Signs and Symptoms Through Day 28- mITT1 Population
NCT04960202 (38) [back to overview]Time to Sustained Alleviation of All Targeted COVID-19 Signs and Symptoms Through Day 28- mITT Population
NCT04960202 (38) [back to overview]Number of COVID-19 Related Medical Visits- mITT Population
NCT04960202 (38) [back to overview]Time to Sustained Resolution of Each Targeted COVID-19 Signs and Symptoms- mITT2 Population
NCT04962022 (16) [back to overview]Change From Baseline in Vital Signs Data - Supine Systolic Blood Pressure
NCT04962022 (16) [back to overview]Number of Participants With Clinically Significant 12-lead Electrocardiogram (ECG) Findings
NCT04962022 (16) [back to overview]Number of Participants With Laboratory Abnormalities (Without Regard to Baseline Abnormality)
NCT04962022 (16) [back to overview]Number of Participants With Treatment Emergent Adverse Events (TEAEs)
NCT04962022 (16) [back to overview]Change From Baseline in Vital Signs Data - Supine Systolic Blood Pressure
NCT04962022 (16) [back to overview]Apparent Clearance(CL/F) of PF-07321332
NCT04962022 (16) [back to overview]Apparent Volume of Distribution (Vz/F) of PF-07321332
NCT04962022 (16) [back to overview]Area Under the Plasma Concentration-time Profile From Time Zero to the Time of the Last Quantifiable Concentration(AUClast) of PF-07321332
NCT04962022 (16) [back to overview]Area Under the Plasma Concentration-time Profile From Time Zero to Time Tau (τ), Where Tau=12-hour Dosing Interval(AUCtau) for PF-07321332
NCT04962022 (16) [back to overview]Maximum Observed Concentration (Cmax) of PF-07321332
NCT04962022 (16) [back to overview]Terminal Half-life(t1/2) of PF-07321332
NCT04962022 (16) [back to overview]Time for Cmax (Tmax) for PF-07321332
NCT04962022 (16) [back to overview]Change From Baseline in Vital Signs Data - Supine Diastolic Blood Pressure
NCT04962022 (16) [back to overview]Change From Baseline in Vital Signs Data - Supine Diastolic Blood Pressure
NCT04962022 (16) [back to overview]Change From Baseline in Vital Signs Data - Supine Pulse Rate
NCT04962022 (16) [back to overview]Change From Baseline in Vital Signs Data - Supine Pulse Rate
NCT04962230 (18) [back to overview]Vz/F of Ritonavir
NCT04962230 (18) [back to overview]Number of Participants With Clinically Significant Change From Baseline in Vital Signs
NCT04962230 (18) [back to overview]Tmax of Ritonavir
NCT04962230 (18) [back to overview]Terminal Half-Life ( t1/2) of PF-07321332
NCT04962230 (18) [back to overview]Number of Participants With Treatment-Emergent Adverse Events (TEAEs)
NCT04962230 (18) [back to overview]Number of Participants With Laboratory Abnormalities
NCT04962230 (18) [back to overview]Apparent Oral Clearance (CL/F) of PF-07321332
NCT04962230 (18) [back to overview]Number of Participants With Clinically Significant Change in Electrocardiogram (ECG) Findings
NCT04962230 (18) [back to overview]Apparent Volume of Distribution (Vz/F) of PF-07321332
NCT04962230 (18) [back to overview]Time to Reach Maximum Observed Plasma Concentration (Tmax) of PF-07321332
NCT04962230 (18) [back to overview]Area Under the Curve From Time Zero to Extrapolated Infinite Time (AUCinf) of PF-07321332
NCT04962230 (18) [back to overview]Area Under the Curve From Time Zero to Last Quantifiable Concentration (AUClast) of PF-07321332
NCT04962230 (18) [back to overview]AUCinf of Ritonavir
NCT04962230 (18) [back to overview]AUClast of Ritonavir
NCT04962230 (18) [back to overview]CL/F of Ritonavir
NCT04962230 (18) [back to overview]Cmax of Ritonavir
NCT04962230 (18) [back to overview]Maximum Observed Plasma Concentration (Cmax) of PF-07321332
NCT04962230 (18) [back to overview]t1/2 of Ritonavir
NCT05005312 (7) [back to overview]Number of Participants With Abnormal Laboratory Assessments (Without Regard to Baseline Abnormality)
NCT05005312 (7) [back to overview]Area Under the Plasma Concentration-Time Profile From Time Zero Extrapolated to Infinite Time (AUCinf) of Plasma PF-07321332
NCT05005312 (7) [back to overview]Maximum Observed Plasma Concentration (Cmax) of Plasma PF-07321332
NCT05005312 (7) [back to overview]Number of Participants With Abnormal Electrocardiograms (ECGs)
NCT05005312 (7) [back to overview]Area Under the Plasma Concentration-Time Profile From Time Zero to Time of the Last Quantifiable Concentration (AUClast) of Plasma PF-07321332
NCT05005312 (7) [back to overview]Number of Participants With an Treatment Emergent Adverse Event (TEAE)
NCT05005312 (7) [back to overview]Number of Participants With Abnormal Vital Signs
NCT05011513 (14) [back to overview]Number of Participants With Treatment Emergent Adverse Events (TEAEs), Serious AEs and AEs Leading to Study and Study Drug Discontinuation
NCT05011513 (14) [back to overview]Duration of Hospitalization and Intensive Care Unit (ICU) Stay Through Day 28
NCT05011513 (14) [back to overview]Change From Baseline in Logarithm to Base10 (Log10) Transformed Viral Load at Days 3, 5, 10 and 14
NCT05011513 (14) [back to overview]Time to Sustained Resolution of Overall COVID-19 Signs and Symptoms Through Day 28
NCT05011513 (14) [back to overview]Percentage of Participants With COVID-19 Related Hospitalization or Death From Any Cause Through Day 28
NCT05011513 (14) [back to overview]Percentage of Participants With Resting Peripheral Oxygen Saturation Greater Than or Equal to (>=) 95% at Day 1 and Day 5
NCT05011513 (14) [back to overview]Plasma Concentration Versus Time Summary of PF-07321332
NCT05011513 (14) [back to overview]Time to Sustained Alleviation of Each COVID-19 Signs and Symptoms Through Day 28
NCT05011513 (14) [back to overview]Time to Sustained Resolution of Each COVID-19 Signs and Symptoms Through Day 28
NCT05011513 (14) [back to overview]Number of COVID-19 Related Medical Visits Per Day Through Day 28
NCT05011513 (14) [back to overview]Percentage of Participants With Death Through Week 24
NCT05011513 (14) [back to overview]Percentage of Participants With Progression to Worsening Status of COVID-19 Signs and Symptoms
NCT05011513 (14) [back to overview]Percentage of Participants With Severe Signs and Symptoms of COVID-19 Through Day 28
NCT05011513 (14) [back to overview]Time to Sustained Alleviation of Overall COVID-19 Signs and Symptoms Through Day 28
NCT05032950 (14) [back to overview]Number of Participants With Laboratory Abnormalities
NCT05032950 (14) [back to overview]Apparent Volume of Distribution (Vz/F) of Midazolam When Administered Alone, With PF-07321332/Ritonavir, and With Ritonavir
NCT05032950 (14) [back to overview]Area Under the Plasma Concentration-time Profile From Time 0 to the Time of the Last Quantifiable Concentration (Clast) (AUClast) of Midazolam When Administered Alone and With PF-07321332/Ritonavir
NCT05032950 (14) [back to overview]Apparent Clearance (CL/F) of Midazolam When Administered Alone, With PF-07321332/Ritonavir, and With Ritonavir
NCT05032950 (14) [back to overview]Area Under the Plasma Concentration-time Profile From Time 0 Extrapolated to Infinity Time (AUCinf) of Midazolam When Administered Alone and With PF-07321332/Ritonavir
NCT05032950 (14) [back to overview]AUCinf of Midazolam When Administered Alone and With Ritonavir
NCT05032950 (14) [back to overview]AUClast of Midazolam When Administered Alone and With Ritonavir
NCT05032950 (14) [back to overview]Cmax of Midazolam When Administered Alone and With Ritonavir
NCT05032950 (14) [back to overview]Maximum Plasma Concentration (Cmax) of Midazolam When Administered Alone and With PF-07321332/Ritonavir
NCT05032950 (14) [back to overview]Number of Participants With Vital Signs Abnormalities
NCT05032950 (14) [back to overview]Terminal Half-life (t1/2) of Midazolam When Administered Alone, With PF-07321332/Ritonavir, and With Ritonavir
NCT05032950 (14) [back to overview]Time for Cmax (Tmax) of Midazolam When Administered Alone, With PF-07321332/Ritonavir, and With Ritonavir
NCT05032950 (14) [back to overview]Number of Participants With Electrocardiogram (ECG) Abnormalities
NCT05032950 (14) [back to overview]Number of Participants With Treatment-emergent Adverse Events (TEAEs)
NCT05047601 (15) [back to overview]Plasma Concentration Versus Time Summary of Nirmatrelvir (PF-07321332)
NCT05047601 (15) [back to overview]Number of COVID-19 Related Medical Visits Through Day 28: Among Participants With Negative RT-PCR at Baseline
NCT05047601 (15) [back to overview]Percentage of Participants Who Developed Symptomatic RT-PCR or RAT Confirmed SARS-CoV-2 Infection Through Day 14: Among Participants With Negative RT-PCR at Baseline
NCT05047601 (15) [back to overview]Percentage of Participants Who Developed Symptomatic RT-PCR or RAT Confirmed SARS-CoV-2 Infection Through Day 14: Among Participants With Negative RT-PCR at Baseline With Increased Risk of Severe COVID-19 Illness
NCT05047601 (15) [back to overview]Percentage of Participants With Asymptomatic RT-PCR or RAT Confirmed SARS-CoV-2 Infection Through Day 14: Among Participants With Negative RT-PCR at Baseline
NCT05047601 (15) [back to overview]Percentage of Participants With COVID-19 Related Hospitalization or Death From Any Cause Through Day 28: Among Participants With Negative RT-PCR at Baseline With Increased Risk of Severe COVID-19 Illness
NCT05047601 (15) [back to overview]Percentage of Participants With Death Event Through Day 38: Among Participants With Negative RT-PCR at Baseline
NCT05047601 (15) [back to overview]Percentage of Participants With no, Mild, Moderate, or Severe Signs and Symptoms Attributed to COVID-19 Through Day 28: Among Participants With Negative RT-PCR at Baseline
NCT05047601 (15) [back to overview]Percentage of Participants With Symptomatic RT-PCR or RAT Confirmed SARS-CoV-2 Infection Through Day 14: Among Participants With Negative, Positive or Missing RT-PCR at Baseline
NCT05047601 (15) [back to overview]Percentage of Participants With Symptomatic RT-PCR or RAT Confirmed SARS-CoV-2 Infection Through Day 14: Among Participants With Positive RT-PCR at Baseline
NCT05047601 (15) [back to overview]Viral Load in Nasal Samples Over Time: Among Participants With Negative RT-PCR at Baseline
NCT05047601 (15) [back to overview]Time to RT-PCR or RAT Confirmed SARS-CoV-2 Infection Through Day 14: Among Participants With Negative RT-PCR at Baseline
NCT05047601 (15) [back to overview]Number of Days of Hospitalization and Intensive Care Unit (ICU) Stay: Among Participants With Negative RT-PCR at Baseline
NCT05047601 (15) [back to overview]Number of Participants With Treatment Emergent Adverse Events (TEAEs), Serious AEs and AEs Leading to Study and Study Drug Discontinuation
NCT05047601 (15) [back to overview]Viral Load in Nasal Samples Over Time: Among Participants With Positive RT-PCR at Baseline
NCT05129475 (11) [back to overview]Apparent Volume of Distribution (Vz/F) of PF-07321332
NCT05129475 (11) [back to overview]Area Under the Plasma Concentration-Time Profile From Time Zero to Extrapolated Infinite Time (AUCinf) of PF-07321332
NCT05129475 (11) [back to overview]Area Under the Plasma Concentration-Time Profile From Time Zero to the Time of the Last Quantifiable Concentration (AUClast) of PF-07321332
NCT05129475 (11) [back to overview]Maximum Observed Plasma Concentration (Cmax) of PF-07321332
NCT05129475 (11) [back to overview]Number of Participants Meeting Pre-Specified Criteria for 12-Lead Electrocardiogram (ECG) Values
NCT05129475 (11) [back to overview]Number of Participants Meeting Pre-Specified Criteria for Vital Signs
NCT05129475 (11) [back to overview]Number of Participants With Clinical Laboratory Abnormalities
NCT05129475 (11) [back to overview]Terminal Elimination Half-life (t1/2) of PF-07321332
NCT05129475 (11) [back to overview]Apparent Clearance (CL/F) of PF-07321332 From Plasma
NCT05129475 (11) [back to overview]Time to Reach Maximum Observed Plasma Concentration (Tmax) of PF-07321332
NCT05129475 (11) [back to overview]Number of Participants With Treatment-Emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs)

Pharmacokinetic (PK) Parameter: Maximum Plasma Concentration (Cmax)

Pharmacokinetics were determined by non-compartmental analysis and Maximum concentration (Cmax) was determined visually. (NCT00006604)
Timeframe: Week 1 (Day 7) Intensive PK-24 hr (Pre-dose, 1, 2, 3, 4, 6, 8, and 12 hours post-dose and the following day at 24-hours post-dose)

Interventionng/mL (Median)
Step I: Group 3: ATV Dose 520mg/m^2 Capsule6463
Step I: Group 4: ATV Dose: 620mg/m^2 Capsule7006
Step I: Group 5: ATV Dose: 310mg/m^2 Powder + RTV6501
Step I: Group 5a: ATV Dose: 310mg/m^2 Powder + RTV4952
Step I: Group 6: ATV Dose: 310mg/m^2 Powder + RTV5593
Step I: Group 7: ATV Dose: 205mg/m^2 Capsule + RTV5013
Step I: Group 8: ATV Dose: 205mg/m^2 Capsule + RTV4095

[back to top]

Pharmacokinetic (PK) Parameter: Minimum Plasma Concentration (C24)

Pharmacokinetics were determined by non-compartmental analysis. C24 determined visually, except in the instance when the patient re-dosed the study medication prior to the 24 hour blood draw or the 24 hour level was not obtained, in which case the C24 was calculated from the elimination rate (ke) and the last measured concentration. (NCT00006604)
Timeframe: Week 1 (Day 7) Intensive PK-24hr (Pre-dose, 1, 2, 3, 4, 6, 8, and 12 hours post-dose and the following day at 24-hours post-dose)

Interventionng/mL (Median)
Step I: Group 3: ATV Dose: 520mg/m^2 Capsule173
Step I: Group 4: ATV Dose: 620mg/m^2 Capsule510
Step I: Group 5: ATV Dose: 310mg/m^2 Powder + RTV482
Step I: Group 5a: ATV Dose: 310mg/m^2 Powder + RTV596
Step I: Group 6: ATV Dose: 310mg/m^2 Powder + RTV947
Step I: Group 7: ATV Dose: 205mg/m^2 Capsule + RTV575
Step I: Group 8: ATV Dose: 205mg/m^2 Capsule + RTV885

[back to top]

Number of Participants Who Died

(NCT00006604)
Timeframe: From study entry up to week 96

,,,,,,
Interventionparticipants (Number)
Death not attributed to ATVDeath attributed to ATV
Step I: Group 3: ATV Dose: 520mg/m^2 Capsule00
Step I: Group 4: ATV Dose: 620mg/m^2 Capsule10
Step I: Group 5: ATV Dose: 310mg/m^2 Powder + RTV10
Step I: Group 5a: ATV Dose: 310mg/m^2 Powder + RTV10
Step I: Group 6: ATV Dose: 310mg/m^2 Powder + RTV00
Step I: Group 7: ATV Dose: 205mg/m^2 Capsule + RTV00
Step I: Group 8: ATV Dose: 205mg/m^2 Capsule + RTV00

[back to top]

Change in CD4 Count (Cells/mm^3) From Baseline to Week 48

(NCT00006604)
Timeframe: Baseline, Week 48

InterventionCells/mm^3 (Median)
Step I: Group 3: ATV Dose: 520mg/m^2 Capsule164
Step I: Group 4: ATV Dose: 620mg/m^2 Capsule140
Step I: Group 5: ATV Dose: 310mg/m^2 Powder + RTV74
Step I: Group 5a: ATV Dose: 310mg/m^2 Powder + RTV-232
Step I: Group 6: ATV Dose: 310mg/m^2 Powder + RTV324
Step I: Group 7: ATV Dose: 205mg/m^2 Capsule + RTV157
Step I: Group 8: ATV Dose: 205mg/m^2 Capsule + RTV135

[back to top]

Change in CD4 Count (Cells/mm^3) From Baseline to Week 96

(NCT00006604)
Timeframe: Baseline, Week 96

Interventioncells/mm^3 (Median)
Step I: Group 3: ATV Dose: 520mg/m^2 Capsule366
Step I: Group 4: ATV Dose: 620mg/m^2 Capsule233
Step I: Group 5: ATV Dose: 310mg/m^2 Powder + RTV340
Step I: Group 5a: ATV Dose: 310mg/m^2 Powder + RTV-600
Step I: Group 6: ATV Dose: 310mg/m^2 Powder + RTV317
Step I: Group 7: ATV Dose: 205mg/m^2 Capsule + RTV260
Step I: Group 8: ATV Dose: 205mg/m^2 Capsule + RTV293

[back to top]

Change in CD4 Percent From Baseline to Week 20

(NCT00006604)
Timeframe: Baseline, Week 20

Interventionpercentage of total lymphocytes (Median)
Step I: Group 3: ATV Dose: 520mg/m^2 Capsule8
Step I: Group 4: ATV Dose: 620mg/m^2 Capsule6
Step I: Group 5: ATV Dose: 310mg/m^2 Powder + RTV8
Step I: Group 5a: ATV Dose: 310mg/m^2 Powder + RTV6
Step I: Group 6: ATV Dose: 310mg/m^2 Powder + RTV11
Step I: Group 7: ATV Dose: 205mg/m^2 Capsule + RTV7
Step I: Group 8: ATV Dose: 205mg/m^2 Capsule + RTV3

[back to top]

Change in CD4 Percent From Baseline to Week 96

(NCT00006604)
Timeframe: Baseline, Week 96

Interventionpercentage of total lymphocytes (Median)
Step I: Group 3: ATV Dose: 520mg/m^2 Capsule14
Step I: Group 4: ATV Dose: 620mg/m^2 Capsule9
Step I: Group 5: ATV Dose: 310mg/m^2 Powder + RTV10
Step I: Group 5a: ATV Dose: 310mg/m^2 Powder + RTV-0.5
Step I: Group 6: ATV Dose: 310mg/m^2 Powder + RTV15
Step I: Group 7: ATV Dose: 205mg/m^2 Capsule + RTV10
Step I: Group 8: ATV Dose: 205mg/m^2 Capsule + RTV9

[back to top]

Change in CD4 Percent From Baseline to Week 48

(NCT00006604)
Timeframe: Baseline, Week 48

Interventionpercentage of total lymphocytes (Median)
Step I: Group 3: ATV Dose: 520mg/m^2 Capsule7
Step I: Group 4: ATV Dose: 620mg/m^2 Capsule9
Step I: Group 5: ATV Dose: 310mg/m^2 Powder + RTV10
Step I: Group 5a: ATV Dose: 310mg/m^2 Powder + RTV0
Step I: Group 6: ATV Dose: 310mg/m^2 Powder + RTV12
Step I: Group 7: ATV Dose: 205mg/m^2 Capsule + RTV8
Step I: Group 8: ATV Dose: 205mg/m^2 Capsule + RTV5

[back to top]

Change in CD4 Count (Cells/mm^3) From Baseline to Week 20

(NCT00006604)
Timeframe: Baseline, Week 20

InterventionCells/mm^3 (Median)
Step I: Group 3: ATV Dose: 520mg/m^2 Capsule120
Step I: Group 4: ATV Dose: 620mg/m^2 Capsule184
Step I: Group 5: ATV Dose: 310mg/m^2 Powder + RTV286
Step I: Group 5a: ATV Dose: 310mg/m^2 Powder + RTV-294
Step I: Group 6: ATV Dose: 310mg/m^2 Powder + RTV312
Step I: Group 7: ATV Dose: 205mg/m^2 Capsule + RTV176
Step I: Group 8: ATV Dose: 205mg/m^2 Capsule + RTV139

[back to top]

Pharmacokinetic (PK) Parameter: Clearance (CL/F)

Pharmacokinetics were determined by non-compartmental analysis and Apparent oral clearance (CL/F) was calculated as ATV dose divided by AUC0-24hr. (NCT00006604)
Timeframe: Week 1 (Day 7) Intensive PK-24 hr (Pre-dose, 1, 2, 3, 4, 6, 8, and 12 hours post-dose and the following day at 24-hours post-dose)

InterventionL/hr/m^2 (Median)
Step I: Group 3: ATV Dose: 520mg/m^2 Capsule12.8
Step I: Group 4: ATV Dose: 620mg/m^2 Capsule11.4
Step I: Group 5: ATV Dose: 310mg/m^2 Powder + RTV7.9
Step I: Group 5a: ATV Dose: 310mg/m^2 Powder + RTV8.3
Step I: Group 6: ATV Dose: 310mg/m^2 Powder + RTV5.3
Step I: Group 7: ATV Dose: 205mg/m^2 Capsule + RTV4.7
Step I: Group 8: ATV Dose: 205mg/m^2 Capsule + RTV4.8

[back to top]

Number of Participants Who Experienced a Safety Endpoint of Interest Attributed to ATV

"Total Bilirubin >= 5.1xULN, ECG Events and Other Grade 3+ toxicities attributed to study treatment.~The AEs were graded by the clinicians according to the Division of AIDS (DAIDS) Toxicity Table (see references in the Protocol Section) as follows: Grade 1=Mild, Grade 2=Moderate, Grade 3=Severe, Grade 4=Potentially Life-Threatening, Grade 5=Death. Relationship to study treatment was determined by the study team." (NCT00006604)
Timeframe: From study entry up to week 96

,,,,,,
Interventionparticipants (Number)
Total Bilirubin >= 5.1xULNIncrease in PR Interval-Grade 1Increase in PR Interval-Grade 2Heart Rate-Grade 2Heart Rate-Grade 3Increase in QTc Interval-Grade 3Other Grade 3+ Toxicities
Step I: Group 3: ATV Dose: 520mg/m^2 Capsule11112110
Step I: Group 4: ATV Dose: 620mg/m^2 Capsule4411003
Step I: Group 5: ATV Dose: 310mg/m^2 Powder + RTV0303100
Step I: Group 5a: ATV Dose: 310mg/m^2 Powder + RTV0101012
Step I: Group 6: ATV Dose: 310mg/m^2 Powder + RTV0401010
Step I: Group 7: ATV Dose: 205mg/m^2 Capsule + RTV4504001
Step I: Group 8: ATV Dose: 205mg/m^2 Capsule + RTV4100000

[back to top]

Percentage of Participants With HIV RNA <400 Copies/mL at Week 24

Over the duration of the protocol, the assay used to determine Plasma HIV RNA levels was transitioned from the Amplicor HIV-1 Assay to the Amplicor HIV-1 Monitor 1.5 UltraSensitive Assay (Roche Molecular Systems, Branchburg, NJ) to finally the Abbott Real time HIV-1 RNA assay. The assays were performed according to the manufacturer's instructions in a laboratory accredited by the College of American Pathologists and certified by the NIH Virology Quality Assurance (VQA) in the United States, and VQA certified in South Africa. (NCT00006604)
Timeframe: Week 24

Interventionpercentage of participants (Number)
Step I: Group 3: ATV Dose: 520mg/m^2 Capsule67
Step I: Group 4: ATV Dose: 620mg/m^2 Capsule44
Step I: Group 5: ATV Dose: 310mg/m^2 Powder + RTV57
Step I: Group 5a: ATV Dose: 310mg/m^2 Powder + RTV44
Step I: Group 6 : ATV Dose: 310mg/m^2 Powder + RTV85
Step I: Group 7: ATV Dose: 205mg/m^2 Capsule + RTV67
Step I: Group 8: ATV Dose: 205mg/m^2 Capsule + RTV38

[back to top]

Percentage of Participants With HIV RNA <400 Copies/mL at Week 48

Over the duration of the protocol, the assay used to determine Plasma HIV RNA levels was transitioned from the Amplicor HIV-1 Assay to the Amplicor HIV-1 Monitor 1.5 UltraSensitive Assay (Roche Molecular Systems, Branchburg, NJ) to finally the Abbott Real time HIV-1 RNA assay. The assays were performed according to the manufacturer's instructions in a laboratory accredited by the College of American Pathologists and certified by the NIH Virology Quality Assurance (VQA) in the United States, and VQA certified in South Africa. (NCT00006604)
Timeframe: Week 48

Interventionpercentage of participants (Number)
Step I: Group 3: ATV Dose: 520mg/m^2 Capsule62
Step I: Group 4: ATV Dose: 620mg/m^2 Capsule32
Step I: Group 5: ATV Dose: 310mg/m^2 Powder + RTV71
Step I: Group 5a: ATV Dose: 310mg/m^2 Powder + RTV55
Step I: Group 6: ATV Dose: 310mg/m^2 Powder + RTV85
Step I: Group 7: ATV Dose: 205mg/m^2 Capsule + RTV63
Step I: Group 8: ATV Dose: 205mg/m^2 Capsule + RTV29

[back to top]

Percentage of Participants With HIV RNA <400 Copies/mL at Week 96

Over the duration of the protocol, the assay used to determine Plasma HIV RNA levels was transitioned from the Amplicor HIV-1 Assay to the Amplicor HIV-1 Monitor 1.5 UltraSensitive Assay (Roche Molecular Systems, Branchburg, NJ) to finally the Abbott Real time HIV-1 RNA assay. The assays were performed according to the manufacturer's instructions in a laboratory accredited by the College of American Pathologists and certified by the NIH Virology Quality Assurance (VQA) in the United States, and VQA certified in South Africa. (NCT00006604)
Timeframe: Week 96

Interventionpercentage of participants (Number)
Step I: Group 3: ATV Dose: 520mg/m^2 Capsule48
Step I: Group 4: ATV Dose: 620mg/m^2 Capsule20
Step I: Group 5: ATV Dose: 310mg/m^2 Powder + RTV75
Step I: Group 5a: ATV Dose: 310mg/m^2 Powder + RTV20
Step I: Group 6: ATV Dose: 310mg/m^2 Powder + RTV73
Step I: Group 7: ATV Dose: 205mg/m^2 Capsule + RTV63
Step I: Group 8: ATV Dose: 205mg/m^2 Capsule + RTV14

[back to top]

Pharmacokinetic (PK) Parameter: Area Under the Curve (AUC24h)

Pharmacokinetics were determined by non-compartmental analysis and AUC0-24hr calculated by the linear trapezoidal method. (NCT00006604)
Timeframe: Week 1 (Day 7) Intensive PK-24hr (Pre-Dose, 1, 2, 3, 4, 6, 8, and 12 hours post-dose and the following day at 24-hours post-dose)

Interventionng*hr/mL (Median)
Step I: Group 3: ATV Dose: 520mg/m^2 Capsule40653
Step I: Group 4: ATV Dose: 620mg/m^2 Capsule51781
Step I: Group 5: ATV 310mg/m^2 Powder + RTV44243
Step I: Group 5a: ATV Dose: 310mg/m^2 Powder + RTV38928
Step I: Group 6: ATV Dose: 310mg/m^2 Powder + RTV52199
Step I: Group 7: ATV Dose: 205mg/m^2 Capsule + RTV45680
Step I: Group 8: ATV Dose: 205mg/m^2 Capsule + RTV42835

[back to top]

Participants Achieving Virologic Half Log Suppression (LOQ = 400 c/mL) at Week 48, (Overall and by PI Sensitivity)

Number of participants with a >=0.5 log10 decrease in HIV RNA from baseline or HIV RNA < 400 c/mL at Week 48, by their baseline phenotypic sensitivity to their randomized PI. (NCT00035932)
Timeframe: Baseline, Week 48

,,
Interventionparticipants (Number)
Overall (n=120, 115, 123)PI Sensitive (n=88, 84, 88)PI Resistant (n=32, 30, 33)
ATV 300 mg / RTV776512
ATV 400 mg / SQV60527
LPV / RTV846716

[back to top]

Participants Achieving Treatment Response (LOQ = 50 c/mL) Without Prior Failure at Week 48

Treatment Response = confirmed suppression to LOQ (50 c/mL). The Algorithm for Treatment Response Without Prior Failure (TRPWF) = participants staying in response at the analysis timepoint without having an intervening, confirmed rebound. (NCT00035932)
Timeframe: Week 48

Interventionparticipants (Number)
ATV 300 mg / RTV43
ATV 400 mg / SQV28
LPV / RTV52

[back to top]

Participants Achieving Treatment Response (LOQ = 50 c/mL) Without Prior Failure at Week 96

Treatment Response = confirmed suppression to LOQ (50 c/mL). The Algorithm for Treatment Response Without Prior Failure (TRPWF) = participants staying in response at the analysis timepoint without having an intervening, confirmed rebound. (NCT00035932)
Timeframe: Week 96

Interventionparticipants (Number)
ATV 300 mg / RTV38
LPV / RTV41

[back to top]

Participants Achieving Virologic Half Log Suppression (LOQ = 400 c/mL) at Week 96

Number of participants with a >=0.5 log10 decrease in HIV RNA from baseline or HIV RNA < 400 c/mL at Week 96. (NCT00035932)
Timeframe: Baseline, Week 96

Interventionparticipants (Number)
ATV 300 mg / RTV61
LPV / RTV58

[back to top]

Participants Achieving Virologic Half Log Suppression (LOQ = 50 c/mL) at Week 24

(NCT00035932)
Timeframe: Week 24

Interventionparticipants (Number)
ATV 300 mg / RTV95
ATV 400 mg / SQV74
LPV / RTV93

[back to top]

Participants Achieving Virologic Half Log Suppression (LOQ = 50 c/mL) at Week 48

(NCT00035932)
Timeframe: Week 48

InterventionParticipants (Number)
ATV 300 mg / RTV76
ATV 400 mg / SQV60
LPV / RTV84

[back to top]

Adherence to Regimen Though Week 48 Based on MACS the Multicenter AIDS Cohort Study (MACS) Adherence Questionnaire

The MACS adherence questionnaire asks patients how many medication doses they missed during the previous day, 2 days, 3 days and 4 days. Drug-specific questions included adherence with dose and frequency. Adherence was defined as taking all doses and numbers of pills as prescribed for each medication. This strict adherence cut-off was based on the guidelines stating that anything less than excellent adherence may result in a virus breakthrough and development of resistance. (NCT00035932)
Timeframe: Baseline, Week 24, Week 48

,,
Interventionparticipants (Number)
Adherent at Baseline (n=27, 25, 33)Adherent at Week 24 (n=18, 11, 25)Adherent at Week 48 (n=11, 4, 20)
ATV 300 mg / RTV12108
ATV 400 mg / SQV1052
LPV / RTV182313

[back to top]

Correlation of ATV Minimum Plasma Concentration (Cmin) Inhibitory Quotient (IQ), and Number of PI Mutations at Baseline and CD4 Cell Count Change From Baseline at Week 24

Pearson correlations of the Cmin (trough plasma concentration), IQ (the ratio of Cmin of ATV to HIV IC50), and Number of baseline PI Mutations with CD4 cell count change from baseline at Week 24 were explored. (NCT00035932)
Timeframe: Baseline, Week 24

,
InterventionPearson Correlation Coefficient (Number)
ATV CminIQ (<10; >=10)# of PI Mutations at baseline (<4; >=4)
ATV 300 mg / RTV0.370.376-0.395
ATV 400 mg / SQV-0.210.105-0.227

[back to top]

Correlation of ATV Minimum Plasma Concentration (Cmin), Inhibitory Quotient (IQ), and Number of Protease Inhibitor (PI) Mutations at Baseline With HIV RNA Change From Baseline at Week 24

Pearson correlations of the Cmin (trough plasma concentration), IQ (the ratio of Cmin of ATV to HIV IC50), and Number of baseline PI Mutations with HIV RNA change from baseline at Week 24 were explored. (NCT00035932)
Timeframe: Baseline, Week 24

,
InterventionPearson Correlation Coefficient (Number)
ATV CminIQ (<10; >=10)# of PI Mutations at baseline (<4; >=4)
ATV 300 mg / RTV-0.056-0.3910.306
ATV 400 mg / SQV0.254-0.0810.437

[back to top]

Deaths, Serious Adverse Events (SAEs), and Adverse Events (AEs) Through Week 48

AE=any new untoward medical occurrence/worsening of a pre-existing medical condition regardless of causal relationship. SAE=any untoward medical occurrence at any dose that: results in death; is life-threatening; requires/prolongs inpatient hospitalization; results in persistent/significant disability; is cancer; is congenital anomaly/birth defect; results in drug dependency/abuse; is an important medical event. (NCT00035932)
Timeframe: From Enrollment through Week 48

,,
Interventionparticipants (Number)
Deaths (n = 120, 115, 123)AEs leading to discontinuation (n = 119, 110, 118)SAEs (n = 120, 115, 123)AEs, grades 1-4 (n = 119, 110, 118)AEs, grades 3-4 (n = 119, 110, 118)
ATV 300 mg / RTV06129711
ATV 400 mg / SQV18149318
LPV / RTV151110312

[back to top]

Grade 3/4 Laboratory Abnormalities Through Week 48

Common Terminology Criteria for Adverse Events v3.0 (CTCAE) Grades:1=Mild, 2=Moderate, 3=Severe, 4=Life-threatening/disabling, 5=Death. Abnormal values: absolute neutrophil count: ≥500 to <750/mm3 (grade 3), <500/mm3 (grade 4); platelets: 20,000-49,999/mm3 (grade 3), <20,000/mm3 or diffuse petechiae (grade 4); alanine transaminase (ALT): 5.1-10 x upper limit of normal (ULN; grade 3), >10 x ULN (grade 4); aspartate transaminase (AST): 5.1-10 x ULN (grade 3), >10 x ULN (grade 4); bilirubin: 2.6-5 x ULN (grade 3), >5 x ULN (grade 4). (NCT00035932)
Timeframe: From Enrollment to Week 48

,,
Interventionparticipants (Number)
Neutrophil ReductionPlatelet ReductionALT ElevationAST ElevationTotal Bilirubin Elevation
ATV 300 mg / RTV825458
ATV 400 mg / SQV844222
LPV / RTV103441

[back to top]

Change From Baseline in CD4 Cell Count at Week 48

(NCT00035932)
Timeframe: Baseline, Week 48

Interventioncells/mm3 (Mean)
ATV 300 mg / RTV110
ATV 400 mg / SQV72
LPV / RTV121

[back to top]

HIV RNA Level - Treated Subjects With Evaluable Cmins at Week 24

Week 24 HIV RNA level and change from baseline were summarized for treated subjects with evaluable Cmins. (NCT00035932)
Timeframe: Baseline, Week 24

,
Interventionlog10 c/mL (Mean)
Baseline ValuesWeek 24 ValuesChange from Baseline at Week 24
ATV 300 mg / RTV4.532.62-1.91
ATV 400 mg / SQV4.412.83-1.57

[back to top]

Lipid Mean Percent Change From Baseline at Week 24

Mean percent change in total cholesterol, high density lipoprotein (HDL) cholesterol, fasting low density lipoprotein (LDL) cholesterol, and fasting triglycerides. (NCT00035932)
Timeframe: Baseline, Week 24

,,
Interventionpercent change (Number)
Total CholesterolHigh Density Lipoprotein (HDL) CholesterolFasting Low Density Lipoprotein (LDL) CholesterolFasting Triglycerides
ATV 300 mg / RTV-8-7-10-2
ATV 400 mg / SQV-9-1-11-14
LPV / RTV30-431

[back to top]

Lipid Mean Percent Change From Baseline at Week 48

Mean percent change in total cholesterol, high density lipoprotein (HDL) cholesterol, fasting low density lipoprotein (LDL) cholesterol, and fasting triglycerides. (NCT00035932)
Timeframe: Week 48

,,
Interventionpercent change in lipid values (Number)
Total CholesterolHDL CholesterolFasting LDL CholesterolFasting Triglycerides
ATV 300 mg / RTV-8-7-10-4
ATV 400 mg / SQV-44-3-14
LPV / RTV62130

[back to top]

Lipid Mean Percent Change From Baseline at Week 96, Observed Values

Mean percent change in total cholesterol, high density lipoprotein (HDL) cholesterol, fasting low density lipoprotein (LDL) cholesterol, and fasting triglycerides. (NCT00035932)
Timeframe: Week 96

,,
Interventionpercent change in lipid values (Number)
Total Cholesterol (n=60, 46, 54)HDL Cholesterol (n=60, 46, 54)Fasting LDL Cholesterol (n=52, 39, 43)Fasting Triglycerides (n=52, 40, 43)
ATV 300 mg / RTV-7-5-11-2
ATV 400 mg / SQV-13-74
LPV / RTV97130

[back to top]

Mean ATV, RTV and SQV Minimum Concentration (Cmin) Values

"The minimum or trough concentration (Cmin) of a drug observed after its administration and just prior to the administration of a subsequent dose." (NCT00035932)
Timeframe: collected at the pre-dose time point after receiving atazanavir for at least four weeks

,
Interventionng/mL (Mean)
ATV (n=40,23)RTV (n=40,0)SQV (n=0,19)
ATV 300 mg / RTV719.53154.83NA
ATV 400 mg / SQV312.01NA52.15

[back to top]

Mean Score of European Quality of Life-5 Dimensions (EQ-5D) Health Index Score at Baseline, Mid-Study (Week 24), and Final (Week 48)

The EQ-5D is a 5-item questionnaire to assess health-related quality of life in 5 health dimensions (mobility, self-care, usual activity, pain/discomfort, anxiety/depression) are scored on a 3-level scale: no problems (1), some problems (2), extreme problems (3). Using a standard algorithm, responses are summarized into a single score, the EQ-5D Health Index Score (HIS), which ranges between 1 (representing perfect health) and 0 (representing the worst imaginable health state or death). The smallest coefficient of change is 0.03. (NCT00035932)
Timeframe: Baseline, Week 24, Week 48

,,
Interventionunits on a scale (Mean)
Baseline (n=99, 86, 100)Mid-Study (n=103, 83, 95)Final (n=93, 84, 96)
ATV 300 mg / RTV0.830.870.84
ATV 400 mg / SQV0.850.860.85
LPV / RTV0.860.890.88

[back to top]

Mean Score of European Quality of Life-5 Dimensions (EQ-5D) Visual Analog Scale (VAS) at Baseline, Mid-Study (Week 24), and Final (Week 48)

The EQ-5D has a Visual Analog Scale (VAS), which is a feeling thermometer-like scale with a range between 0 and 100. Patients are required to draw a line from a box on the VAS scale to an actual mark on the thermometer-like scale that corresponds with a number that reflects their self-assessed health status at the time they are completing the questionnaire. Higher VAS scores indicate better overall health. There is no minimum clinically important difference reported in the literature for VAS. (NCT00035932)
Timeframe: Baseline, Week 24, Week 48

,,
Interventionunits on a scale (Mean)
Baseline (n=98, 85, 101)Mid-Study (n=102, 83, 97)Final (n=95, 81, 96)
ATV 300 mg / RTV81.3384.8982.77
ATV 400 mg / SQV81.7283.3485.80
LPV / RTV81.5285.0986.16

[back to top]

Participants Achieving Virologic Half Log Suppression (Limit of Quantification [LOQ] = 400 c/mL) at Week 24 (Overall and by Protease Inhibitor [PI] Sensitivity)

Number of participants with a >=0.5 log10 decrease in HIV RNA from baseline or HIV RNA < 400 c/mL at Week 24, by their baseline phenotypic sensitivity to their randomized PI. (NCT00035932)
Timeframe: Baseline, Week 24

,,
Interventionparticipants (Number)
Overall (n=120, 115, 123)PI Sensitive (n=88, 83, 88)PI Resistant (n=32, 30, 33)
ATV 300 mg / RTV957916
ATV 400 mg / SQV745815
LPV / RTV937219

[back to top]

Fridericia-corrected QT (QTcF) Interval and Change From Baseline by Analysis Time Point

The QT interval is a measure of the time between the start of the Q wave and the end of the T wave in the heart's electrical cycle. The QT interval was corrected for heart rate using Fridericia's (QTcF) formula. (NCT00035932)
Timeframe: Baseline, Week 4 predose, 2-3 hours postdose, 6-12 hours postdose, Week 12, Week 24, Week 48

,,
Interventionmsec (Mean)
Baseline Mean (n=119, 110, 118)Mean Change at Week 4 predose (n=117, 104, 110)Mean Change Wk 4 2-3 hrs postdose (n=113,102,106)Mean Change Wk 4 6-12 hrs postdose (n=112,101,105)Mean Change at Week 12 (n=110, 97, 107)Mean Change at Week 24 (n=108, 92, 109)Mean Change at Week 48 (n=89, 75, 97)
ATV 300 mg / RTV390-3-2-421-1
ATV 400 mg / SQV3871-3-133-1
LPV / RTV390-2-7-8220

[back to top]

Change From Baseline in CD4 Cell Count at Week 24

(NCT00035932)
Timeframe: Baseline, Week 24

Interventioncells/mm3 (Mean)
ATV 300 mg / RTV83
ATV 400 mg / SQV59
LPV / RTV90

[back to top]

PR Interval and Change From Baseline by Analysis Time Point

The PR interval is measured from the beginning of the P wave to the beginning of the QRS complex, and reflects the time the electrical impulse takes to travel from the sinus node through the atrioventricular (AV) node and entering the ventricles. The PR interval is therefore a good estimate of AV node function. (NCT00035932)
Timeframe: Baseline, Week 4 predose, 2-3 hours postdose, 6-12 hours postdose, Week 12, Week 24, Week 48

,,
Interventionmsec (Mean)
Baseline Mean (n=119, 110, 118)Mean Change at Week 4 predose (n=117, 104, 110)Mean Change Wk 4 2-3 hrs postdose (n=113,102,106)Mean Change Wk 4 6-12 hrs postdose (n=112,101,105)Mean Change at Week 12 (n=110, 97, 107)Mean Change at Week 24 (n=108, 92, 109)Mean Change at Week 48 (n=89, 75, 97)
ATV 300 mg / RTV153412520
ATV 400 mg / SQV155966772
LPV / RTV154312854

[back to top]

Change From Baseline in CD4 Cell Count at Week 96

(NCT00035932)
Timeframe: Baseline, Week 96

Interventioncells/mm3 (Mean)
ATV 300 mg / RTV122
LPV / RTV154

[back to top]

Fasting Glucose Mean Change From Baseline at Week 24

(NCT00035932)
Timeframe: Baseline, Week 24

Interventionmg/dL (Mean)
ATV 300 mg / RTV0
ATV 400 mg / SQV-3
LPV / RTV0

[back to top]

Most Common AEs and AEs of Interest Through Week 48

Prespecified AEs of interest included jaundice, ocular icterus, and hyperbilirubinemia. (NCT00035932)
Timeframe: From Enrollment to Week 48

,,
Interventionparticipants (Number)
Diarrhea (Most Common)Headache (Most Common)Nausea (Most Common)Jaundice (AE of Interest)Ocular Icterus (AE of Interest)Hyperbilirubinemia (AE of Interest)
ATV 300 mg / RTV252119191324
ATV 400 mg / SQV292424638
LPV / RTV541815001

[back to top]

Mean Change From Baseline in HIV RNA at Week 48

(NCT00035932)
Timeframe: Baseline, Week 48

Interventionlog10 c/mL (Mean)
ATV 300 mg / RTV-1.93
ATV 400 mg / SQV-1.55
LPV / RTV-1.87

[back to top]

Fasting Glucose Mean Change From Baseline at Week 48

(NCT00035932)
Timeframe: Week 48

Interventionmg/dL (Mean)
ATV 300 mg / RTV4
ATV 400 mg / SQV-1
LPV / RTV1

[back to top]

HIV IC50 at Week 24

IC50: inhibitory concentration of drug required to reduce viral replication by 50%. (NCT00035932)
Timeframe: Week 24

Interventionng/mL (Mean)
ATV 300 mg / RTV17.83
ATV 400 mg / SQV22.84

[back to top]

Inhibitory Quotient at Week 24

Inhibitory quotient is a measure of drug exposure and susceptibility in an individual. The IQ is typically calculated as the ratio of Cmin to HIV IC50. (NCT00035932)
Timeframe: Baseline, Week 24

Interventionratio (Mean)
ATV 300 mg / RTV136.94
ATV 400 mg / SQV25.04

[back to top]

Mean Change From Baseline in HIV Ribonucleic Acid (RNA) at Week 24

(NCT00035932)
Timeframe: Baseline, Week 24

Interventionlog10 c/mL (Mean)
ATV 300 mg / RTV-1.86
ATV 400 mg / SQV-1.52
LPV / RTV-1.89

[back to top]

Mean Change From Baseline in HIV RNA at Week 2

(NCT00035932)
Timeframe: Baseline, Week 2

Interventionlog10 c/mL (Mean)
ATV 300 mg / RTV-1.18
ATV 400 mg / SQV-1.14
LPV / RTV-1.30

[back to top]

Participants Achieving Virologic Half Log Suppression (LOQ = 50 c/mL) at Week 96

(NCT00035932)
Timeframe: Week 96

InterventionParticipants (Number)
ATV 300 mg / RTV61
LPV / RTV57

[back to top]

Mean Change From Baseline in HIV RNA at Week 96

(NCT00035932)
Timeframe: Baseline, Week 96

Interventionlog10 c/mL (Mean)
ATV 300 mg / RTV-2.29
LPV / RTV-2.08

[back to top]

Participants Achieving Treatment Response (LOQ = 400 c/mL) Without Prior Failure at Week 24

Treatment Response = confirmed suppression to LOQ (400 c/mL). The Algorithm for Treatment Response Without Prior Failure (TRPWF) = participants staying in response at the analysis timepoint without having an intervening, confirmed rebound. (NCT00035932)
Timeframe: Week 24

Interventionparticipants (Number)
ATV 300 mg / RTV76
ATV 400 mg / SQV50
LPV / RTV74

[back to top]

Participants Achieving Treatment Response (LOQ = 400 c/mL) Without Prior Failure at Week 48

Treatment Response = confirmed suppression to LOQ (400 c/mL). The Algorithm for Treatment Response Without Prior Failure (TRPWF) = participants staying in response at the analysis timepoint without having an intervening, confirmed rebound. (NCT00035932)
Timeframe: Week 48

Interventionparticipants (Number)
ATV 300 mg / RTV64
ATV 400 mg / SQV42
LPV / RTV67

[back to top]

Participants Achieving Treatment Response (LOQ = 400 c/mL) Without Prior Failure at Week 96

Treatment Response = confirmed suppression to LOQ (400 c/mL). The Algorithm for Treatment Response Without Prior Failure (TRPWF) = participants staying in response at the analysis timepoint without having an intervening, confirmed rebound. (NCT00035932)
Timeframe: Week 96

Interventionparticipants (Number)
ATV 300 mg / RTV52
LPV / RTV53

[back to top]

Participants Achieving Treatment Response (LOQ = 50 c/mL) Without Prior Failure at Week 24

Treatment Response = confirmed suppression to LOQ (50 c/mL). The Algorithm for Treatment Response Without Prior Failure (TRPWF) = participants staying in response at the analysis timepoint without having an intervening, confirmed rebound. (NCT00035932)
Timeframe: Week 24

Interventionparticipants (Number)
ATV 300 mg / RTV46
ATV 400 mg / SQV25
LPV / RTV50

[back to top]

Geometric Mean of Steady State Plasma Amprenavir (APV) Parameter: AUC(0-tau)

"Geometric mean is a type of average that indicates the central tendency of a set of values and is calculated by multiplying all the numbers in a set, and then taking the nth root of the resulting product. AUC(0-tau)=area under the concentration curve from time 0 to tau." (NCT00040664)
Timeframe: 0, 1, 2, 4, 8, 12, and 24 hours post dosing at Week 4

Interventionhours*micrograms/milliliter (Geometric Mean)
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 2-5 Years47.3
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 6-11 Years47.6
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 12-18 Years75.5
FPV/RTV 1400/200 mg Once Daily (Tablet), 12-18 Years71.8

[back to top]

Least Squares Mean of Plasma APV Parameter: Ctau

A blood sample was drawn on Week 4 over 24 hours (at 0, 1, 2, 4, 8, 12, and 24 hours post dosing). Ratio of geometric least squares mean (90% CI) are presented. Ctau=trough concentration. PK Parameters for QD and BID are compared with Historical adult data. Least squares mean (LSM) are the group means after having controlled for a covariate (i.e., holding it constant at some typical value of the covariate, such as its mean value). LSM is calculated by taking the average of the means within a treatment. (NCT00040664)
Timeframe: 0, 1, 2, 4, 8, 12, and 24 hours post dosing at Week 4.

Interventionmicrogram per milliliter (Least Squares Mean)
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 2-5 Years0.716
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 6-11 Years0.837
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 12-18 Years0.963
FPV/RTV 1400/200 mg Once Daily (Tablet), 12-18 Years0.706

[back to top]

Percentage of Participants With HIV-1 RNA <400 Copies Per mL at Weeks 12, 48, 96, and 168 (Time to Loss of Virologic Response [TLOVR] Analysis)

A blood sample was drawn to determine the amount of HIV-1 RNA virus in copies per milliliter (mL) at Weeks 12, 48, 96, and 196. The percentage of participants with HIV-1 RNA <400 copies/mL at Weeks 12, 48, 96, 168 was determined by the TLOVR algorithm with stratification by the six randomization strata. TLOVR analysis categorizes participants by treatment response. Responders were participants with confirmed viral load <400copies/mL on two consecutive visits. (NCT00040664)
Timeframe: Weeks 12, 48, 96, and 168

,
Interventionpercentage of participants (Number)
Week 12Week 48Week 96Week 168
PI-Experienced2016117
PI-Naive22161311

[back to top] [back to top]

Number of Participants Who Discontinued Treatment Due to Adverse Events

The number of participants who prematurely discontinued study drug due to adverse events was tabulated. Data are summarized by individual adverse event. (NCT00040664)
Timeframe: Baseline through end of study (at least Week 168)

InterventionParticipants (Number)
Any eventNauseaVomitingStomach discomfortHyperglycaemiaHypertriglyceridaemiaBlood alkaline phosphatase increasedHodgkin's diseaseHaemoptysis
FPV/RTV1233111111

[back to top]

Median Change From Baseline HIV-1 RNA Values at Weeks 12, 48, 96, and 168 Visits

A blood sample was drawn to determine the amount of HIV-1 RNA virus in copies/mL at Weeks 12, 24, 48, 96, and 168. Change from Baseline was defined as the HIV-1 RNA level at Weeks 12, 24, 48, 96, and 168 minus the HIV-1 RNA level at Baseline. (NCT00040664)
Timeframe: Baseline and Weeks 12, 48, 96, and 168

,
Interventionlog10 copies/mL (Median)
Week 12Week 48Week 96Week 168
PI-Experienced-2.03-1.65-1.76-2.39
PI-Naive-2.85-2.65-2.52-2.88

[back to top]

Number of Participants With APV Resistance Associated HIV-1 RNA Genotypic Mutations and Phenotypic Resistance at Time of Virologic Failure Not Present at Baseline

A blood sample was drawn for participants failing to respond to therapy, and the mutations present in the virus were identified. For each participant, the mutations found at the time of failure were compared with any mutations found in the blood sample at baseline. New mutations that developed at the time of virologic failure were tabulated by drug class. Virologic failure is defined as HIV-1 RNA greater than or equal to 400 copies/mL. (NCT00040664)
Timeframe: Time of virologic failure

InterventionParticipants (Number)
PI-Naive2
PI-Experienced2

[back to top]

Median Steady State Plasma APV Tmax

tmax: time after administration of the drug when maximum concentration is reached (NCT00040664)
Timeframe: 0, 1, 2, 4, 8, 12, and 24 hours post dosing at Week 4

Interventionhours (Median)
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 2-5 Years1.04
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 6-11 Years1.12
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 12-18 Years1.08
FPV/RTV 1400/200 mg Once Daily (Tablet), 12-18 Years3.78

[back to top]

Least Squares Mean of Plasma APV Parameter: Cmax

A blood sample was drawn on Week 4 over 24 hours (at 0, 1, 2, 4, 8, 12, and 24 hours post dosing). Ratio of geometric least squares mean (90% CI) are presented. Ctau=trough concentration. PK Parameters for QD and BID are compared with Historical adult data. Least squares mean (LSM) are the group means after having controlled for a covariate (i.e., holding it constant at some typical value of the covariate, such as its mean value). LSM is calculated by taking the average of the means within a treatment. (NCT00040664)
Timeframe: 0, 1, 2, 4, 8, 12, and 24 hours post dosing at Week 4.

Interventionmicrograms/milliliters (Least Squares Mean)
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 2-5 Years0.663
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 6-11 Years0.676
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 12-18 Years0.917
FPV/RTV 1400/200 mg Once Daily (Tablet), 12-18 Years1.03

[back to top]

Least Squares Mean of Plasma APV Parameter: AUC0-tau

A blood sample was drawn on Week 4 over 24 hours (at 0, 1, 2, 4, 8, 12, and 24 hours post dosing). Ratio of geometric least squares mean (90% CI) are presented. Ctau=trough concentration. PK Parameters for QD and BID are compared with Historical adult data. Least squares mean (LSM) are the group means after having controlled for a covariate (i.e., holding it constant at some typical value of the covariate, such as its mean value). LSM is calculated by taking the average of the means within a treatment. (NCT00040664)
Timeframe: 0, 1, 2, 4, 8, 12, and 24 hours post dosing at Week 4

Interventionhours*micrograms/milliliters (Least Squares Mean)
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 2-5 Years0.695
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 6-11 Years0.699
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 12-18 Years1.11
FPV/RTV 1400/200 mg Once Daily (Tablet), 12-18 Years1.06

[back to top]

Median Change From Baseline in CD4+ Values at Week 12, 48, 96, and 168 Visits

A blood sample was drawn to determine the CD4+ cell count at Weeks 24, 48, 96, and 168. Change from Baseline was defined as the CD4+ cell count at Weeks 24, 48, 96, and 168 minus the CD4+ cell count at Baseline. (NCT00040664)
Timeframe: Baseline and Weeks 12, 48, 96, and 168

,
InterventionCells/mm3 (Median)
Week 12Week 48Week 96Week 168
PI-Experienced40120400
PI-Naive95150160180

[back to top]

Number of Participants With Grade 3 or 4 Treatment-emergent Laboratory Abnormalities

The number of participants with Grade 3 (severe) or Grade 4 (life-threatening) laboratory abnormalities while on study treatment. (NCT00040664)
Timeframe: Baseline through end of study (at least Week 168)

InterventionParticipants (Number)
All parametersAlanine aminotransferaseAspartate aminotransferaseCholesterolHyperglycemiaHypoglycemiaSerum lipaseTriglyceridesLeucopeniaNeutropeniaThrombocytopeniaAnemia
FPV/RTV6230000111221

[back to top]

Geometric Mean of Steady State Plasma APV Parameter: t1/2

"Geometric mean is a type of average that indicates the central tendency of a set of values and is calculated by multiplying all the numbers in a set, and then taking the nth root of the resulting product. t1/2=elimination half-life. t1/2=elimination half-life." (NCT00040664)
Timeframe: 0, 1, 2, 4, 8, 12, and 24 hours post dosing at Week 4

Interventionhours (Geometric Mean)
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 2-5 Years17.5
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 6-11 Years13.6
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 12-18 Years15.0
FPV/RTV 1400/200 mg Once Daily (Tablet), 12-18 Years14.9

[back to top]

Geometric Mean of Steady State Plasma APV Parameter: Cmax

"Geometric mean is a type of average that indicates the central tendency of a set of values and is calculated by multiplying all the numbers in a set, and then taking the nth root of the resulting product. Cmax= concentration maximum." (NCT00040664)
Timeframe: 0, 1, 2, 4, 8, 12, and 24 hours post dosing at Week 4

Interventionmicrograms/milliliter (Geometric Mean)
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 2-5 Years4.97
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 6-11 Years5.07
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 12-18 Years6.88
FPV/RTV 1400/200 mg Once Daily (Tablet), 12-18 Years7.70

[back to top]

Geometric Mean of Steady State Plasma APV Parameter: CL/F

"Geometric mean is a type of average that indicates the central tendency of a set of values and is calculated by multiplying all the numbers in a set, and then taking the nth root of the resulting product. CL/F=apparent plasma clearance." (NCT00040664)
Timeframe: 0, 1, 2, 4, 8, 12, and 24 hours post dosing at Week 4

Interventionmilliliters/minute/kilogram (Geometric Mean)
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 2-5 Years10.5
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 6-11 Years10.6
FPV/RTV 30/6 mg/kg Once Daily (Suspension), 12-18 Years6.57
FPV/RTV 1400/200 mg Once Daily (Tablet), 12-18 Years4.95

[back to top]

Geometric Mean of Steady State Plasma APV Parameter: CL/F

"Geometric mean is a type of average that indicates the central tendency of a set of values and is calculated by multiplying all the numbers in a set, and then taking the nth root of the resulting product. CL/F=apparent plasma clearance." (NCT00040664)
Timeframe: 0, 1, 2, 4, 8, 12, and 24 hours post dosing at Week 4

Interventionmilliliters/minute (Geometric Mean)
FPV/RTV 1400/200 mg Once Daily (Tablet), 12-18 Years278

[back to top]

PK Parameter: Trough Concentration (C12) With Median (Range) for ARVs and TB Drugs

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. Trough concentration was the measured concentration from the 12h post-dose sample after an observed dose. (NCT00042289)
Timeframe: Measured at 2nd trimester (20-26 wks gestation); 3rd trimester (30-38 gestation); and either 2-3 wks, 2-8 wks, or 6-12 wks postpartum, depending on study arm. Trough concentration was measured 12 hrs after an observed dose.

,,,
Interventionmg/L (Median)
2nd Trimester3rd TrimesterPostpartum
ETR 200mg b.i.d.0.360.480.38
IDV/RTV Arm 2: 400/100mg q.d. (Only THA)0.130.130.28
LPV/RTV Arm 3: 400/100mg b.i.d. Then 600/150mg b.i.d. Then 400/100mg b.i.d.3.75.17.2
RAL 400mg b.i.d.0.06210.0640.0797

[back to top]

PK Parameter: Trough Concentration (C24) With Median (IQR) for ARVs and TB Drugs

"Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. Trough concentration was the measured concentration from the 24h post-dose sample after an observed dose.~For the TAF 25 mg q.d., 10 mg q.d. w/COBI, and 25 mg q.d. w/COBI or RTV boosting arms, samples were all below the limit of quantification and statistical analyses were not conducted." (NCT00042289)
Timeframe: Measured at 2nd trimester (20-26 wks gestation), 3rd trimester (30-38 wks gestation), and either 2-3 wks, 2-8 wks or 6-12 wks postpartum depending on study arm. Trough concentration was measured 24 hrs after an observed dose.

,,,,,,,,,,,,,
Interventionmg/L (Median)
2nd Trimester3rd TrimesterPostpartum
ATV/COBI 300/150 mg q.d.0.210.210.61
ATV/RTV Arm 1: 300/100mg q.d.2.00.71.2
ATV/RTV Arm 2: 300/100mg q.d. Then 400/100mg q.d. Then 300/100mg q.d.0.490.710.90
DRV/COBI 800/150 mg q.d.0.330.271.43
DRV/RTV 800/100mg q.d.0.991.172.78
DTG 50mg q.d.0.730.931.28
EFV 600 mg q.d. (Outside THA)1.491.481.94
EVG/COBI 150/150mg q.d.0.02580.04870.3771
TAF 10mg q.d. w/COBI0.001950.001950.00195
TAF 25mg q.d.0.001950.001950.00195
TAF 25mg q.d. w/COBI or RTV Boosting0.001950.001950.00195
TFV 300mg q.d.0.0390.0540.061
TFV/ATV/RTV Arm 1: 300/300/100mg q.d.0.30.50.8
TFV/ATV/RTV Arm 2: 300/300/100mg q.d. Then 300/400/100mg q.d Then 300/300/100mg q.d.0.440.571.26

[back to top]

PK Parameter: Trough Concentration (C24) With Median (Range) for ARVs and TB Drugs

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. Trough concentration was the measured concentration from the 24h post-dose sample after an observed dose. (NCT00042289)
Timeframe: Measured at 2nd trimester (20-26 wks gestation), 3rd trimester (30-38 wks gestation), and either 2-3 wks, 2-8 wks or 6-12 wks postpartum depending on study arm. Trough concentration was measured 24 hrs after an observed dose.

Interventionmg/L (Median)
3rd TrimesterPostpartum
EFV 600mg q.d.1.602.05

[back to top]

Number of Women Who Met PK Target of Area Under the Curve (AUC) for ARVs

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. AUC (area under the curve) were determined using the linear trapezoidal rule. See PK target in the Protocol Appendix V. (NCT00042289)
Timeframe: Measured at 2nd trimester (20-26 wks gestation), 3rd trimester (30-38 wks gestation), and either 2-3 wks, 2-8 wks or 6-12 wks postpartum depending on study arm. Blood samples were drawn pre-dose and at 1, 2, 4, 6, 8, 12 (and 24) hours post dosing.

,,,,,,,,,,,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
2nd Trimester3rd TrimesterPostpartum
ATV/RTV Arm 1: 300/100mg q.d.11212
DRV/COBI 800/150 mg q.d.3414
DRV/RTV 600 or 800 or 900/100mg b.i.d. Then 800 or 900/100mg b.i.d. Then 600/100mg b.i.d.71622
DRV/RTV 600/100mg b.i.d.71922
DRV/RTV 800/100mg q.d.91922
DTG 50mg q.d.92023
EFV 600 mg q.d. (Outside THA)123334
ATV/RTV Arm 2: 300/100mg q.d. Then 400/100mg q.d. Then 300/100mg q.d.82927
ETR 200mg b.i.d.5137
EVG/COBI 150/150mg q.d.81018
FPV/RTV 700/100mg b.i.d.82622
IDV/RTV Arm 2: 400/100mg q.d. (Only THA)101926
LPV/RTV Arm 3: 400/100mg b.i.d. Then 600/150mg b.i.d. Then 400/100mg b.i.d.93027
ATV/COBI 300/150 mg q.d.125
NFV Arm 2: 1250mg b.i.d. Then 1875mg b.i.d. Then 1250mg b.i.d.NA1514
RAL 400mg b.i.d.113330
RPV 25mg q.d.142625
TAF 10mg q.d. w/COBI152322
TAF 25mg q.d.132324
TAF 25mg q.d. w/COBI or RTV Boosting102418
TFV 300mg q.d.22727
TFV/ATV/RTV Arm 1: 300/300/100mg q.d.11112
TFV/ATV/RTV Arm 2: 300/300/100mg q.d. Then 300/400/100mg q.d Then 300/300/100mg q.d.72332

[back to top]

PK Parameter: Maximum Concentration (Cmax) in ng/mL With Median (95% CI) for ARVs and TB Drugs

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. Cmax was the maximum observed concentration after a dose. (NCT00042289)
Timeframe: Measured at 2nd trimester (20-26 wks gestation), 3rd trimester (30-38 wks gestation), and either 2-3 wks, 2-8 wks or 6-12 wks postpartum depending on study arm; Blood samples were drawn pre-dose and at 1, 2, 4, 6, 8,12 (and 24) hours post dosing.

Interventionng/mL (Median)
3rd TrimesterPostpartum
MVC 150 or 300mg b.i.d.448647

[back to top]

PK Parameter: Cord/Maternal Blood Concentration Ratio With Median (IQR) for ARVs and TB Drugs

Cord blood and maternal plasma concentrations were collected and measured at delivery, and compared as a ratio. (NCT00042289)
Timeframe: Measured at time of delivery with single cord blood and single maternal plasma sample.

Interventionunitless (Median)
DRV/RTV 600 or 800 or 900/100mg b.i.d. Then 800 or 900/100mg b.i.d. Then 600/100mg b.i.d.0.15
DTG 50mg q.d.1.25
EVG/COBI 150/150mg q.d.0.91
DRV/COBI 800/150 mg q.d.0.07
ATV/COBI 300/150 mg q.d.0.07
TFV 300mg q.d.0.88

[back to top]

Number of Women Who Met PK Target of Area Under the Curve (AUC) for ARVs

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. AUC (area under the curve) were determined using the linear trapezoidal rule. See PK target in the Protocol Appendix V. (NCT00042289)
Timeframe: Measured at 2nd trimester (20-26 wks gestation), 3rd trimester (30-38 wks gestation), and either 2-3 wks, 2-8 wks or 6-12 wks postpartum depending on study arm. Blood samples were drawn pre-dose and at 1, 2, 4, 6, 8, 12 (and 24) hours post dosing.

,
InterventionParticipants (Count of Participants)
3rd TrimesterPostpartum
EFV 600mg q.d.2021
MVC 150 or 300mg b.i.d.87

[back to top]

Area Under the Curve From 0 to 24 Hours (AUC24) of ARVs for Contraceptive Arms

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. AUC24h (area-under-the-curve from 0 to 24 hours) were determined using the linear trapezoidal rule. (NCT00042289)
Timeframe: Measured at 2-12 wks postpartum before contraceptive initiation and 6-7 wks after contraceptive initiation. Blood samples were drawn pre-dose and at 0, 1, 2, 6, 8, 12, and 24 hours post dosing.

,
Interventionmcg*hr/mL (Median)
Before contraceptive initiationAfter contraceptive initiation
ATV/RTV/TFV 300/100/300mg q.d. With ENG53.9655.25
EFV 600mg q.d. With ENG53.6456.65

[back to top]

Area Under the Curve From 0 to 12 Hours (AUC12) of ARVs for Contraceptive Arms

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. AUC12h (area-under-the-curve from 0 to 12 hours) were determined using the linear trapezoidal rule. (NCT00042289)
Timeframe: Measured at 2-12 wks postpartum before contraceptive initiation and 6-7 wks after contraceptive initiation. Blood samples were drawn pre-dose and at 0, 1, 2, 6, 8 and 12 hours post dosing.

Interventionmcg*hr/mL (Median)
Before contraceptive initiationAfter contraceptive initiation
LPV/RTV 400/100 b.i.d. With ENG115.97100.20

[back to top]

Plasma Concentration for Contraceptives

Serum concentrations of the contraceptives. Note that no historical controls were provided by team pharmacologists and thus no comparisons were done for contraceptive concentrations in women using hormonal contraceptives and selected ARV drugs as compared to historical controls not using those ARV drugs. (NCT00042289)
Timeframe: Measured at 6-7 weeks after contraceptive initiation postpartum

Interventionpg/mL (Median)
ATV/RTV/TFV 300/100/300mg q.d. With ENG604
LPV/RTV 400/100 b.i.d. With ENG428
EFV 600mg q.d. With ENG125

[back to top]

PK Parameter: Trough Concentration (C24) With Median (Range) for ARVs and TB Drugs

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. Trough concentration was the measured concentration from the 24h post-dose sample after an observed dose. (NCT00042289)
Timeframe: Measured at 2nd trimester (20-26 wks gestation), 3rd trimester (30-38 wks gestation), and either 2-3 wks, 2-8 wks or 6-12 wks postpartum depending on study arm. Trough concentration was measured 24 hrs after an observed dose.

Interventionmg/L (Median)
2nd Trimester3rd TrimesterPostpartum
RPV 25mg q.d.0.0630.0560.081

[back to top]

PK Parameter: Cord/Maternal Blood Concentration Ratio With Median (Range) for ARVs and TB Drugs

Cord blood and maternal plasma concentrations were collected and measured at delivery, and compared as a ratio. For arms with zero overall participants analyzed, samples were below the limit of quantification and ratios could not be calculated. (NCT00042289)
Timeframe: Measured at time of delivery with single cord blood and single maternal plasma sample.

Interventionunitless (Median)
TAF 10mg q.d. w/COBI0.97
EFV 600 mg q.d. (Outside THA)0.67
EFV 600mg q.d.0.49
LPV/RTV Arm 3: 400/100mg b.i.d. Then 600/150mg b.i.d. Then 400/100mg b.i.d.0.2
RAL 400mg b.i.d.1.5
ETR 200mg b.i.d.0.52
MVC 150 or 300mg b.i.d.0.33
ATV/RTV Arm 2: 300/100mg q.d. Then 400/100mg q.d. Then 300/100mg q.d.0.14
TFV/ATV/RTV Arm 2: 300/300/100mg q.d. Then 300/400/100mg q.d Then 300/300/100mg q.d.0.16
NFV Arm 2: 1250mg b.i.d. Then 1875mg b.i.d. Then 1250mg b.i.d.0.19
IDV/RTV Arm 2: 400/100mg q.d. (Only THA)0.12
RPV 25mg q.d.0.55
ATV/RTV 300/100mg q.d. or TFV/ATV/RTV 300/300/100mg q.d.0.18
DRV/RTV 800/100mg q.d. or DRV/RTV 600/100mg b.i.d.0.18

[back to top]

Pharmacokinetic (PK) Parameter: Infant Plasma Washout Half-life (T1/2) of ARVs and TB Drugs

Infant plasma concentrations were collected and measured during the first 9 days of life. Half-life is defined as 0.693/k, where k, the elimination rate constant, is the slope of the decline in concentrations. (NCT00042289)
Timeframe: Infant plasma samples at 2-10, 18-28, 36-72 hours and 5-9 days after birth.

Interventionhour (Median)
DTG 50mg q.d.32.8
EVG/COBI 150/150mg q.d.7.6
DRV/COBI 800/150 mg q.d.NA
EFV 600 mg q.d. (Outside THA)65.6

[back to top]

PK Parameter: Maximum Concentration (Cmax) in mg/L With Median (IQR) for ARVs and TB Drugs

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. Cmax was the maximum observed concentration after a dose. (NCT00042289)
Timeframe: Measured at 2nd trimester (20-26 wks gestation), 3rd trimester (30-38 wks gestation), and either 2-3 wks, 2-8 wks or 6-12 wks postpartum depending on study arm; Blood samples were drawn pre-dose and at 1, 2, 4, 6, 8,12 (and 24) hours post dosing.

Interventionmg/L (Median)
3rd TrimesterPostpartum
EFV 600mg q.d.5.445.10

[back to top]

PK Parameter: Maximum Concentration (Cmax) in mg/L With Median (IQR) for ARVs and TB Drugs

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. Cmax was the maximum observed concentration after a dose. (NCT00042289)
Timeframe: Measured at 2nd trimester (20-26 wks gestation), 3rd trimester (30-38 wks gestation), and either 2-3 wks, 2-8 wks or 6-12 wks postpartum depending on study arm; Blood samples were drawn pre-dose and at 1, 2, 4, 6, 8,12 (and 24) hours post dosing.

,,,,,,,,,,,,,,
Interventionmg/L (Median)
2nd Trimester3rd TrimesterPostpartum
ATV/COBI 300/150 mg q.d.2.822.203.90
ATV/RTV Arm 1: 300/100mg q.d.NA3.64.1
ATV/RTV Arm 2: 300/100mg q.d. Then 400/100mg q.d. Then 300/100mg q.d.3.114.514.52
DRV/COBI 800/150 mg q.d.4.593.677.04
DRV/RTV 600 or 800 or 900/100mg b.i.d. Then 800 or 900/100mg b.i.d. Then 600/100mg b.i.d.6.226.558.96
DRV/RTV 600/100mg b.i.d.5.645.537.78
DRV/RTV 800/100mg q.d.6.775.788.11
DTG 50mg q.d.3.623.544.85
EFV 600 mg q.d. (Outside THA)3.875.134.41
FPV/RTV 700/100mg b.i.d.5.615.126.75
IDV/RTV Arm 2: 400/100mg q.d. (Only THA)3.893.625.37
NFV Arm 2: 1250mg b.i.d. Then 1875mg b.i.d. Then 1250mg b.i.d.NA5.15.0
TFV 300mg q.d.0.2500.2450.298
TFV/ATV/RTV Arm 1: 300/300/100mg q.d.1.22.54.1
TFV/ATV/RTV Arm 2: 300/300/100mg q.d. Then 300/400/100mg q.d Then 300/300/100mg q.d.2.733.565.43

[back to top]

PK Parameter: Maximum Concentration (Cmax) in mg/L With Median (Range) for ARVs and TB Drugs

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. Cmax was the maximum observed concentration after a dose. (NCT00042289)
Timeframe: Measured at 2nd trimester (20-26 wks gestation), 3rd trimester (30-38 wks gestation), and either 2-3 wks, 2-8 wks or 6-12 wks postpartum depending on study arm; Blood samples were drawn pre-dose and at 1, 2, 4, 6, 8,12 (and 24) hours post dosing.

,,,
Interventionmg/L (Median)
2nd Trimester3rd TrimesterPostpartum
ETR 200mg b.i.d.0.701.010.63
LPV/RTV Arm 3: 400/100mg b.i.d. Then 600/150mg b.i.d. Then 400/100mg b.i.d.8.410.714.6
RAL 400mg b.i.d.2.2501.7703.035
RPV 25mg q.d.0.1450.1340.134

[back to top]

PK Parameter: Maximum Concentration (Cmax) in ng/mL With Median (IQR) for ARVs and TB Drugs

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. Cmax was the maximum observed concentration after a dose. (NCT00042289)
Timeframe: Measured at 2nd trimester (20-26 wks gestation), 3rd trimester (30-38 wks gestation), and either 2-3 wks, 2-8 wks or 6-12 wks postpartum depending on study arm; Blood samples were drawn pre-dose and at 1, 2, 4, 6, 8,12 (and 24) hours post dosing.

,,,
Interventionng/mL (Median)
2nd Trimester3rd TrimesterPostpartum
EVG/COBI 150/150mg q.d.1447.11432.81713.1
TAF 10mg q.d. w/COBI80.491.298.2
TAF 25mg q.d.69.796133
TAF 25mg q.d. w/COBI or RTV Boosting87.8107141

[back to top]

PK Parameter: Trough Concentration (C12) With Geometric Mean (95% CI) for ARVs and TB Drugs

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. Trough concentration was the measured concentration from the 12h post-dose sample after an observed dose. (NCT00042289)
Timeframe: Measured at 2nd trimester (20-26 wks gestation), 3rd trimester (30-38 wks gestation), and either 2-3 wks, 2-8 wks or 6-12 wks postpartum depending on study arm. Trough concentration was measured 12 hrs after an observed dose.

Interventionng/mL (Geometric Mean)
3rd TrimesterPostpartum
MVC 150 or 300mg b.i.d.108128

[back to top]

PK Parameter: Trough Concentration (C12) With Median (IQR) for ARVs and TB Drugs

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. Trough concentration was the measured concentration from the 12h post-dose sample after an observed dose. (NCT00042289)
Timeframe: Measured at 2nd trimester (20-26 wks gestation); 3rd trimester (30-38 gestation); and either 2-3 wks, 2-8 wks, or 6-12 wks postpartum, depending on study arm. Trough concentration was measured 12 hrs after an observed dose.

,,,
Interventionmg/L (Median)
2nd Trimester3rd TrimesterPostpartum
DRV/RTV 600 or 800 or 900/100mg b.i.d. Then 800 or 900/100mg b.i.d. Then 600/100mg b.i.d.2.842.524.51
DRV/RTV 600/100mg b.i.d.2.122.222.51
FPV/RTV 700/100mg b.i.d.2.121.642.87
NFV Arm 2: 1250mg b.i.d. Then 1875mg b.i.d. Then 1250mg b.i.d.NA0.470.52

[back to top]

PK Parameter: Area Under the Curve From 0 to 24 Hours (AUC24) With Median (Range) for ARVs and TB Drugs

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. AUC24h (area-under-the-curve from 0 to 24 hours) were determined using the trapezoidal rule. (NCT00042289)
Timeframe: Measured at 2nd trimester (20-26 wks gestation), 3rd trimester (30-38 wks gestation), and either 2-3 wks, 2-8 wks or 6-12 wks postpartum depending on study arm. Blood samples were drawn pre-dose and at 1, 2, 4, 6, 8, 12 and 24 hours post dosing.

Interventionmg*hour/L (Median)
2nd Trimester3rd TrimesterPostpartum
RPV 25mg q.d.1.9691.6692.387

[back to top]

PK Parameter: Area Under the Curve From 0 to 24 Hours (AUC24) With Median (Range) for ARVs and TB Drugs

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. AUC24h (area-under-the-curve from 0 to 24 hours) were determined using the trapezoidal rule. (NCT00042289)
Timeframe: Measured at 2nd trimester (20-26 wks gestation), 3rd trimester (30-38 wks gestation), and either 2-3 wks, 2-8 wks or 6-12 wks postpartum depending on study arm. Blood samples were drawn pre-dose and at 1, 2, 4, 6, 8, 12 and 24 hours post dosing.

Interventionmg*hour/L (Median)
3rd TrimesterPostpartum
EFV 600mg q.d.55.458.3

[back to top]

PK Parameter: Area Under the Curve From 0 to 24 Hours (AUC24) With Median (IQR) for ARVs and TB Drugs

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. AUC24 (area under the curve from 0 to 24 hours) were determined using the linear trapezoidal rule. (NCT00042289)
Timeframe: Measured at 2nd trimester (20-26 wks gestation), 3rd trimester (30-38 wks gestation), and either 2-3 wks, 2-8 wks or 6-12 wks postpartum depending on study arm. Blood samples were drawn pre-dose and at 1, 2, 4, 6, 8, 12 and 24 hours post dosing.

,,,,,,,,,,,,,
Interventionmg*hour/L (Median)
2nd Trimester3rd TrimesterPostpartum
ATV/COBI 300/150 mg q.d.25.3318.8536.20
ATV/RTV Arm 1: 300/100mg q.d.88.241.957.9
ATV/RTV Arm 2: 300/100mg q.d. Then 400/100mg q.d. Then 300/100mg q.d.30.645.748.8
DRV/COBI 800/150 mg q.d.50.0042.0595.55
DRV/RTV 800/100mg q.d.64.663.5103.9
DTG 50mg q.d.47.649.265.0
EFV 600 mg q.d. (Outside THA)47.3060.0262.70
EVG/COBI 150/150mg q.d.15.314.021.0
TAF 10mg q.d. w/COBI0.1970.2060.216
TAF 25mg q.d.0.1710.2120.271
TAF 25mg q.d. w/COBI or RTV Boosting0.1810.2570.283
TFV 300mg q.d.1.92.43.0
TFV/ATV/RTV Arm 1: 300/300/100mg q.d.14.528.839.6
TFV/ATV/RTV Arm 2: 300/300/100mg q.d. Then 300/400/100mg q.d Then 300/300/100mg q.d.26.237.758.7

[back to top]

PK Parameter: Area Under the Curve From 0 to 12 Hours (AUC12) With Median (Range) for ARVs and TB Drugs

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. AUC12 (area under the curve from 0 to 12 hours) were determined using the linear trapezoidal rule. (NCT00042289)
Timeframe: Measured in 2nd trimester (20-26 wks gestation), 3rd trimester (30-38 wks gestation), and either 2-3 wks, 2-8 wks, or 6-12 wks postpartum depending on study arm. Blood samples were drawn pre-dose and at 1, 2, 4, 6, 8, and 12 hrs post dosing.

,,,
Interventionmg*hour/L (Median)
2nd Trimester3rd TrimesterPostpartum
ETR 200mg b.i.d.4.58.35.3
IDV/RTV Arm 2: 400/100mg q.d. (Only THA)14.916.127.1
LPV/RTV Arm 3: 400/100mg b.i.d. Then 600/150mg b.i.d. Then 400/100mg b.i.d.7296133
RAL 400mg b.i.d.6.65.411.6

[back to top]

PK Parameter: Area Under the Curve From 0 to 12 Hours (AUC12) With Median (IQR) for ARVs and TB Drugs

Pharmacokinetic parameters were determined from plasma concentration-time profiles using noncompartmental methods. AUC12 (area under the curve from 0 to 12 hours) were determined using the linear trapezoidal rule. (NCT00042289)
Timeframe: Measured in 2nd trimester (20-26 wks gestation), 3rd trimester (30-38 wks gestation), and either 2-3 wks, 2-8 wks, or 6-12 wks postpartum depending on study arm. Blood samples were drawn pre-dose and at 1, 2, 4, 6, 8, and 12 hrs post dosing.

,,,
Interventionmg*hour/L (Median)
2nd Trimester3rd TrimesterPostpartum
DRV/RTV 600 or 800 or 900/100mg b.i.d. Then 800 or 900/100mg b.i.d. Then 600/100mg b.i.d.55.151.879.6
DRV/RTV 600/100mg b.i.d.45.845.961.7
FPV/RTV 700/100mg b.i.d.43.5032.1551.60
NFV Arm 2: 1250mg b.i.d. Then 1875mg b.i.d. Then 1250mg b.i.d.NA34.233.5

[back to top]

PK Parameter: Area Under the Curve From 0 to 12 Hours (AUC12) With Geometric Mean (95% CI) for ARVs and TB Drugs

Measured in 2nd trimester (20-26 wks gestation), 3rd trimester (30-38 wks gestation), and either 2-3 wks, 2-8 wks, or 6-12 wks postpartum depending on study arm. Blood samples were drawn pre-dose and at 1, 2, 4, 6, 8, and 12 hrs post dosing. (NCT00042289)
Timeframe: Measured in 2nd trimester (20-26 wks gestation), 3rd trimester (30-38 wks gestation), and either 2-3 wks, 2-8 wks, or 6-12 wks postpartum depending on study arm. Blood samples were drawn pre-dose and at 1, 2, 4, 6, 8, and 12 hrs post dosing.

Interventionng*hour/mL (Geometric Mean)
2nd Trimester3rd TrimesterPostpartum
MVC 150 or 300mg b.i.d.NA27173645

[back to top]

Pharmacokinetic (PK) Parameter: Infant Plasma Washout Concentration of ARVs and TB Drugs

Infant plasma concentrations were collected and measured during the first 9 days of life. (NCT00042289)
Timeframe: Blood samples were collected at 2-10, 18-28, 36-72 hours and 5-9 days after birth.

,,,
Interventionmcg/mL (Median)
2-10 hours after birth18-28 hours after birth36-72 hours after birth5-9 days after birth
DRV/COBI 800/150 mg q.d.0.351.431.871.72
DTG 50mg q.d.1.731.531.000.06
EFV 600 mg q.d. (Outside THA)1.11.00.90.4
EVG/COBI 150/150mg q.d.0.1320.0320.0050.005

[back to top]

Mean Change From Baseline to Week 40 in CD4+ Cell Count

(NCT00054717)
Timeframe: Baseline to Week 40

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)50
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)24

[back to top]

Mean Change From Baseline to Week 48 in CD4+ Cell Count

(NCT00054717)
Timeframe: Baseline to Week 48

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)46
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)28

[back to top]

Mean Change From Baseline to Week 56 in CD4+ Cell Count

(NCT00054717)
Timeframe: Baseline to Week 56

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)52
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)26

[back to top]

Mean Change From Baseline to Week 64 in CD4+ Cell Count

(NCT00054717)
Timeframe: Baseline to Week 64

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)50
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)26

[back to top]

Mean Change From Baseline to Week 72 in CD4+ Cell Count

(NCT00054717)
Timeframe: Baseline to Week 72

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)50
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)24

[back to top]

Mean Change From Baseline to Week 8 in CD4+ Cell Count

(NCT00054717)
Timeframe: Baseline to Week 8

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)49
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)25

[back to top]

Mean Change From Baseline to Week 80 in CD4+ Cell Count

(NCT00054717)
Timeframe: Baseline to Week 80

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)55
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)26

[back to top]

Mean Change From Baseline to Week 88 in CD4+ Cell Count

(NCT00054717)
Timeframe: Baseline to Week 88

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)60
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)29

[back to top]

Mean Change From Baseline to Week 96 in CD4+ Cell Count

(NCT00054717)
Timeframe: Baseline to Week 96

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)60
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)28

[back to top]

Median Change From Baseline in Viral Load to Week 2

(NCT00054717)
Timeframe: Baseline to Week 2

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-1.36
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)-0.57

[back to top]

Median Change From Baseline in Viral Load to Week 24

(NCT00054717)
Timeframe: Baseline to Week 24

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.81
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)-0.3

[back to top]

Median Change From Baseline in Viral Load to Week 32

(NCT00054717)
Timeframe: Baseline to Week 32

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.73
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)-0.24

[back to top]

Median Change From Baseline in Viral Load to Week 4

(NCT00054717)
Timeframe: Baseline to Week 4

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-1.56
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)-0.46

[back to top]

Median Change From Baseline in Viral Load to Week 40

(NCT00054717)
Timeframe: Baseline to Week 40

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.74
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)-0.23

[back to top]

Median Change From Baseline in Viral Load to Week 48

(NCT00054717)
Timeframe: Baseline to Week 48

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.61
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)-0.24

[back to top]

Median Change From Baseline in Viral Load to Week 56

(NCT00054717)
Timeframe: Baseline to Week 56

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.57
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)-0.23

[back to top]

Median Change From Baseline in Viral Load to Week 64

(NCT00054717)
Timeframe: Baseline to Week 64

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.6
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)-0.23

[back to top]

Median Change From Baseline in Viral Load to Week 72

(NCT00054717)
Timeframe: Baseline to Week 72

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.65
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)-0.21

[back to top]

Median Change From Baseline in Viral Load to Week 80

(NCT00054717)
Timeframe: Baseline to Week 80

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.64
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)-0.21

[back to top]

Median Change From Baseline in Viral Load to Week 88

(NCT00054717)
Timeframe: Baseline to Week 88

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.64
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)-0.21

[back to top]

Median Change From Baseline in Viral Load to Week 96

(NCT00054717)
Timeframe: Baseline to Week 96

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.6
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)-0.21

[back to top]

Time to Confirmed Virologic Failure Through 48 Weeks of Treatment

Time to virologic failure is defined as the time from the start of treatment to the last measurement with a viral load reduction greater than 1.0 log before a confirmed drop of viral load reduction below 1.0 log. (NCT00054717)
Timeframe: Week 48

InterventionDays (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)113
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)0

[back to top]

Time to Confirmed Virologic Failure Through 96 Weeks of Treatment

Time to virologic failure is defined as the time from the start of treatment to the last measurement with a viral load reduction greater than 1.0 log before a confirmed drop of viral load reduction below 1.0 log. (NCT00054717)
Timeframe: Week 96

InterventionDays (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)116
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)0

[back to top]

Time to New CDC Class C Progression Event or Death.

Time to new Centers for Disease Control and Prevention (CDC) class C progression event (i.e., new AIDS defining illness) or death (NCT00054717)
Timeframe: after 48 weeks of treatment

InterventionDays (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)448
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)284

[back to top]

Time to Treatment Failure Through 48 Weeks of Treatment

Time to treatment failure is defined as 0 for patients who never achieve TR otherwise time to treatment failure is the earliest time of death, discontinuation of the study drug or introduction of a new anti-retroviral drug to the regimen if it is not solely related to either toxicity or intolerance clearly attributable to a background, or the first of two consecutive visits with VL measurements <1 log10 below baseline. (NCT00054717)
Timeframe: Week 48

InterventionDays (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)113
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)0

[back to top]

Time to Treatment Failure Through 96 Weeks of Treatment

time to treatment failure is defined as 0 for patients who never achieve TR otherwise time to treatment failure is the earliest time of death, discontinuation of the study drug or introduction of a new anti-retroviral drug to the regimen if it is not solely related to either toxicity or intolerance clearly attributable to a background, or the first of two consecutive visits with VL measurements <1 log10 below baseline. (NCT00054717)
Timeframe: Week 96

InterventionDays (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)114
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)0

[back to top]

Treatment Response at Week 16

Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00054717)
Timeframe: week 16

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)45.7
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)27.5

[back to top]

Treatment Response at Week 2

Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00054717)
Timeframe: week 2

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)53.1
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)34.3

[back to top]

Treatment Response at Week 24

Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00054717)
Timeframe: Week 24

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)41.8
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)23.9
Tipranavir(TPV)/Low Dose Ritonavir(r)42.1
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)23.6

[back to top]

Treatment Response at Week 32

Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00054717)
Timeframe: Week 32

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)37.9
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)19.4

[back to top]

Treatment Response at Week 4

Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00054717)
Timeframe: week 4

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)52.4
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)33.3

[back to top]

Treatment Response at Week 40

Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00054717)
Timeframe: Week 40

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)35.4
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)17.8

[back to top]

Treatment Response at Week 48

Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00054717)
Timeframe: At week 48

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)33.80
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)16.20

[back to top]

Treatment Response at Week 48

Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00054717)
Timeframe: Week 48

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)33.8
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)16.2

[back to top]

Treatment Response at Week 56

Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00054717)
Timeframe: week 56

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)32.5
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)15.9

[back to top]

Treatment Response at Week 64

Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00054717)
Timeframe: week 64

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)31.3
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)15.5

[back to top]

Treatment Response at Week 72

Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00054717)
Timeframe: Week 72

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)29.6
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)14.9

[back to top]

Treatment Response at Week 8

Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00054717)
Timeframe: week 8

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)52.4
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)33.3

[back to top]

Treatment Response at Week 80

Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00054717)
Timeframe: Week 80

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)28
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)14.2

[back to top]

Treatment Response at Week 88

Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00054717)
Timeframe: Week 88

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)27.3
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)12.9

[back to top]

Treatment Response at Week 96

Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00054717)
Timeframe: Week 96

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)26.7
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)11.7

[back to top]

Virologic Response (Viral Load >= 1 Log Drop) at Viral Load Nadir, LOCF

Percentage of participants with Viral Load (VL) >= 1 log reduction from baseline (NCT00054717)
Timeframe: Week 2 through Week 96 (at any point during trial)

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)79.1
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)50.5

[back to top]

Virologic Response (Viral Load >= 1 Log Drop) at Week 16

Percentage of participants with Viral Load (VL) >= 1 log reduction from baseline (NCT00054717)
Timeframe: Week 16

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)47.3
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)26.5

[back to top]

Virologic Response (Viral Load >= 1 Log Drop) at Week 2

Percentage of participants with Viral Load (VL) >= 1 log reduction from baseline (NCT00054717)
Timeframe: Week 2

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)63.3
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)40.1

[back to top]

Virologic Response (Viral Load >= 1 Log Drop) at Week 24

Percentage of participants with Viral Load (VL) >= 1 log reduction from baseline (NCT00054717)
Timeframe: Week 24

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)43.1
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)22.3

[back to top]

Virologic Response (Viral Load >= 1 Log Drop) at Week 32

Percentage of participants with Viral Load (VL) >= 1 log reduction from baseline (NCT00054717)
Timeframe: Week 32

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)38.9
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)19.1

[back to top]

Virologic Response (Viral Load >= 1 Log Drop) at Week 4

Percentage of participants with Viral Load (VL) >= 1 log reduction from baseline (NCT00054717)
Timeframe: Week 4

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)63.3
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)40.1

[back to top]

Virologic Response (Viral Load >= 1 Log Drop) at Week 40

Percentage of participants with Viral Load (VL) >= 1 log reduction from baseline (NCT00054717)
Timeframe: Week 40

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)36.7
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)17.5

[back to top]

Virologic Response (Viral Load >= 1 Log Drop) at Week 48

Percentage of participants with Viral Load (VL) >= 1 log reduction from baseline (NCT00054717)
Timeframe: Week 48

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)36.3
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)16.2

[back to top]

Virologic Response (Viral Load >= 1 Log Drop) at Week 56

Percentage of participants with Viral Load (VL) >= 1 log reduction from baseline (NCT00054717)
Timeframe: Week 56

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)34.1
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)15.5

[back to top]

Virologic Response (Viral Load >= 1 Log Drop) at Week 64

Percentage of participants with Viral Load (VL) >= 1 log reduction from baseline (NCT00054717)
Timeframe: Week 64

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)34.1
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)16.2

[back to top]

Virologic Response (Viral Load >= 1 Log Drop) at Week 8

Percentage of participants with Viral Load (VL) >= 1 log reduction from baseline (NCT00054717)
Timeframe: Week 8

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)55.9
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)34.3

[back to top]

Virologic Response (VL < 400 Copies/ml) at Week 16

Percentage of participants with Viral Load < 400 copies/mL (NCT00054717)
Timeframe: Week 16

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)35.7
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)20.7

[back to top]

Virologic Response (VL < 400 Copies/ml) at Week 2

Percentage of participants with Viral Load < 400 copies/mL (NCT00054717)
Timeframe: Week 2

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)14.8
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)13.6

[back to top]

Virologic Response (VL < 400 Copies/ml) at Week 24

Percentage of participants with Viral Load < 400 copies/mL (NCT00054717)
Timeframe: Week 24

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)34.4
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)16.5

[back to top]

Virologic Response (VL < 400 Copies/ml) at Week 32

Percentage of participants with Viral Load < 400 copies/mL (NCT00054717)
Timeframe: week 32

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)32.8
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)14.9

[back to top]

Virologic Response (VL < 400 Copies/ml) at Week 4

Percentage of participants with Viral Load < 400 copies/mL (NCT00054717)
Timeframe: Week 4

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)28.3
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)19.1

[back to top]

Virologic Response (VL < 400 Copies/ml) at Week 40

Percentage of participants with Viral Load < 400 copies/mL (NCT00054717)
Timeframe: Week 40

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)32.8
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)14.6

[back to top]

Virologic Response (VL < 400 Copies/ml) at Week 48

Percentage of participants with Viral Load < 400 copies/mL (NCT00054717)
Timeframe: Week 48

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)30.5
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)13.6

[back to top]

Virologic Response (VL < 400 Copies/ml) at Week 56

Percentage of participants with Viral Load < 400 copies/mL (NCT00054717)
Timeframe: Week 56

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)29.6
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)13.9

[back to top]

Virologic Response (VL < 400 Copies/ml) at Week 64

Percentage of participants with Viral Load < 400 copies/mL (NCT00054717)
Timeframe: Week 64

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)28.6
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)13.3

[back to top]

Virologic Response (VL < 400 Copies/ml) at Week 72

Percentage of participants with Viral Load < 400 copies/mL (NCT00054717)
Timeframe: Week 72

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)28.9
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)12.9

[back to top]

Virologic Response (VL < 400 Copies/ml) at Week 8

Percentage of participants with Viral Load < 400 copies/mL (NCT00054717)
Timeframe: Week 8

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)37.3
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)23.6

[back to top]

Virologic Response (VL < 400 Copies/ml) at Week 80

Percentage of participants with Viral Load < 400 copies/mL (NCT00054717)
Timeframe: Week 80

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)27.7
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)13.6

[back to top]

Virologic Response (VL < 400 Copies/ml) at Week 88

Percentage of participants with Viral Load < 400 copies/mL (NCT00054717)
Timeframe: week 88

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)27.7
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)12.6

[back to top]

Virologic Response (VL < 400 Copies/ml) at Week 96

Percentage of participants with Viral Load < 400 copies/mL (NCT00054717)
Timeframe: week 96

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)26.7
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)11.7

[back to top]

Virologic Response (VL < 50 Copies/ml) at Viral Load Nadir, LOCF

Percentage of participants with Viral Load < 50 copies/mL (NCT00054717)
Timeframe: Week 2 through Week 96 (at any point during trial)

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)37.9
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)20.4

[back to top]

Virologic Response (VL < 50 Copies/ml) at Week 16

Percentage of participants with Viral Load < 50 copies/mL (NCT00054717)
Timeframe: Week 16

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)23.5
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)9.4

[back to top]

Virologic Response (VL < 50 Copies/ml) at Week 2

Percentage of participants with Viral Load < 50 copies/mL (NCT00054717)
Timeframe: Week 2

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)1.9
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)1.9

[back to top]

Virologic Response (VL < 50 Copies/ml) at Week 24

Percentage of participants with Viral Load < 50 copies/mL (NCT00054717)
Timeframe: Week 24

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)24.8
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)10

[back to top]

Virologic Response (VL < 50 Copies/ml) at Week 32

Percentage of participants with Viral Load < 50 copies/mL (NCT00054717)
Timeframe: Week 32

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)24.8
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)10

[back to top]

Virologic Response (VL < 50 Copies/ml) at Week 4

Percentage of participants with Viral Load < 50 copies/mL (NCT00054717)
Timeframe: Week 4

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)6.4
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)4.2

[back to top]

Virologic Response (VL < 50 Copies/ml) at Week 40

Percentage of participants with Viral Load < 50 copies/mL (NCT00054717)
Timeframe: Week 40

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)23.5
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)8.4

[back to top]

Virologic Response (VL < 50 Copies/ml) at Week 48

Percentage of participants with Viral Load < 50 copies/mL (NCT00054717)
Timeframe: Week 48

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)22.5
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)9.7

[back to top]

Virologic Response (VL < 50 Copies/ml) at Week 56

Percentage of participants with Viral Load < 50 copies/mL (NCT00054717)
Timeframe: Week 56

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)21.5
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)10

[back to top]

Virologic Response (VL < 50 Copies/ml) at Week 64

Percentage of participants with Viral Load < 50 copies/mL (NCT00054717)
Timeframe: Week 64

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)22.8
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)10.7

[back to top]

Virologic Response (VL < 50 Copies/ml) at Week 72

Percentage of participants with Viral Load < 50 copies/mL (NCT00054717)
Timeframe: Week 72

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)21.2
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)11.3

[back to top]

Virologic Response (VL < 50 Copies/ml) at Week 8

Percentage of participants with Viral Load < 50 copies/mL (NCT00054717)
Timeframe: Week 8

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)16.1
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)8.1

[back to top]

Virologic Response (VL < 50 Copies/ml) at Week 80

Percentage of participants with Viral Load < 50 copies/mL (NCT00054717)
Timeframe: Week 80

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)20.6
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)10.7

[back to top]

Virologic Response (VL < 50 Copies/ml) at Week 88

Percentage of participants with Viral Load < 50 copies/mL (NCT00054717)
Timeframe: Week 88

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)22.2
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)9.7

[back to top]

Virologic Response (VL < 50 Copies/ml) at Week 96

Percentage of participants with Viral Load < 50 copies/mL (NCT00054717)
Timeframe: Week 96

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)20.9
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)9.4

[back to top]

Percentage of Patients With Division of Acquired Immunodeficiency Syndrome (DAIDS) Grade 3 or 4 Laboratory Abnormalities

NIH Division of Acquired Immunodeficiency Syndrome (DAIDS) Table for Grading Severity of Adult Adverse Experiences, December 2004. (NCT00054717)
Timeframe: 240 Weeks

,
InterventionPercentage of participants (Number)
HaemoglobinWhite Blood Cell Count IncreaseWhite Blood Cell Count DecreasePlateletsProthrombin TimeAlanine Transaminase (ALT)Aspartate Transaminase (AST)ALT or ASTBilirubin, TotalAlkaline phosphataseAmylaseLipaseCholesterolTriglyceridesGlucose, increaseGlucose, decreaseCreatinine
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)0.705.30.72.01.61.62.60.709.21.38.615.52.01.02.3
Tipranavir(TPV)/Low Dose Ritonavir(r)1.305.32.01.613.57.915.11.01.011.82.023.729.33.302.0

[back to top]

Virologic Response (VL < 400 Copies/ml) at Viral Load Nadir, LOCF

Percentage of participants with Viral Load < 400 copies/mL (NCT00054717)
Timeframe: Week 2 through Week 96 (at any point during trial)

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)53.4
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)32.4

[back to top]

Median Change From Baseline in Viral Load to Week 8

(NCT00054717)
Timeframe: Baseline to Week 8

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-1.6
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)-0.43

[back to top]

Median Change From Baseline in Viral Load to Week 16

(NCT00054717)
Timeframe: Baseline to Week 16

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-1.08
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)-0.32

[back to top]

Mean Change From Baseline to Week 16 in CD4+ Cell Count

(NCT00054717)
Timeframe: Baseline to Week 16

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)55
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)30

[back to top]

Mean Change From Baseline to Week 2 in CD4+ Cell Count

(NCT00054717)
Timeframe: Baseline to Week 2

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)23
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)19

[back to top]

Mean Change From Baseline to Week 24 in CD4+ Cell Count

(NCT00054717)
Timeframe: Baseline to Week 24

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)54
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)24

[back to top]

Mean Change From Baseline to Week 32 in CD4+ Cell Count

(NCT00054717)
Timeframe: Baseline to Week 32

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)51
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)26

[back to top]

Mean Change From Baseline to Week 4 in CD4+ Cell Count

(NCT00054717)
Timeframe: Baseline to Week 4

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)41
Comparitor Protease Inhibitor(CPR)/Low Dose Ritonavir(r)22

[back to top]

Number of Confirmed Virologic Failure Participants (Par.) With Treatment-emergent Reductions in Drug Susceptibility (DS)

A blood sample was drawn for par. failing to respond to therapy, and changes in DS for HIV isolated from the par. for each drug used in the study were assessed. The changes in DS detected by phenotypic assay in virus from the sample collected at the time of failure was compared with DS in the virus from the blood sample at baseline. Par. are grouped by study arm and prior therapy experience. DS is the state of HIV being susceptible to the antiretroviral agent (the virus can be inhibited by the drug). Reduced DS (i.e., HIV is resistant to the antiretroviral agent) can lead to treatment failure. (NCT00071760)
Timeframe: Baseline through Week 48

,,
Interventionparticipants (Number)
Any NRTIEmtricitabineLamivudineAny NNRTIAny PIRitonavir-boosted Fosamprenavir
ART-experienced, PI-naïve FPV/RTV Treatment Group000000
ART-naïve FPV/RTV Treatment Group111000
PI-experienced, ART-experienced FPV/RTV Treatment Group000011

[back to top]

Number of Participants With Plasma HIV-1 Ribonucleic Acid (RNA) <400 Copies Per Milliliter at Baseline and Weeks 4, 12, 24, 36, and 48 (MSD=F)

Blood samples of participants were collected to measure plasma HIV-1 RNA concentrations. Viral load, measured in RNA copies per milliliter of plasma, is an efficacy measure for antiretroviral drugs. In the Missing, Switch, or Discontinuation=Failure (MSD=F) analysis, participants who had missing data at or had discontinued the study prior to a certain time point or had changed their background antiretroviral regimen are classified as non-responders. (NCT00071760)
Timeframe: Baseline and Weeks 4, 12, 24, 36, and 48

,
Interventionparticipants (Number)
BaselineWeek 4Week 12Week 24Week 36Week 48
Cohort 1, Arm A - 6 Months to <2 Years0618202022
Cohort 2, Arm A - 4 Weeks to <6 Months1513181915

[back to top]

Number of Participants With Hematology Toxicities

Blood samples were collected for the analysis of all hematology parameters. Laboratory toxicities were graded using the DAIDS Table for grading the severity of adult and pediatric adverse events, where grade 1=mild, grade 2=moderate, grade 3=severe and grade 4=potentially life-threatening. Results for participants with Grade 1 to 4 hematology toxicities is presented. (NCT00071760)
Timeframe: Day 1 and up to Week 684

,
InterventionParticipants (Count of Participants)
Grade 1Grade 2Grade 3Grade 4
Cohort 1, Arm A - 6 Months to <2 Years6622
Cohort 2, Arm A - 4 Weeks to <6 Months6530

[back to top]

Number of Participants With Clinical Chemistry Toxicities

Blood samples were collected for the analysis of all clinical chemistry parameters. Laboratory toxicities were graded using the Division of AIDS (DAIDS) table for grading the severity of adult and pediatric adverse events, where grade 1=mild, grade 2=moderate, grade 3=severe and grade 4=potentially life-threatening. Results for participants with Grade 1 to 4 clinical chemistry toxicities is presented. (NCT00071760)
Timeframe: Day 1 and up to Week 684

,
InterventionParticipants (Count of Participants)
Grade 1Grade 2Grade 3Grade 4
Cohort 1, Arm A - 6 Months to <2 Years51471
Cohort 2, Arm A - 4 Weeks to <6 Months5931

[back to top]

Absolute Values of Cholesterol, Glucose, High Density Lipoprotein (HDL) Cholesterol, Low Density Lipoprotein (LDL) and Triglyceride (TG) at Baseline (Day 1), Weeks 4, 12, 24, 36, and 48

Blood samples of all participants were generally collected under non-fasting conditions (given the age of participants) for the evaluation of cholesterol, serum glucose, HDL cholesterol, LDL cholesterol and triglyceride (TG). Clinical chemistry analyses were carried out using the observed analysis strategy. (NCT00071760)
Timeframe: Baseline (Day 1) and Weeks 4, 12, 24, 36, and 48

InterventionMillimoles per liter (mmol/L) (Median)
Cholesterol, Day 1; n=22;29Cholesterol, Week 4; n=4;2Cholesterol, Week 24; n=22;26Cholesterol, Week 36; n=3;2Cholesterol, Week 48; n=18;24Glucose, Day 1; n=26;30Glucose, Week 4; n=23;28Glucose, Week 12; n=22;27Glucose, Week 24; n=22;26Glucose, Week 36; n=20;25Glucose, Week 48; n=18;24HDL, Day 1; n=22;29HDL, Week 4; n=4;2HDL, Week 24; n=22;26HDL, Week 36; n=3;2HDL, Week 48; n=18;24LDL, Day 1; n=22;28LDL, Week 4; n=4;2LDL, Week 24; n=22;26LDL, Week 36; n=3;2LDL, Week 48; n=18;24TG, Day 1; n=22;29TG, Week 4; n=4;2TG, Week 24; n=22;26TG, Week 36; n=3;2TG, Week 48; n=18;24
Cohort 2, Arm A - 4 Weeks to <6 Months2.2353.0854.1753.0303.8904.804.604.704.554.504.450.6550.8250.9000.6400.8250.901.252.081.902.331.6952.0152.1551.3101.590

[back to top]

Number of Participants With at Least a 1.0 log10 HIV-1 RNA Decrease From Baseline at Weeks 4, 12, 24, 36, and 48 (MSD=F Analysis)

Blood samples of participants were collected to assess the decrease in the number of HIV-1 RNA copies. In the MSD=F analysis, participants who had missing data at or had discontinued the study prior to a certain time point or had changed their background antiretroviral regimen are classified as non-responders. (NCT00071760)
Timeframe: Baseline and Weeks 4, 12, 24, 36, and 48

,
Interventionparticipants (Number)
Week 4Week 12Week 24Week 36Week 48
Cohort 1, Arm A - 6 Months to <2 Years2525232221
Cohort 2, Arm A - 4 Weeks to <6 Months1621211916

[back to top]

Number of Participants Reporting Perfect Adherence at Weeks 2, 12, 24, and 48 as Assessed by the Study Coordinator Using the Adherence Questionnaire

A separate questionnaire was administered for FPV and RTV. Items 1-4 of the Adherence Questionnaire measured a participant's adherence with FPV or RTV during the last 3 days and the weekend prior to the indicated study visits. Perfect adherence was defined as not missing any doses of FPV or RTV since the last study visit. (NCT00071760)
Timeframe: Weeks 2, 12, 24, and 48

,
Interventionparticipants (Number)
Week 2Week 12Week 24Week 48
Cohort 1, Arm A - 6 Months to <2 Years24242117
Cohort 2, Arm A - 4 Weeks to <6 Months18171913

[back to top]

Number of Participants With Treatment-Emergent Changes in HIV-1 Phenotypic Drug Susceptibility

A blood sample was drawn from participant failing to respond to therapy, and the mutations present in the virus were identified. Phenotypic resistance was assessed for virologic failure population and evaluated for Protease Inhibitors (PIs), Nucleotide reverse transcriptase inhibitors (NRTIs), and non-NRTIs (NNTRIs) using Monogram PhenoSense Assay. Virologic failure was defined as having failed to achieve a plasma HIV-RNA of <400 copies/mL by Week 24 or having had a confirmed HIV-RNA rebound to ≥400 c/mL at any time after achieving a plasma HIV-RNA of <400 c/mL. (NCT00071760)
Timeframe: Post-Week 48 to Week 684

,
InterventionParticipants (Count of Participants)
Treatment emergent reduced NRTI drug susceptibilityTreatment emergent reduced NNRTI drug susceptibilityTreatment emergent reduced PI drug susceptibility
ART-Naive000
PI-Naive000

[back to top]

Number of Confirmed Virologic Failure Participants (Par.) With Treatment-emergent HIV Genotypic Resistance in Reverse Transcriptase and Protease

A blood sample was drawn for par. failing to respond to therapy, and the mutations present in the virus were identified. For each par., the mutations found at the time of failure were compared with any mutations found in the blood sample at baseline. New International AIDS Society-USA defined resistance mutations that developed at the time of failure were tabulated by drug class. VF, virologic failure; NRTI, nucleoside reverse transcriptase inhibitor; NNRTI, non-nucleoside reverse transcriptase inhibitor; PI, protease inhibitor. Par. are grouped by study arm and prior therapy experience. (NCT00071760)
Timeframe: Baseline through Week 48

,,
Interventionparticipants (Number)
Any HIV NRTI MutationHIV NRTI mutation M184VAny HIV NNRTI MutationHIV NNRTI Mutation K101K/EAny HIV Major PI MutationsAny Minor HIV PI MutationsMinor HIV PI Mutation L10FMinor HIV PI Mutation L10IMinor HIV PI Mutation L33L/FMinor HIV PI Mutation L33F
ART-naïve FPV/RTV Treatment Group1100020110
PI-experienced, ART-experienced FPV/RTV Treatment Group0011011001
PI-naïve, ART-experienced FPV/RTV Treatment Group0000000000

[back to top]

Median Plasma HIV-1 RNA (log10 Copies/mL) at Baseline and Weeks 4, 12, 24, 36, and 48 (Observed Analysis)

Blood samples of participants were collected to assess the decrease in the number of HIV-1 RNA copies. (NCT00071760)
Timeframe: Baseline and Weeks 4, 12, 24, 36, and 48

,
Interventionlog10 copies/mL (Median)
Baseline, n=26;28Week 4, n=21;28Week 12, n=22;27Week 24, n=22;25Week 36, n=21;25Week 48, n=18;24
Cohort 1, Arm A - 6 Months to <2 Years5.512.882.071.741.691.69
Cohort 2, Arm A - 4 Weeks to <6 Months5.803.292.281.691.691.69

[back to top]

Median Percent Cluster of Differentiation Antigen 4 (CD4+) Cell Count at Baseline and at Weeks 4, 12, 24, 36, and 48

Blood samples of participants were collected for the measurement of the percentage of total lymphocytes that are CD4+ cells. Observed analysis was used for the summary of proportion endpoints using viral load data. CD4+ cells are white blood cells that are important in fighting infection. HIV infects CD4+ cells, replicates in them, and destroys them. A CD4+ cell count provides a measure of the status of the immune system and to what extent it is affected by HIV. (NCT00071760)
Timeframe: Baseline and Weeks 4, 12, 24, 36, and 48

,
InterventionPercentage of cells (Median)
Baseline, n=23;28Week 4, n=23;28Week 12, n=22;27Week 24, n=22;26Week 36, n=21;25Week 48, n=18;24
Cohort 1, Arm A - 6 Months to <2 Years2527.53232.829.531.6
Cohort 2, Arm A - 4 Weeks to <6 Months272927.531.53228

[back to top]

Median Percent Change From Baseline in CD4+ Cell Count at Weeks 4, 12, 24, 36, and 48

Blood samples of participants were collected for the measurement of the percentage of total lymphocytes that are CD4+ cells. Observed analysis was used for the summary of proportion endpoints using viral load data. Change from Baseline in percentage was calculated as the value at indicated time points minus the value at Baseline. (NCT00071760)
Timeframe: Baseline and Weeks 4, 12, 24, 36, and 48

,
InterventionPercentage of cells (Median)
Week 4, n=20;28Week 12, n=19;27Week 24, n=19;26Week 36, n=18;25Week 48, n=15;24
Cohort 1, Arm A - 6 Months to <2 Years3.46654.7
Cohort 2, Arm A - 4 Weeks to <6 Months32785

[back to top]

Number of Participants With the Indicated Treatment-emergent (TE) Grade 3/4 Laboratory Abnormalities

TE toxicities were presented for each laboratory parameter. A toxicity was considered TE if it was greater than the Baseline grade, and if it was observed on/after the date of the first dose of study drug (SD), and on/before the date of the last dose of SD. Per the Division of AIDS Table for Grading the Severity of Adult and Pediatric AEs, Grade 3=severe; Grade 4=potentially life-threatening. (NCT00071760)
Timeframe: Baseline (Day 1) until Week 48

,
Interventionparticipants (Number)
Clinical Chemistry Toxicities - Grade 3Clinical Chemistry Toxicities - Grade 4Hematology Toxicities - Grade 3Hematology Toxicities - Grade 4
Cohort 1, Arm A - 6 Months to <2 Years4121
Cohort 2, Arm A - 4 Weeks to <6 Months2020

[back to top]

Number of Participants With the Indicated Virological Outcome at Week 48

Blood samples of participants were collected to measure plasma HIV-1 RNA concentrations. PI-exp = PI-experienced. Virologic success was defined as plasma HIV-1 RNA <400 copies/mL. Virologic failure: (1) HIV-1 RNA >=400 copies/mL, (2) change of background antiretroviral treatment (ART), (3) discontinued study due to lack of efficacy, (4) discontinued study with last HIV-1 >=400 copies/mL. No virologic data at Week 48 window: (a) discontinued study due to an adverse event or death, (b) discontinued study due to other reasons (withdrew consent, loss to follow-up, moved, etc.). (NCT00071760)
Timeframe: Week 48

,
Interventionparticipants (Number)
Virological (V) successV failure (1)V failure (2)V failure (3)V failure (4)No V data at Week 48 (a)No V data at Week 48 (b)
Cohort 1, Arm A - 6 Months to <2 Years20221111
Cohort 2, Arm A - 4 Weeks to <6 Months15301214

[back to top]

Number of Participants With the Indicated Response Scores for the Parent/Guardian (P/G) Perception of FPV Oral Suspension Questionnaire: Items 1 to 4

P/G perceptions of FPV/RTV BID were assessed using a P/G Perception of Study Medication questionnaire administered during Weeks 2, 24, and 48/premature study discontinuation. Questions 1 to 4 ask directly about the P/G's assessment of 1=color, 2=texture/consistency, 3=odor, and 4=general satisfaction. Questions 5 to 10 ask about the P/G's perception of the child's assessment of the oral suspension. Data are reported as the number of participants with the indicated response by question, response category (1-3=dislike, 4=neutral, 5-7=like), and timing of visit. (NCT00071760)
Timeframe: Weeks 2, 24, and 48/premature study discontinuation

,
Interventionparticipants (Number)
Week 2, color, dislike, n=23;27Week 2, color, neutral, n=23;27Week 2, color, like, n=23;27Week 24, color, dislike, n=20;24Week 24, color, neutral, n=20;24Week 24, color, like, n=20;24Week 48, color, dislike, n=15;22Week 48, color, neutral, n=15;22Week 48, color, like, n=15;22Week 2, texture, dislike, n=23;27Week 2, texture, neutral, n=23;27Week 2, texture, like, n=23;27Week 24, texture, dislike, n=20;24Week 24, texture, neutral, n=20;24Week 24, texture, like, n=20;24Week 48, texture, dislike, n=15;21Week 48, texture, neutral, n=15;21Week 48, texture, like, n=15;21Week 2, odor, dislike, n=23;27Week 2, odor, neutral, n=23;27Week 2, odor, like, n=23;27Week 24, odor, dislike, n=20;24Week 24, odor, neutral, n=20;24Week 24, odor, like, n=20;24Week 48, odor, dislike, n=15;22Week 48, odor, neutral, n=15;22Week 48, odor, like, n=15;22Week 2, general satisfaction, dislike, n=23;27Week 2, general satisfaction, neutral, n=23;27Week 2, general satisfaction, like, n=23;27Week 24, general satisfaction, dislike, n=20;24Week 24, general satisfaction, neutral, n=20;24Week 24, general satisfaction, like, n=20;24Week 48, general satisfaction, dislike, n=15;22Week 48, general satisfaction, neutral, n=15;22Week 48, general satisfaction, like, n=15;22
Cohort 1, Arm A - 6 Months to <2 Years513941010778510124128678771378975105715699949
Cohort 2, Arm A - 4 Weeks to <6 Months3713421461847126410618131942144110341640164110

[back to top]

Number of Participants With Treatment-Emergent Mutations at Virologic Failure Timepoint

A blood sample was drawn from participants failing to respond to therapy, and genotyping was performed to identify the mutations present in the baseline (pre-therapy) and the sample obtained at virologic failure (VF). For each participant, the HIV-1 mutations found at the time of failure were compared with any HIV-1 mutations detected in the blood sample at baseline. New International AIDS Society-USA defined resistance mutations that developed at the time of failure were tabulated by drug class- NRTI, nucleoside reverse transcriptase inhibitor; NNRTI, non-nucleoside reverse transcriptase inhibitor; PI, protease inhibitor. Participants are grouped by study arm (prior therapy experience), results displayed in this table are from participants who met virologic failure criteria from Week 48 through Week 684. (NCT00071760)
Timeframe: Week 48 to Week 684

,
InterventionParticipants (Count of Participants)
PRO, L10L/RPRO, I62I/V
ART-Naive11
PI-Naive00

[back to top]

Plasma Amprenavir (APV) AUC (0-tau[τ])

"Plasma samples were assayed for APV concentrations using a validated assay. The GlaxoSmithKline (GSK) Department of Clinical Pharmacology Modeling and Simulation conducted pharmacokinetic (PK) analysis of the plasma APV concentration-time data using a model-independent approach. As a measure of total drug exposure, the area under the plasma-concentration-versus-time curve over the dosing interval at steady-state (AUC[0-τ]), where τ is the length of the dosing interval, was calculated by the linear up/log down trapezoidal method. hours, hr." (NCT00071760)
Timeframe: Week 48

InterventionHr per microgram/milliliter (hr*µg/mL) (Geometric Mean)
45/10 mg/kg BID; n=9, 160/10 mg/kg BID; n=2, 245/7 mg/kg BID; n=2, 10
FPV/RTV BID: 4 Weeks to <6 Months26.654.235.08

[back to top]

Plasma Amprenavir (APV) AUC (0-tau[τ])

"Plasma samples were assayed for APV concentrations using a validated assay. The GlaxoSmithKline (GSK) Department of Clinical Pharmacology Modeling and Simulation conducted pharmacokinetic (PK) analysis of the plasma APV concentration-time data using a model-independent approach. As a measure of total drug exposure, the area under the plasma-concentration-versus-time curve over the dosing interval at steady-state (AUC[0-τ]), where τ is the length of the dosing interval, was calculated by the linear up/log down trapezoidal method. hours, hr." (NCT00071760)
Timeframe: Week 48

InterventionHr per microgram/milliliter (hr*µg/mL) (Geometric Mean)
45/10 mg/kg BID; n=9, 160/10 mg/kg BID; n=2, 245/7 mg/kg BID; n=2, 1060/7 mg/kg BID; n=0, 8
FPV/RTV BID: 6 Months to <2 Years64.5126.2227.548.4

[back to top]

Median Change From Baseline in Plasma HIV-1 RNA (log10 Copies/mL) at Weeks 4, 12, 24, 36, and 48 (Observed Analysis)

Blood samples of participants were collected to assess the decrease in the number of HIV-1 RNA copies. Change from Baseline in plasma HIV-1 RNA was calculated as the value at the indicated time point minus the value at Baseline. (NCT00071760)
Timeframe: Baseline and Weeks 4, 12, 24, 36, and 48

,
Interventionlog10 copies/mL (Median)
Week 4, n=21;28Week 12, n=22;27Week 24, n=22;25Week 36, n=21;25Week 48, n=18;24
Cohort 1, Arm A - 6 Months to <2 Years-2.33-3.14-3.38-3.52-3.56
Cohort 2, Arm A - 4 Weeks to <6 Months-2.38-3.11-3.77-3.72-3.90

[back to top]

Change From Baseline in Plasma HIV-1 RNA Levels at Each Study Visit

Blood samples of participants were collected to measure plasma HIV-1 RNA copies. Baseline value was defined as the value observed at the day 1 visit or if this value is missing the last value observed before the start of investigational product. Change from baseline value was defined as post-dose value minus baseline value. (NCT00071760)
Timeframe: Baseline (Day 1) and up to Week 684

InterventionLog10 copies/milliliter (Median)
Week 4Week 12Week 24Week 36Week 48Week 60Week 72Week 84Week 96Week 108Week 120Week 132Week 144Week 156Week 168Week 180Week 192Week 204Week 216Week 228Week 240Week 252Week 264Week 276Week 288Week 300Week 312Week 324Week 336Week 348Week 360Week 372Week 384Week 396Week 408Week 420Week 432Week 444Week 456Week 468Week 480Week 492Week 504Week 516Week 528Week 540Week 552Week 564Week 576Week 588Week 600Week 612Week 624Week 636Week 648Week 660Week 672Week 684
Cohort 1, Arm A - 6 Months to <2 Years-2.33-3.14-3.38-3.52-3.56-3.12-3.60-3.62-3.70-3.80-3.58-3.52-3.52-3.34-3.52-3.52-3.31-3.52-3.52-3.71-3.71-3.71-3.71-3.71-3.71-3.80-3.98-3.87-3.62-3.36-3.62-3.36-3.15-3.62-3.62-3.62-3.62-3.62-3.62-3.62-3.62-3.62-3.62-3.62-3.80-3.99-3.99-3.99-3.99-3.99-3.99-3.99-3.99-4.27-4.27-4.27-4.27-3.99

[back to top]

Change From Baseline in Plasma HIV-1 RNA Levels at Each Study Visit

Blood samples of participants were collected to measure plasma HIV-1 RNA copies. Baseline value was defined as the value observed at the day 1 visit or if this value is missing the last value observed before the start of investigational product. Change from baseline value was defined as post-dose value minus baseline value. (NCT00071760)
Timeframe: Baseline (Day 1) and up to Week 684

InterventionLog10 copies/milliliter (Median)
Week 4Week 12Week 24Week 36Week 48Week 60Week 72Week 84Week 96Week 108Week 120Week 132Week 144Week 156Week 168Week 180Week 192Week 204Week 216Week 228Week 240Week 252Week 264Week 276Week 288Week 300Week 312Week 324Week 336Week 348Week 360Week 372Week 384Week 396Week 408Week 420Week 432Week 444Week 456Week 468Week 480Week 492Week 504Week 516Week 528Week 540Week 552Week 564Week 576Week 588Week 600Week 612Week 624Week 636Week 648Week 660
Cohort 2, Arm A - 4 Weeks to <6 Months-2.38-3.11-3.77-3.72-3.90-3.89-3.95-3.95-4.26-4.26-4.12-4.09-4.12-4.12-3.95-4.36-4.28-4.22-4.36-4.45-4.22-4.22-4.22-4.22-4.22-4.22-4.05-4.01-4.09-4.33-4.09-4.09-4.01-4.09-4.09-4.09-4.33-4.04-4.09-4.09-4.09-3.55-4.09-4.09-4.09-4.09-4.09-4.28-4.04-4.04-3.55-4.09-4.09-4.00-4.00-4.09

[back to top]

Number of Participants With the Indicated Response Scores for the Parent/Guardian Perception of the Child's Assessment of FPV Oral Suspension Questionnaire: Items (I) 5 to 10

Parent/guardian perceptions of FPV/RTV BID was assessed using a Parent/Guardian Perception of Study Medication questionnaire. Questions 1 to 4 ask directly about the parent/guardian's assessment of the color, texture/consistency, odor, and general satisfaction. Questions 5 to 10 ask about the parent/guardian's perception of the child's assessment of the oral suspension (Items: 5=reaction to new medicine [med.]; 6=taste; 7=acceptance; 8=swallowing; 9=willingness compared to other med.; 10=overall liking. Data for items 6/10 are reported in response categories: 1-3=dislike; 4=neutral; 5-7=like. (NCT00071760)
Timeframe: Weeks (W) 2, 24, and 48/premature study discontinuation

,
Interventionparticipants (Number)
W2, Item 5, takes all/most med. easily, n=23;27W2, Item 5, problem taking a few med., n=23;27W2, Item 5, problem taking most med., n=23;27W2, Item 5, impossible to take med., n=23;27W24, Item 5, takes all/most med. easily, n=20;23W24, Item 5, problem taking a few med., n=20;23W24, Item 5, problem taking most med., n=20;23W24, Item 5, impossible to take med., n=20;23W48, Item 5, takes all/most med. easily, n=14;22Week 48, Item 5, problem taking a few med., n=14;22W48, Item 5, problem taking most med., n=14;22W48, Item 5, impossible to take med., n=14;22W2, Item 6, dislike, n=23;27W2, Item 6, neutral, n=23;27W2, Item 6, like, n=23;27W24, Item 6, dislike, n=20;23W24, Item 6, neutral, n=20;23W24, Item 6, like, n=20;23W48, Item 6, dislike, n=15;22W48, Item 6, neutral, n=15;22W48, Item 6, like, n=15;22W2, Item 7, no problem taking FPV, n=23;27W2, Item 7, few problems taking FPV, n=23;27W2, Item 7, problem taking most of time, n=23;27W2, Item 7, impossible to take, n=23;27W24, Item 7, no problem taking FPV, n=20;23W24, Item 7, few problems taking FPV, n=20;23W24, Item 7, problem taking most of time, n=20;23W24, Item 7, impossible to take, n=20;23W48, Item 7, no problem taking FPV, n=15;22W48, Item 7, few problems taking FPV, n=15;22W48, Item 7, problem taking most of time, n=15;22W48, Item 7, impossible to take, n=15;22W2, Item 8, swallows with no problem, n=23;27W2, Item 8, swallows with struggle, n=23;27W2, Item 8, spits out suspension, n=23;27W2, Item 8, vomits the suspension, n=23;27W24, Item 8, swallows with no problem, n=20;23W24, Item 8, swallows with struggle, n=20;23W24, Item 8, spits out suspension, n=20;23W24, Item 8, vomits the suspension, n=20;23W48, Item 8, swallows with no problem, n=15;22W48, Item 8, swallows with struggle, n=15;22W48, Item 8, spits out suspension, n=15;22W48, Item 8, vomits the suspension, n=15;22W2, I9, take more willingly than other med., n=23;27W2, Item 9, about the same, n=23;27W2, I9, not as willing to take as other med., n=23;27W24, I9, take more willingly than other med., n=20;22W24, Item 9, about the same, n=20;22W24, I9, not as willing to take as other med, n=20;22W48, I9, take more willingly than other med., n=15;22W48, Item 9, about the same, n=15;22W48, I9, not as willing to take as other med, n=15;22Week 2, Item 10, dislike, n=23;27Week 2, Item 10, neutral, n=23;27Week 2, Item 10, like, n=23;27Week 24, Item 10, dislike, n=20;24Week 24, Item 10, neutral, n=20;24Week 24, Item 10, like, n=20;24Week 48, Item 10, dislike, n=15;22Week 48, Item 10, neutral, n=15;22Week 48, Item 10, like, n=15;22
Cohort 1, Arm A - 6 Months to <2 Years111051107511750091081256106669102105801082210105212920128113159120161429991374877
Cohort 2, Arm A - 4 Weeks to <6 Months101030128001040012471136915795213340842191103107301131031373116510011577211744

[back to top]

Absolute Values of Serum Lipase at Baseline (Day 1), Weeks 4, 12, 24, and 48

Blood samples of all participants were collected for the evaluation of serum lipase. Clinical chemistry analyses were carried out using the observed analysis strategy. Change from Baseline in serum lipase was calculated as the value at the indicated time point minus the value at Baseline. (NCT00071760)
Timeframe: Baseline (Day 1) and Weeks 4, 12, 24, and 48

InterventionUnits per liter (U/L) (Median)
Day 1; n=2;11Week 4; n=0;1Week 12; n=0;1Week 24; n=0;8Week 48; n=1;8
Cohort 1, Arm A - 6 Months to <2 Years22.016.068.019.516.0

[back to top]

Absolute Values of Serum Lipase at Baseline (Day 1), Weeks 4, 12, 24, and 48

Blood samples of all participants were collected for the evaluation of serum lipase. Clinical chemistry analyses were carried out using the observed analysis strategy. Change from Baseline in serum lipase was calculated as the value at the indicated time point minus the value at Baseline. (NCT00071760)
Timeframe: Baseline (Day 1) and Weeks 4, 12, 24, and 48

InterventionUnits per liter (U/L) (Median)
Day 1; n=2;11Week 48; n=1;8
Cohort 2, Arm A - 4 Weeks to <6 Months20.516.0

[back to top]

Absolute Values of Cluster of Differentiation 4 (CD4+) Cell Counts by Study Visit

Blood samples of participants were collected for the measurement of the CD4+ cells. (NCT00071760)
Timeframe: Baseline (Day 1) and up to week 684

InterventionCells/cubic millimeter (Median)
Baseline (Day 1)Week 4Week 12Week 24Week 36Week 48Week 60Week 72Week 84Week 96Week 108Week 120Week 132Week 144Week 156Week 168Week 180Week 192Week 204Week 216Week 228Week 240Week 252Week 264Week 276Week 288Week 300Week 312Week 324Week 336Week 348Week 360Week 372Week 384Week 396Week 408Week 420Week 432Week 444Week 456Week 468Week 480Week 492Week 504Week 516Week 528Week 540Week 552Week 564Week 576Week 588Week 600Week 612Week 624Week 636Week 648Week 660Week 672Week 684
Cohort 1, Arm A - 6 Months to <2 Years11201734.518221475.515231602131116671490.514601343.51148.5144015621307123697813271082909110610671098.5853970911.5990928.512421160976962926884900938.5850918860832863101093091099489287710008049901100717949720535455610710580

[back to top]

Absolute Values of Cluster of Differentiation 4 (CD4+) Cell Counts by Study Visit

Blood samples of participants were collected for the measurement of the CD4+ cells. (NCT00071760)
Timeframe: Baseline (Day 1) and up to week 684

InterventionCells/cubic millimeter (Median)
Baseline (Day 1)Week 4Week 12Week 24Week 36Week 48Week 60Week 72Week 84Week 96Week 108Week 120Week 132Week 144Week 156Week 168Week 180Week 192Week 204Week 216Week 228Week 240Week 252Week 264Week 276Week 288Week 300Week 312Week 324Week 336Week 348Week 360Week 372Week 384Week 396Week 408Week 420Week 432Week 444Week 456Week 468Week 480Week 492Week 504Week 516Week 528Week 540Week 552Week 564Week 576Week 588Week 600Week 612Week 624Week 636Week 648Week 660
Cohort 2, Arm A - 4 Weeks to <6 Months13781610140515951780169018901407156015301440148012851250142215501331.513391160133611361132.5137911091180.5106310629249808561030115110519509711005840940.581110039101063.5839887776860851756908792873865.5720753770720750

[back to top]

Plasma APV CL/F Following Dosing Expressed in mL/Min

Apparent clearance of drug from plasma following extravascular administration (CL/F) was calculated as dose/AUC(0-τ). For FPV, doses were expressed in APV molar equivalents (50 mg of FPV = 43.2 mg of APV). (NCT00071760)
Timeframe: Week 48

InterventionmL/min (Geometric Mean)
45/10 mg/kg BID; n=9, 160/10 mg/kg BID; n=2, 245/7 mg/kg BID; n=2, 10
FPV/RTV BID: 4 Weeks to <6 Months13586.4106.7

[back to top]

Plasma APV CL/F Following Dosing Expressed in mL/Min

Apparent clearance of drug from plasma following extravascular administration (CL/F) was calculated as dose/AUC(0-τ). For FPV, doses were expressed in APV molar equivalents (50 mg of FPV = 43.2 mg of APV). (NCT00071760)
Timeframe: Week 48

InterventionmL/min (Geometric Mean)
45/10 mg/kg BID; n=9, 160/10 mg/kg BID; n=2, 245/7 mg/kg BID; n=2, 1060/7 mg/kg BID; n=0, 8
FPV/RTV BID: 6 Months to <2 Years62.5234.3190172

[back to top]

Plasma APV CL/F Following Dosing Expressed in mL/Min/kg

Apparent clearance of drug from plasma following extravascular administration (CL/F) was calculated using the formulation: APV Dose in mg/kg units divided by AUC(0-τ). For FPV, doses were expressed in APV molar equivalents (50 mg of FPV = 43.2 mg of APV). Normalizing CL/F for bodyweight allows for comparison of CL/F across populations. (NCT00071760)
Timeframe: Week 48

InterventionMilliliters/minute/kilogram (mL/min/kg) (Geometric Mean)
45/10 mg/kg BID; n=9, 160/10 mg/kg BID; n=2, 245/7 mg/kg BID; n=2, 10
FPV/RTV BID: 4 Weeks to <6 Months22.915.317.50

[back to top]

Plasma APV CL/F Following Dosing Expressed in mL/Min/kg

Apparent clearance of drug from plasma following extravascular administration (CL/F) was calculated using the formulation: APV Dose in mg/kg units divided by AUC(0-τ). For FPV, doses were expressed in APV molar equivalents (50 mg of FPV = 43.2 mg of APV). Normalizing CL/F for bodyweight allows for comparison of CL/F across populations. (NCT00071760)
Timeframe: Week 48

InterventionMilliliters/minute/kilogram (mL/min/kg) (Geometric Mean)
45/10 mg/kg BID; n=9, 160/10 mg/kg BID; n=2, 245/7 mg/kg BID; n=2, 1060/7 mg/kg BID; n=0, 8
FPV/RTV BID: 6 Months to <2 Years10.4231.9222.817.8

[back to top]

Number of Participants With Treatment Limiting Toxicities

Treatment limiting toxicities are defined as those that are related to investigational medicinal products and deemed to be unacceptable, leading to restriction of further dose escalation. (NCT00071760)
Timeframe: Day 1 and up to Week 684

InterventionParticipants (Count of Participants)
Cohort 2, Arm A - 4 Weeks to <6 Months0
Cohort 1, Arm A - 6 Months to <2 Years0

[back to top]

Plasma APV Cmax

The maximum concentration at steady state (Cmax) was measured. (NCT00071760)
Timeframe: Week 48

InterventionMicrograms per milliliter (µg/mL) (Least Squares Mean)
45/10 mg/kg BID; n=9, 160/10 mg/kg BID; n=2, 245/7 mg/kg BID; n=2, 10
FPV/RTV BID: 4 Weeks to <6 Months6.2510.448.20

[back to top]

Absolute Values of Cholesterol, Glucose, High Density Lipoprotein (HDL) Cholesterol, Low Density Lipoprotein (LDL) and Triglyceride (TG) at Baseline (Day 1), Weeks 4, 12, 24, 36, and 48

Blood samples of all participants were generally collected under non-fasting conditions (given the age of participants) for the evaluation of cholesterol, serum glucose, HDL cholesterol, LDL cholesterol and triglyceride (TG). Clinical chemistry analyses were carried out using the observed analysis strategy. (NCT00071760)
Timeframe: Baseline (Day 1) and Weeks 4, 12, 24, 36, and 48

InterventionMillimoles per liter (mmol/L) (Median)
Cholesterol, Day 1; n=22;29Cholesterol, Week 4; n=4;2Cholesterol, Week 12; n=0;2Cholesterol, Week 24; n=22;26Cholesterol, Week 36; n=3;2Cholesterol, Week 48; n=18;24Glucose, Day 1; n=26;30Glucose, Week 4; n=23;28Glucose, Week 12; n=22;27Glucose, Week 24; n=22;26Glucose, Week 36; n=20;25Glucose, Week 48; n=18;24HDL, Day 1; n=22;29HDL, Week 4; n=4;2HDL, Week 12; n=0;2HDL, Week 24; n=22;26HDL, Week 36; n=3;2HDL, Week 48; n=18;24LDL, Day 1; n=22;28LDL, Week 4; n=4;2LDL, Week 12; n=0;2LDL, Week 24; n=22;26LDL, Week 36; n=3;2LDL, Week 48; n=18;24TG, Day 1; n=22;29TG, Week 4; n=4;2TG, Week 12; n=0;2TG, Week 24; n=22;26TG, Week 36; n=3;2TG, Week 48; n=18;24
Cohort 1, Arm A - 6 Months to <2 Years2.9103.8853.9504.0003.1504.7504.704.804.704.304.604.700.6800.6900.7750.9600.8051.0501.502.452.332.322.082.801.6501.6451.8101.5501.7001.390

[back to top]

Number of Participants Who Permanently Discontinued the Treatment Due to Adverse Event

An AE is any untoward medical occurrence in a patient or clinical investigation subject administered a pharmaceutical product and which does not necessarily have a causal relationship with this treatment. An AE can therefore be any unfavorable and unintended sign (that could include a clinically significant abnormal laboratory finding), symptom, or disease temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product. (NCT00071760)
Timeframe: Day 1 and up to Week 684

InterventionParticipants (Count of Participants)
Cohort 2, Arm A - 4 Weeks to <6 Months1
Cohort 1, Arm A - 6 Months to <2 Years1

[back to top]

Plasma APV Cmax

The maximum concentration at steady state (Cmax) was measured. (NCT00071760)
Timeframe: Week 48

InterventionMicrograms per milliliter (µg/mL) (Least Squares Mean)
45/10 mg/kg BID; n=9, 160/10 mg/kg BID; n=2, 245/7 mg/kg BID; n=2, 1060/7 mg/kg BID; n=0, 9
FPV/RTV BID: 6 Months to <2 Years21.827.475.8410.4

[back to top]

Number of Participants With Treatment Emergent Adverse Events (AEs)

An AE is any untoward medical occurrence in a patient or clinical investigation subject administered a pharmaceutical product and which does not necessarily have a causal relationship with this treatment. An AE can therefore be any unfavorable and unintended sign (that could include a clinically significant abnormal laboratory finding), symptom, or disease temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product. (NCT00071760)
Timeframe: Day 1 and up to week 684

InterventionParticipants (Count of Participants)
Cohort 2, Arm A - 4 Weeks to <6 Months25
Cohort 1, Arm A - 6 Months to <2 Years29

[back to top]

Absolute Values of Alanine Amino Transferase (ALT) and Aspartate Amino Transferase (AST) at Baseline (Day 1), Weeks 4, 12, 24, 36, and 48

Blood samples of the participants were collected for the evaluation of ALT and AST. Clinical chemistry analyses were carried out using the observed analysis strategy. (NCT00071760)
Timeframe: Baseline (Day 1) and Weeks 4, 12, 24, 36, and 48

,
InterventionInternational units per liter (IU/L) (Median)
ALT, Day 1; n=26;30ALT, Week 4; n=23;28ALT, Week 12; n=22;27ALT, Week 24; n=22;26ALT, Week 36; n=20;25ALT, Week 48; n=18;24AST, Day 1; n=26;30AST, Week 4; n=23;28AST, Week 12; n=22;27AST, Week 24; n=22;26AST, Week 36; n=20;24AST, Week 48; n=18;23
Cohort 1, Arm A - 6 Months to <2 Years20.015.515.015.516.016.044.035.040.036.534.034.0
Cohort 2, Arm A - 4 Weeks to <6 Months22.014.015.016.015.515.043.532.035.034.035.034.5

[back to top]

Plasma Unbound APV Percent Protein Binding (%Cτ)

Participants who are <2 years old may have reduced protein binding; therefore, plasma unbound APV concentrations were measured to determine dosing recommendations at acceptable dosing volumes. APV %Cτ unbound is the percentage of the total APV Cτ that is unbound. (NCT00071760)
Timeframe: Week 48

InterventionPercentage of total APV Cτ unbound (Mean)
45/10 mg/kg BID; n=7, 1560/10 mg/kg BID; n=1, 745/7 mg/kg BID; n=1, 1660/7 mg/kg BID; n=0, 9
FPV/RTV BID: 6 Months to <2 Years6.565.818.239.20

[back to top]

Plasma RTV CL/F Expressed in mL/Min

Apparent clearance of drug from plasma following extravascular administration (CL/F) was calculated as dose in mg/kg units divided by AUC(0-τ). (NCT00071760)
Timeframe: Week 48

,
InterventionmL/min (Geometric Mean)
7 mg/kg BID, n=2, 2210 mg/kg BID, n=11, 2
FPV/RTV BID: 4 Weeks to <6 Months335.272.1
FPV/RTV BID: 6 Months to <2 Years134.157.9

[back to top]

Plasma Unbound APV Percent Protein Binding (%Cτ)

Participants who are <2 years old may have reduced protein binding; therefore, plasma unbound APV concentrations were measured to determine dosing recommendations at acceptable dosing volumes. APV %Cτ unbound is the percentage of the total APV Cτ that is unbound. (NCT00071760)
Timeframe: Week 48

InterventionPercentage of total APV Cτ unbound (Mean)
45/10 mg/kg BID; n=7, 1560/10 mg/kg BID; n=1, 745/7 mg/kg BID; n=1, 16
FPV/RTV BID: 4 Weeks to <6 Months5.795.327.55

[back to top]

Plasma Unbound APV Cτ

"Participants who are <2 years old may have reduced protein binding; therefore, plasma unbound APV concentrations were measured to determine dosing recommendations at acceptable dosing volumes. Unbound or free APV is the fraction of drug that is not bound to protein. Cτ is the plasma concentration at the end of the dosing interval at steady state." (NCT00071760)
Timeframe: Week 48

Interventionµg/mL (Mean)
45/10 mg/kg BID; n=7, 1660/10 mg/kg BID; n=1, 745/7 mg/kg BID; n=1, 1660/7 mg/kg BID; n=0, 12
FPV/RTV BID: 6 Months to <2 Years0.0870.0690.1500.290

[back to top]

Plasma Unbound APV Cτ

"Participants who are <2 years old may have reduced protein binding; therefore, plasma unbound APV concentrations were measured to determine dosing recommendations at acceptable dosing volumes. Unbound or free APV is the fraction of drug that is not bound to protein. Cτ is the plasma concentration at the end of the dosing interval at steady state." (NCT00071760)
Timeframe: Week 48

Interventionµg/mL (Mean)
45/10 mg/kg BID; n=7, 1660/10 mg/kg BID; n=1, 745/7 mg/kg BID; n=1, 16
FPV/RTV BID: 4 Weeks to <6 Months0.0910.0030.027

[back to top]

Plasma RTV Cτ

The plasma concentration at the end of the dosing interval at steady state (Cτ) was measured. (NCT00071760)
Timeframe: Week 48

,
Interventionµg/mL (Geometric Mean)
7 mg/kg BID, n=4, 3310 mg/kg BID, n=15, 19
FPV/RTV BID: 4 Weeks to <6 Months0.07950.1855
FPV/RTV BID: 6 Months to <2 Years0.24680.4200

[back to top]

Plasma RTV Cmax

The maximum concentration at steady state (Cmax) was measured. (NCT00071760)
Timeframe: Week 48

,
Interventionµg/mL (Geometric Mean)
7 mg/kg BID, n=2, 2310 mg/kg BID, n=12, 2
FPV/RTV BID: 4 Weeks to <6 Months0.4042.388
FPV/RTV BID: 6 Months to <2 Years1.5763.823

[back to top]

Plasma RTV CL/F Expressed in mL/Min/kg

Apparent clearance of drug from plasma following extravascular administration (CL/F) was calculated as dose in mg/kg units divided by AUC(0-τ). Normalizing CL/F for bodyweight allows for comparison of CL/F across populations. (NCT00071760)
Timeframe: Week 48

,
InterventionmL/min/kg (Geometric Mean)
7 mg/kg BID, n=2, 2210 mg/kg BID, n=11, 2
FPV/RTV BID: 4 Weeks to <6 Months58.66812.118
FPV/RTV BID: 6 Months to <2 Years14.9608.938

[back to top]

Number of Participants With Plasma Human Immuno Virus Type 1 (HIV-1) Ribonucleic Acid (RNA) Levels < 400 Copies/Milliliter at Each Study Visit

Blood samples of participants were collected to measure plasma HIV-1 RNA concentrations. (NCT00071760)
Timeframe: Baseline (Day 1) and up to Week 684

,
InterventionParticipants (Count of Participants)
Baseline (Day 1)Week 4Week 12Week 24Week 36Week 48Week 60Week 72Week 84Week 96Week 108Week 120Week 132Week 144Week 156Week 168Week 180Week 192Week 204Week 216Week 228Week 240Week 252Week 264Week 276Week 288Week 300Week 312Week 324Week 336Week 348Week 360Week 372Week 384Week 396Week 408Week 420Week 432Week 444Week 456Week 468Week 480Week 492Week 504Week 516Week 528Week 540Week 552Week 564Week 576Week 588Week 600Week 612Week 624Week 636Week 648Week 660Week 672Week 684
Cohort 1, Arm A - 6 Months to <2 Years06182020221214151414111181099899888888888767777777777777765555533322221
Cohort 2, Arm A - 4 Weeks to <6 Months1513181915151414151514141414121010101012121212121212101111101011101111810999979999966655511100

[back to top]

Plasma Ritonavir (RTV) AUC (0-τ)

Plasma samples were assayed for RTV concentrations using a validated assay. The GSK Department of Clinical Pharmacology Modeling and Simulation conducted PK analysis of the plasma RTV concentration-time data using a model-independent approach. As a measure of total drug exposure, the area under the plasma-concentration-versus-time curve over the dosing interval at steady-state (AUC[0-τ]), where τ is the length of the dosing interval, was calculated by the linear up/log down trapezoidal method. (NCT00071760)
Timeframe: Week 48

,
Interventionhr*µg/mL (Geometric Mean)
7 mg/kg BID; n=2, 2210 mg/kg BID; n=11, 2
FPV/RTV BID: 4 Weeks to <6 Months1.92112.952
FPV/RTV BID: 6 Months to <2 Years7.36318.750

[back to top]

Plasma APV Cτ

The plasma concentration at the end of the dosing interval at steady state (Cτ) was measured. (NCT00071760)
Timeframe: Week 48

InterventionMicrograms per milliliter (µg/mL) (Geometric Mean)
45/10 mg/kg BID; n=11, 1560/10 mg/kg BID; n=3, 545/7 mg/kg BID; n=3, 2960/7 mg/kg BID; n=0, 12
FPV/RTV BID: 6 Months to <2 Years1.922.582.172.81

[back to top]

Number of Participants With the Indicated Treatment-emergent (TE) Grade 3/4 Adverse Events (AE)

An AE is any untoward medical occurrence in a patient or clinical investigation subject, temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product. An AE is considered TE if it has an onset date on or after the date of the first dose of study drug, and on or before the date of the final dose of study drug. As per the Division of AIDS Table for Grading the Severity of Adult and Pediatric AEs, Grade 3=severe; Grade 4=potentially life-threatening. (NCT00071760)
Timeframe: Baseline (Day 1) until Week 48

Interventionparticipants (Number)
Cohort 2, Arm A - 4 Weeks to <6 Months11
Cohort 1, Arm A - 6 Months to <2 Years10

[back to top]

Number of Participants Who Permanently Discontinued the Treatment Due to an AE

An AE is any untoward medical occurrence in a patient or clinical investigation subject, temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product. (NCT00071760)
Timeframe: Baseline (Day 1) until Week 48

Interventionparticipants (Number)
Cohort 2, Arm A - 4 Weeks to <6 Months1
Cohort 1, Arm A - 6 Months to <2 Years2

[back to top]

Plasma APV Cτ

The plasma concentration at the end of the dosing interval at steady state (Cτ) was measured. (NCT00071760)
Timeframe: Week 48

InterventionMicrograms per milliliter (µg/mL) (Geometric Mean)
45/10 mg/kg BID; n=11, 1560/10 mg/kg BID; n=3, 545/7 mg/kg BID; n=3, 29
FPV/RTV BID: 4 Weeks to <6 Months0.860.600.44

[back to top]

All Partner HIV Infection Rates in Early-ART and Delayed-ART Arms

All Incident HIV infections occurring in the partners (HIV-negative at enrollment) of randomized HIV-infected index (HIV-positive at enrollment) cases are assessed, by arm. (NCT00074581)
Timeframe: Throughout study

Interventionevent rate per 100 person-yr (Number)
Early-ART0.44
Delayed-ART1.41

[back to top]

Linked Partner HIV Infection Rates in Early-ART and Delayed-ART Arms

incident HIV infections occurring in the partners (HIV-negative at enrollment) of randomized HIV-infected index (HIV-positive at enrollment) cases are assessed, by arm. Only acquisition from the index partner were included in the primary analysis, therefore, each endpoint was required to be confirmed (by genotyping) such that the viral envelop sequence in the index case matched that of the partner. (NCT00074581)
Timeframe: Throughout study

Interventionevent rate per 100 person-yr (Number)
Early-ART0.07
Delayed-ART1.03

[back to top]

Levels of Proviral DNA in Peripheral Blood Mononuclear Cells (PBMC) (log10)

(NCT00084149)
Timeframe: At 48 weeks after the start of treatment

Interventionlog10(copies/mL) (Median)
Cyclosporine1.88
No Cyclosporine1.92

[back to top] [back to top]

CD4 T Cell Levels

(NCT00084149)
Timeframe: At Week 48

Interventioncells/mm^3 (Median)
Cyclosporine301
No Cyclosporine287

[back to top]

HIV-1 Viral Load Levels

(NCT00084149)
Timeframe: At Week 48

Interventionlog10(copies/mL) (Mean)
Cyclosporine1.70
No Cyclosporine1.70

[back to top]

Number of Patients With Viral Load Less Than 50 Copies/ml

(NCT00084149)
Timeframe: Week 48

InterventionParticipants (Count of Participants)
Cyclosporine27
No Cyclosporine13

[back to top]

Proviral DNA (log10)

(NCT00084149)
Timeframe: At Week 12

Interventionlog10(copies/mL) (Median)
Cyclosporine2.22
No Cyclosporine2.13

[back to top]

Proviral DNA Levels (log10)

(NCT00084149)
Timeframe: At Week 24

Interventionlog10(copies/mL) (Median)
Cyclosporine2.12
No Cyclosporine1.96

[back to top]

Number of Participants Who Experienced Virologic Failure or Died.

Virologic failure (VF) is defined as a plasma HIV-1 RNA level that is 1 log10 below baseline 12 weeks after treatment is initiated or as a plasma HIV-1 RNA level that is >=400 copies/mL at or after 24 weeks of treatment, regardless of whether randomized treatment was being taken at the time of VF. (NCT00089505)
Timeframe: Through database cutoff for DSMB review (by October 6, 2008) for NVP/NVP and NVP/LPV_r. Throughout study for NoNVP/NVP and NoNVP/LPV_r.

Interventionparticipants (Number)
NVP/NVP32
NVP/LPV_r10
NoNVP/NVP42
NoNVP/LPV_r50

[back to top] [back to top] [back to top]

Percent of Participants Who Reported to Never Missed Any of the Study Drug Regimen in the Past Month

Self-reported adherence at week 48 and 96 while participants remained on randomized regimen. Adherence interviews for each antiretroviral drug drug the participant is taking was performed by site personnel every 24 weeks. For NVP/NVP and NVP/LPV_r arms, data through DSMB review cutoff (October 6, 2008) were used to report the outcome. For NoNVP/NVP and NoNVP/LPV_r arms, since the follow-up continued as planned, data through overall study were used. (NCT00089505)
Timeframe: Through database cutoff for DSMB review (by October 6, 2008) for NVP/NVP and NVP/LPV_r arms. Throughout study for NoNVP/NVP and NoNVP/LPV_r arms.

,,,
Interventionpercent of participants (Number)
week 48 percent of full adherence in past monthweek 96 percent of full adherence in past month
NoNVP/LPV_r8687
NoNVP/NVP9093
NVP/LPV_r8895
NVP/NVP8994

[back to top]

Percent of Participants Who Experienced Virologic Failure or Died

Results report cumulative percent of participants reaching virologic failure (VF) or death by week 48 and week 96 calculated using the Kaplan-Meier method. VF is defined as a plasma HIV-1 RNA level that is 1 log10 below baseline 12 weeks after treatment is initiated or as a plasma HIV-1 RNA level that is >=400 copies/mL at or after 24 weeks of treatment, regardless of whether randomized treatment was being taken at the time of VF. (NCT00089505)
Timeframe: Through database cutoff for DSMB review (by October 6, 2008) for NVP/NVP and NVP/LPV_r arms. Throughout study for NoNVP/NVP and NoNVP/LPV_r arms.

,,,
InterventionPercent of participants (Number)
week 48 percent of virologic failure or deathweek 96 percent of virologic failure or death
NoNVP/LPV_r1420
NoNVP/NVP1417
NVP/LPV_r412
NVP/NVP2331

[back to top]

CD4 Count Change From Randomization

Change was calculated as the CD4 count at Week 48 (or at Week 96) minus the baseline CD4 count (last CD4 before/on treatment start date). For NVP/NVP and NVP/LPV_r arms, data through DSMB review cutoff (October 6, 2008) were used to report the outcome. For NoNVP/NVP and NoNVP/LPV_r arms, since the follow-up continued as planned, data through overall study were used. (NCT00089505)
Timeframe: Through database cutoff for DSMB review (by October 6, 2008) for NVP/NVP and NVP/LPV_r. Throughout study for NoNVP/NVP and NoNVP/LPV_r. Week 48 and 96.

,,,
Interventioncells/mm^3 (Median)
Week 48 CD4 count change from randomizationWeek 96 CD4 count change from randomization
NoNVP/LPV_r172256
NoNVP/NVP172223
NVP/LPV_r201278
NVP/NVP191291

[back to top]

Number of Participants Who Received NVP-containing Regimens at Randomization and Experienced NVP-associated Rash or Grade 2+ Liver Lab Abnormality

Any grade of rash or grade 2+ liver lab abnormality events that were claimed to be NVP associated (definitely, probably, or possibly) by site investigators were evaluated. Grade 2+ liver lab abnormality is defined as aspartate aminotransferase (AST)>=2.6 x ULN or alanine aminotransferase (ALT)>=2.6 x ULN. (NCT00089505)
Timeframe: Through database cutoff for DSMB review (by October 6, 2008) for NVP/NVP arm. Throughout study for NoNVP/NVP arm.

Interventionparticipants (Number)
NVP/NVP20
NoNVP/NVP51

[back to top]

Time From Randomization to Virologic Failure or Death for Participants Without SD NVP Exposure Prior to Study Entry

5th and 10th Percentiles in weeks from randomization to virologic failure (VF) or death. VF is defined as a plasma HIV-1 RNA level that is 1 log10 below baseline 12 weeks after treatment is initiated or as a plasma HIV-1 RNA level that is >=400 copies/mL at or after 24 weeks of treatment, regardless of whether randomized treatment was being taken at the time of VF. (NCT00089505)
Timeframe: Throughout study with median follow-up 72 weeks and range from 0 to 180 weeks.

,
Interventionweeks (Number)
5th percentile10th percentile25th percentile
NoNVP/LPV_r1236132
NoNVP/NVP2436NA

[back to top]

Time From Randomization to Virologic Failure or Death for Participants Who Had SD NVP Exposure Prior to Study Entry

5th and 10th Percentiles in weeks from randomization to virologic failure (VF) or death. VF is defined as a plasma HIV-1 RNA level that is 1 log10 below baseline 12 weeks after treatment is initiated or as a plasma HIV-1 RNA level that is >=400 copies/mL at or after 24 weeks of treatment, regardless of whether randomized treatment was being taken at the time of VF. (NCT00089505)
Timeframe: Through database cutoff for DSMB review (by October 6, 2008) with median follow-up 72 weeks and range from 0 to 144 weeks.

,
Interventionweeks (Number)
5th percentile10th percentile25th percentile
NVP/LPV_r6084NA
NVP/NVP121260

[back to top]

Plasma APV t1/2

The apparent terminal phase half-life (t1/2) is calculated as loge2/λz. The apparent terminal phase rate constant (λz) is the slope of the terminal portion of the logarithmically transformed concentration-time data as estimated by linear regression. (NCT00089583)
Timeframe: Week 48

,
Interventionhours (Geometric Mean)
2 to <6yrs, 30 mg/kg BID; n=9, 02 to <6yrs, 40 mg/kg BID; n=5, 02 to <6yrs, 23/3 mg/kg BID; n=0, 146 to <12 yrs, 15/3 mg/kg BID; n=0, 76 to <12 yrs, 18/3 mg/kg BID; n=0, 106 to <12 yrs, 700/100 mg BID; n=0, 212 to 18 yrs, 15/3 mg/kg BID; n=0, 412 to 18 yrs, 18/3 mg BID; n=0, 412 to 18 yrs, 700/100 mg BID; n=0, 11
FPV Treatment Group3.033.18NANANANANANANA
FPV/RTV Treatment GroupNANA5.2110.58.417.436.128.767.64

[back to top]

Number of Participants (Par.) With Plasma HIV-1 Ribonucleic Acid (RNA) <400 Copies Per Milliliter at Baseline and Weeks 2,12, 24, and 48 (MSD=F)

Blood samples of participants were collected to measure plasma HIV-1 RNA concentrations. PI-exp = PI-experienced. Viral load, measured in RNA copies per milliliter of plasma, is an efficacy measure for antiretroviral drugs. In the Missing, Switch, or Discontinuation = Failure (MSD=F) analysis, participants who had missing data at or had discontinued the study prior to a certain time point or had changed their background antiretroviral regimen are classified as non-responders. (NCT00089583)
Timeframe: Baseline and Weeks 2, 12, 24, and 48

,
Interventionparticipants (Number)
PI-naïve, Baseline; n=20, 49PI-exp, Baseline; n=0, 40PI-naïve, Week 2; n=20, 49PI-exp, Week 2; n=0, 40PI-naïve, Week 12; n=20, 49PI-exp, Week 12; n=0, 40PI-naïve, Week 24; n=20, 49PI-exp, Week 24; n=0, 40PI-naïve, Week 48; n=20, 49PI-exp, Week 48; n=0, 40
FPV Treatment Group0NA3NA13NA13NA12NA
FPV/RTV Treatment Group0095351935223619

[back to top]

Change From Baseline in the Percentage of Total Lymphocytes (TLs) That Are CD4+ Cells at Weeks 2, 12, 24, and 48

Blood samples of participants were collected for the measurement of the percentage of total lymphocytes that are CD4+ cells. Observed analysis was used for the summary of proportion endpoints using viral load data. Change from Baseline in percentage was calculated as the value at Weeks 2, 12, 24, and 48 minus the value at Baseline. (NCT00089583)
Timeframe: Baseline and Week 2, 12, 24, 48

,
InterventionPercentage of TLs that are CD4+ cells (Median)
PI-naïve, Week 2; n=13, 41PI-naïve, Week 12; n=19, 46PI-naïve, Week 24; n=18, 44PI-naïve, Week 48; n=17, 42PI-exp, Week 2; n=0, 32PI-exp, Week 12; n=0, 31PI-exp, Week 24; n=0, 34PI-exp, Week 48; n=0, 29
FPV Treatment Group3678NANANANA
FPV/RTV Treatment Group158102356

[back to top]

Change From Baseline in Triglycerides, Total Cholesterol, Low-density Lipoprotein (LDL) Cholesterol, High-density Lipoprotein (HDL) Cholesterol, and Serum Glucose at Week 48

Blood samples of all participants were collected under fasting conditions for the evaluation of triglycerides, total cholesterol, HDL cholesterol, LDL cholesterol, and serum glucose. Clinical chemistry analyses were carried out using the observed analysis strategy. Change from Baseline in triglycerides, total cholesterol, HDL cholesterol, LDL cholesterol, and serum glucose was calculated as the value at Week 48 minus the value at Baseline. (NCT00089583)
Timeframe: Baseline (Day 1) and Week 48

,
InterventionMillimoles per liter (mmol/L) (Median)
Triglycerides; n=17, 65Total cholesterol; n=17, 65HDL cholesterol; n=17, 65LDL cholesterol; n=17, 64Glucose; n=18, 69
FPV Treatment Group0.11.10.40.60.0
FPV/RTV Treatment Group0.20.90.30.50.1

[back to top]

Cluster of Differentiation Antigen 4 (CD4+) Cell Count at Baseline and at Weeks 2, 12, 24, and 48

Blood samples of participants were collected for the measurement of CD4+ cell count. Observed analysis was used for the summary of proportion endpoints using viral load data. CD4+ cells are white blood cells that are important in fighting infection. HIV infects CD4+ cells, replicates in them, and destroys them. CD4+ cell count provides a measure of the status of the immune system and to what extent it is affected by HIV. (NCT00089583)
Timeframe: Baseline and Weeks 2, 12, 24, and 48

,
InterventionCells per cubic millimeter (cells/cu mm) (Median)
PI-naïve, Baseline; n= 19, 49PI-naïve, Week 2; n= 13, 41PI-naïve, Week 12; n= 19, 46PI-naïve, Week 24; n= 18, 44PI-naïve, Week 48; n= 18, 42PI-exp, Baseline; n= 0, 40PI-exp, Week 2; n= 0, 32PI-exp, Week 12; n= 0, 31PI-exp, Week 24; n= 0, 34PI-exp, Week 48; n= 0, 29
FPV Treatment Group810820104012601080NANANANANA
FPV/RTV Treatment Group370450581609670440605720620540

[back to top]

Median Change From Plasma HIV-1 RNA (log10 Copies/mL) at Weeks 2, 12, 24, and 48 (Observed Analysis)

Blood samples of participants were collected to assess the decrease in the number of HIV-1 RNA. Change from Baseline at Weeks 2, 12, 24, and 48 was calculated as value at Week 2, 12, 24, and 48 minus the value at Baseline. (NCT00089583)
Timeframe: Baseline and Weeks 2, 12, 24, and 48

,
Interventionlog10/copies (Median)
PI-naïve, Week 2; n=14, 39PI-exp, Week 2; n=0, 32PI-naïve, Week 12; n=19, 46PI-exp, Week 12; n=0, 33PI-naïve, Week 24; n=18, 44PI-exp, Week 24; n=0, 35PI-naïve, Week 48; n=18, 44PI-exp, Week 48; n=0, 33
FPV Treatment Group-1.91NA-3.04NA-3.16NA-3.02NA
FPV/RTV Treatment Group-1.84-1.58-2.77-2.23-2.87-2.28-2.83-2.14

[back to top]

Number of Confirmed Virologic Failure Participants (Par.) With Treatment-emergent Reductions in Drug Susceptibility (DS)

A blood sample was drawn for par. failing to respond to therapy, and changes in DS for HIV isolated from the par. for each drug used in the study were assessed. The changes in DS detected by phenotypic assay in virus from the sample collected at the time of failure was compared with DS in the virus from the blood sample at baseline. Par. are grouped by study arm and prior therapy experience. DS is the state of HIV being susceptible to the antiretroviral agent (the virus can be inhibited by the drug). Reduced DS (i.e., HIV is resistant to the antiretroviral agent) can lead to treatment failure. (NCT00089583)
Timeframe: Baseline through 48 Weeks

,,,
Interventionparticipants (Number)
Any NRTIAbacavirDidanosineEmtricitabineLamivudineAny NNRTIAny PIUnboosted FosamprenavirRitonavir- boosted FosamprenavirRitonavir
ART-Naïve, FPV Treatment Group31333022NA2
ART-Naïve, FPV/RTV Treatment Group0000000NA00
PI Naïve, ART-experienced, FPV/RTV Treatment Group0000001NA10
PI-experienced, ART-experienced FPV/RTV Treatment Group2011101NA10

[back to top]

Number of Participants With Treatment-emergent (TE) Grade 3/4 Clinical Chemistry Laboratory Abnormalities

"A toxicity was considered TE if it was > than the Baseline grade, and if it was observed on/after the date of the first dose of study drug (SD), and on/before the date of the last dose of SD. Leucopenia is the decrease in the number of leucocytes (white blood cells [WBCs]); neutropenia is the decrease in the number of neutrophils (type of WBCs). Per the Division of AIDS Table for Grading the Severity of Adult and Pediatric AEs: Grade 3 is severe; Grade 4 is potentially life-threatening. ULN, upper limit of normal; LDL, low-density lipoprotein; PC, platelet count." (NCT00089583)
Timeframe: Baseline (Day 1) until Week 48

,
Interventionparticipants (Number)
ALT increased (inc.) (>5.0x ULN); n=20, 87AST inc. (>5.0x ULN); n=20, 87Cholesterol (Chol.) inc. (>7.77 mmol/L); n=16, 43Hyperglycemia (>13.88 mmol/L); n=16, 58Hypoglycemia (<2.22 mmol/L); n=16, 58LDL Chol. inc. (>=4.91 mmol/L); n=16, 43Triglycerides inc. (>8.48 mmol/L); n=16, 43Lipase inc. (>3.0x ULN); n=19, 85Leucopenia (<1.500 x 10^9/L); n=20, 84Neutropenia (<0.750 x 10^9/L); n=20, 84Hemoglobin > anemia (<1.16 mmol/L); n=20, 85PC > thrombocytopenia (<50.000 x 10^9/L); n=20, 85
FPV Treatment Group220000000800
FPV/RTV Treatment Group222004000701

[back to top]

Number of Confirmed Virologic Failure Participants (Par.) With Treatment-emergent HIV Genotypic Resistance in Reverse Transcriptase and Protease

A blood sample was drawn for par. failing to respond to therapy, and the mutations present in the virus were identified. For each par., the mutations found at the time of failure were compared with any mutations found in the blood sample at baseline. New International AIDS Society-USA defined resistance mutations that developed at the time of failure were tabulated by drug class. VF, virologic failure; NRTI, nucleoside reverse transcriptase inhibitor; NNRTI, non-nucleoside reverse transcriptase inhibitor; PI, protease inhibitor. Par. are grouped by study arm and prior therapy experience. (NCT00089583)
Timeframe: Week 48

,,,
Interventionparticipants (Number)
Any HIV NRTI MutationHIV NRTI mutation M184VAny HIV NNRTI MutationHIV NNRTI Mutation V179D/EAny HIV Major PI MutationsHIV Major PI Mutation M46M/IHIV Major PI Mutation M46M/LHIV Major PI Mutation I50I/VHIV Major PI Mutation I54I/LHIV Major PI Mutation I54I/MHIV Major PI Mutation I54I/M/VHIV Major PI Mutation Q58Q/EHIV Major PI Mutation V82A/VHIV Major PI Mutation V82F/IHIV Major PI Mutation I84I/VAny Minor HIV PI MutationsMinor HIV PI Mutation L10L/FMinor HIV PI Mutation K20K/RMinor HIV PI Mutation L33L/FMinor HIV PI Mutation K43K/TMinor HIV PI Mutation F53F/LMinor HIV PI Mutation F53LMinor HIV PI Mutation I62I/VMinor HIV PI Mutation A71I/VMinor HIV PI Mutation I85I/V
ART-Naïve, FPV Treatment Group3300201110011002011011000
ART-Naïve, FPV/RTV Treatment Group0000000000000000000000000
PI Experienced, ART Experienced, FPV/RTV Treatment Group1111100101000101000100001
PI Naïve, ART Experienced, FPV/RTV Treatment Group0000110100100011101000111

[back to top]

Median Plasma HIV-1 RNA (log10 Copies/mL) at Baseline and Weeks 2, 12, 24, and 48 (Observed Analysis)

Blood samples of participants were collected to assess the decrease in the number of HIV-1 RNA. (NCT00089583)
Timeframe: Baseline and Weeks 2, 12, 24, and 48

,
Interventionlog10 copies/mL (Median)
PI-naïve, Baseline; n=20, 49PI-exp, Baseline; n=0, 39PI-naïve, Week 2; n=14, 39PI-exp, Week 2; n=0, 33PI-naïve, Week 12; n=19, 46PI-exp, Week 12; n=0, 34PI-naïve, Week 24; n=18, 44PI-exp, Week 24; n=0, 35PI-naïve, Week 48; n=18, 44PI-exp, Week 48; n=0, 33
FPV Treatment Group5.13NA3.27NA2.03NA1.85NA1.85NA
FPV/RTV Treatment Group4.724.533.063.041.942.201.691.801.691.69

[back to top]

Change From Baseline in Aspartate Aminotransferase (AST) and Alanine Aminotransferase (ALT) at Week 48

Blood samples of the participants were collected for the evaluation of AST and ALT. Clinical chemistry analyses were carried out using the observed analysis strategy. Change from Baseline in AST and ALT was calculated as the value at Week 48 minus the value at Baseline. (NCT00089583)
Timeframe: Baseline (Day 1) and Week 48

,
InterventionInternational units per liter (IU/L) (Median)
ALTAST
FPV Treatment Group-3-6
FPV/RTV Treatment Group-7-9

[back to top]

Change From Baseline in CD4+ Cell Count at Weeks 2, 12, 24, and 48

Blood samples of participants were collected for the measurement of CD4+ cell count. Observed analysis was used for the summary of proportion endpoints using viral load data. Change from Baseline was calculated as the value at Weeks 2, 12, 24, and 48 minus the value at Baseline. (NCT00089583)
Timeframe: Baseline and Weeks 2, 12, 24, and 48

,
Interventioncells/cu mm (Median)
PI-naïve, Week 2; n=13, 41PI-naïve, Week 12; n=19, 46PI-naïve, Week 24; n=18, 44PI-naïve, Week 48; n=17, 42PI-exp, Week 2; n=0, 32PI-exp, Week 12; n=0, 31PI-exp, Week 24; n=0, 34PI-exp, Week 48; n=0, 29
FPV Treatment Group20170350340NANANANA
FPV/RTV Treatment Group6018018421790200150180

[back to top]

Number of Participants Who Permanently Discontinued the Treatment Due to Any Adverse Event (AE)

An AE is any untoward medical occurrence in a patient or clinical investigation subject, temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product. (NCT00089583)
Timeframe: Week 48

Interventionparticipants (Number)
FPV Treatment Group0
FPV/RTV Treatment Group4

[back to top]

Change From Baseline in Serum Lipase at Week 48

Blood samples of all participants were collected for the evaluation of serum lipase. Clinical chemistry analyses were carried out using the observed analysis strategy. Change from Baseline in serum lipase was calculated as the value at Week 48 minus the value at Baseline. (NCT00089583)
Timeframe: Baseline (Day 1) and Week 48

InterventionUnits per liter (U/L) (Median)
FPV Treatment Group-2.0
FPV/RTV Treatment Group-1.0

[back to top]

Plasma RTV CL/F Following Dosing Expressed in mg/kg

Apparent clearance of drug from plasma following extravascular administration (CL/F) was calculated using the formulation: RTV Dose in mg/kg units divided by AUC(0-τ). Normalizing CL/F for bodyweight allows for comparison of CL/F across populations. (NCT00089583)
Timeframe: Week 48

InterventionmL/min/kg (Geometric Mean)
2 to <6yrs, 3 mg/kg BID; n=0, 106 to <12 yrs, 3 mg/kg BID; n=0, 126 to <12 yrs, 100 mg/kg BID; n=0, 1212 to 18 yrs, 3 mg/kg BID; n=0, 112 to 18 yrs, 100 mg BID; n=0, 15
FPV/RTV Treatment Group12.96.815.948.615.59

[back to top]

Plasma RTV Tmax

The time to reach the maximum concentration (Cmax) at steady state is defined as (tmax). (NCT00089583)
Timeframe: Week 48

Interventionhours (Median)
2 to <6 yrs, 3 mg/kg BID; n=0, 106 to <12 yrs, 3 mg/kg BID; n=0, 146 to <12 yrs, 100 mg BID; n=0, 612 to 18 yrs, 3 mg/kg BID; n=0, 312 to 18 yrs, 100 mg BID; n=0, 16
FPV/RTV Treatment Group3.924.004.015.923.96

[back to top]

Number of Confirmed Virologic Failure Participants (Par.) Since the Week 48 Analysis With Treatment-emergent HIV Genotypic Resistance in Reverse Transcriptase and Protease

A blood sample was drawn for par. remaining in the study after Week 48 and failing to respond to therapy, and the mutations present in the virus were identified. For each par., the mutations found at the time of failure were compared with any mutations found in the blood sample at baseline. New International AIDS Society-USA defined resistance mutations that developed at the time of failure were tabulated by drug class. VF, virologic failure; NRTI, nucleoside reverse transcriptase inhibitor; NNRTI, non-nucleoside reverse transcriptase inhibitor; PI, protease inhibitor. Par. are grouped by study arm and prior therapy experience. (NCT00089583)
Timeframe: After Week 48 through Week 240

,,
InterventionParticipants (Number)
Any HIV NRTI MutationHIV NRTI mutation M41LHIV NRTI mutation M184IHIV NRTI mutation M184VHIV NRTI mutation T215S/YAny Major HIV NNRTI MutationHIV NNRTI Mutation K103NAny Minor HIV NNRTI MutationHIV NNRTI Mutation V179D/EAny HIV Major PI MutationsHIV Major PI Mutation V32IHIV Major PI Mutation M46LHIV Major PI Mutation I47IVHIV Major PI Mutation T74PHIV Major PI Mutation I84I/VAny Minor HIV PI MutationsMinor HIV PI Mutation L10FMinor HIV PI Mutation L33FMinor HIV PI Mutation I62I/VMinor HIV PI Mutation I85V
ART-Naïve, FPV Treatment Group10010000010101011001
ART-Naïve, FPV/RTV Treatment Group10100000000000000000
PI Experienced, ART Experienced, FPV/RTV Treatment Group21021110011010020110

[back to top]

Plasma RTV t1/2

alf-life (t1/2) is calculated as loge2/λz. The apparent terminal phase rate constant (λz) is the slope of the terminal portion of the logarithmically transformed concentration-time data as estimated by linear regression. (NCT00089583)
Timeframe: Week 48

Interventionhours (Geometric Mean)
2 to <6 yrs, 3 mg/kg BID; n=0, 106 to <12 yrs, 3 mg/kg BID; n=0, 116 to <12 yrs, 100 mg BID; n=0, 512 to 18 yrs, 3 mg/kg BID; n=0, 112 to 18 yrs, 100 mg BID; n=0, 14
FPV/RTV Treatment Group3.433.393.972.843.64

[back to top]

Plasma APV Tmax

The time to reach the maximum concentration (Cmax) at steady state is defined as tmax. (NCT00089583)
Timeframe: Week 48

,
Interventionhours (Median)
2 to <6yrs, 30 mg/kg BID; n=9, 02 to <6yrs, 40 mg/kg BID; n=7, 02 to <6yrs, 23/3 mg/kg BID; n=0, 146 to <12 yrs, 15/3 mg/kg BID; n=0, 106 to <12 yrs, 18/3 mg/kg BID; n=0, 126 to <12 yrs, 700/100 mg BID; n=0, 312 to 18 yrs, 15/3 mg/kg BID; n=0, 412 to 18 yrs, 18/3 mg BID; n=0, 312 to 18 yrs, 700/100 mg BID; n=0, 13
FPV Treatment Group1.171.00NANANANANANANA
FPV/RTV Treatment GroupNANA1.252.001.963.921.001.502.00

[back to top]

Plasma RTV Cτ

The plasma concentration at the end of the dosing interval at steady-state (Cτ) was measured. (NCT00089583)
Timeframe: Week 48

Interventionµg/mL (Geometric Mean)
2 to <6 yrs, 3 mg/kg BID; n=0, 166 to <12 yrs, 3 mg/kg BID; n=0, 246 to <12 yrs, 100 mg BID; n=0, 1012 to 18 yrs, 3 mg/kg BID; n=0, 612 to 18 yrs, 100 mg BID; n=0, 41
FPV/RTV Treatment Group0.2240.2970.2280.2630.220

[back to top]

Plasma RTV Cmax

The maximum concentration at steady state (Cmax) was measured. (NCT00089583)
Timeframe: Week 48

Interventionµg/mL (Geometric Mean)
2 to <6 yrs, 3 mg/kg BID; n=0, 106 to <12 yrs, 3 mg/kg BID; n=0, 146 to <12.yrs, 100 mg BID; n=0, 612 to 18 yrs, 3 mg/kg BID; n=0, 312 to 18 yrs, 100 mg BID; n=0, 16
FPV/RTV Treatment Group0.6331.1000.9800.7501.06

[back to top]

Plasma RTV CL/F Following Dosing Expressed in mg

Apparent clearance of drug from plasma following extravascular administration (CL/F) was calculated as dose/AUC(0-τ). (NCT00089583)
Timeframe: Week 48

InterventionmL/min (Geometric Mean)
2 to <6yrs, 3 mg/kg BID; n=0, 106 to <12 yrs, 3 mg/kg BID; n=0, 126 to <12 yrs, 100 mg/kg BID; n=0, 612 to 18 yrs, 3 mg/kg BID; n=0, 112 to 18 yrs, 100 mg BID; n=0, 15
FPV/RTV Treatment Group195190258279272

[back to top]

Plasma Ritonavir (RTV) AUC (0-τ)

Plasma samples were assayed for RTV concentrations using a validated assay. The GlaxoSmithKline (GSK) Department of Clinical Pharmacology Modeling and Simulation conducted pharmacokinetic (PK) analysis of the plasma RTV concentration-time data using a model-independent approach. As a measure of total drug exposure, the area under the plasma-concentration-versus-time curve over the dosing interval at steady-state (AUC[0-τ]), where τ is the length of the dosing interval, was calculated by the linear up/log down trapezoidal method. (NCT00089583)
Timeframe: Week 48

Interventionhr*µg/mL (Geometric Mean)
2 to <6 yrs, 3 mg/kg BID; n=0, 106 to <12 yrs, 3 mg/kg BID; n=0, 126 to <12 yrs, 100 mg BID; n=0, 612 to 18 yrs, 3 mg/kg BID; n=0, 112 to 18 yrs, 100 mg BID; n=0, 15
FPV/RTV Treatment Group3.987.136.465.746.13

[back to top]

Plasma APV CL/F Following Dosing Expressed in mg

Apparent clearance of drug from plasma following extravascular administration (CL/F) was calculated as dose/AUC(0-τ). For FPV, doses were expressed in APV molar equivalents (50 mg of FPV = 43.2 mg of APV). (NCT00089583)
Timeframe: Week 48

,
InterventionMilliliters per minute (mL/min) (Geometric Mean)
2 to <6yrs, 30 mg/kg BID; n=9, 02 to <6yrs, 40 mg/kg BID; n=7, 02 to <6yrs, 23/3 mg/kg BID; n=0, 146 to <12 yrs, 15/3 mg/kg BID; n=0, 96 to <12 yrs, 18/3 mg/kg BID; n=0, 126 to <12 yrs, 700/100 mg BID; n=0, 312 to 18 yrs, 15/3 mg/kg BID; n=0, 412 to 18 yrs, 18/3 mg BID; n=0, 312 to 18 yrs, 700/100 mg BID; n=0, 13
FPV Treatment Group269330NANANANANANANA
FPV/RTV Treatment GroupNANA91195149266392198284

[back to top]

Number of Confirmed Virologic Failure Participants (Par.) Since the Week 48 Analysis With Treatment-emergent Reductions in Drug Susceptibility (DS)

A blood sample was drawn for par. remaining in the study after Week 48 and failing to respond to therapy, and changes in DS for HIV isolated from the par. for each drug used in the study were assessed. The changes in DS detected by phenotypic assay in virus from the sample collected at the time of failure was compared with DS in the virus from the blood sample at baseline. Par. are grouped by study arm and prior therapy experience. DS is the state of HIV being susceptible to the antiretroviral agent (the virus can be inhibited by the drug). Reduced DS (i.e., HIV is resistant to the antiretroviral agent) can lead to treatment failure. (NCT00089583)
Timeframe: Week 60 through Week 240

,,
InterventionParticipants (Number)
Any NRTIAbacavirDidanosineEmtricitabineLamivudineZidovudineAny NNRTIDelaviridineEfavirenzNevirapineAny PIUnboosted FosamprenavirRitonavir- boosted FosamprenavirNelfinavirTipranavir
ART-Naïve, FPV Treatment Group100110000011NA00
ART-Naïve, FPV/RTV Treatment Group10111000001NA010
PI-experienced, ART-experienced FPV/RTV Treatment Group31332111113NA101

[back to top]

Plasma APV Cτ

The plasma concentration at the end of the dosing interval at steady-state (Cτ) was measured. (NCT00089583)
Timeframe: Week 48

,
InterventionMicrograms per milliliter (µg/mL) (Geometric Mean)
2 to <6 yrs, 30 mg/kg BID; n=19, 02 to <6 yrs, 40 mg/kg BID; n=10, 02 to <6 yrs, 23/3 mg/kg BID; n=0, 166 to <12 yrs, 15/3 mg/kg BID; n=0, 136 to <12 yrs, 18/3 mg/kg BID; n=0, 236 to <12 yrs, 700/100 mg BID; n=0, 712 to 18 yrs, 15/3 mg/kg BID; n=0, 612 to 18 yrs, 18/3 mg/kg BID; n=0, 1012 to 18 yrs, 700/100 mg BID; n=0, 40
FPV Treatment Group0.550.70NANANANANANANA
FPV/RTV Treatment GroupNANA3.392.242.421.811.451.802.01

[back to top]

Plasma APV Cmax

The maximum concentration at steady state (Cmax) was measured. (NCT00089583)
Timeframe: Week 48

,
InterventionMicrograms per milliliter (µg/mL) (Geometric Mean)
2 to <6 yrs, 30 mg/kg BID; n=9, 02 to <6 yrs, 40 mg/kg BID; n=7, 02 to <6 yrs, 23/3 mg/kg BID; n=0, 146 to <12 yrs, 15/3 mg/kg BID; n=0, 106 to <12.yrs, 18/3 mg/kg BID; n=0, 126 to <12 yrs, 700/100 mg BID; n=0, 312 to 18 yrs, 15/3 mg/kg BID; n=0, 412 to 18 yrs, 18/3 mg/kg BID; n=0, 412 to 18 yrs, 700/100 mg BID; n=0, 13
FPV Treatment Group7.156.52NANANANANANANA
FPV/RTV Treatment GroupNANA8.664.346.405.853.924.914.93

[back to top]

Plasma APV CL/F Following Dosing Expressed in mg/kg

Apparent clearance of drug from plasma following extravascular administration (CL/F) was calculated using the formulation: APV Dose in mg/kg units divided by AUC(0-τ). For FPV, doses were expressed in APV molar equivalents (50 mg of FPV = 43.2 mg of APV). Normalizing CL/F for bodyweight allows for comparison of CL/F across populations. (NCT00089583)
Timeframe: Week 48

,
InterventionMilliliters/minute/kilogram (mL/min/kg) (Geometric Mean)
2 to <6yrs, 30 mg/kg BID; n=9, 02 to <6yrs, 40 mg/kg BID; n=7, 02 to <6yrs, 23/3 mg/kg BID; n=0, 146 to <12 yrs, 15/3 mg/kg BID; n=0, 96 to <12 yrs, 18/3 mg/kg BID; n=0, 126 to <12 yrs, 700/100 mg BID; n=0, 312 to 18 yrs, 15/3 mg/kg BID; n=0, 412 to 18 yrs, 18/3 mg BID; n=0, 312 to 18 yrs, 700/100 mg BID; n=0, 13
FPV Treatment Group19.323.4NANANANANANANA
FPV/RTV Treatment GroupNANA6.066.485.275.9410.16.005.33

[back to top]

Plasma Amprenavir (APV) AUC (0-tau[τ])

Plasma samples were assayed for APV concentrations using a validated assay. The GlaxoSmithKline (GSK) Department of Clinical Pharmacology Modeling and Simulation conducted pharmacokinetic (PK) analysis of the plasma APV concentration-time data using a model-independent approach. As a measure of total drug exposure, the area under the plasma-concentration-versus-time curve over the dosing interval at steady-state (AUC[0-τ]), where τ is the length of the dosing interval, was calculated by the linear up/log down trapezoidal method. hr, hour; µg, micrograms; mL, milliliter. (NCT00089583)
Timeframe: Week 48

,
Interventionhr*µg/mL (Geometric Mean)
2 to <6 yrs, 30 mg/kg BID; n=9, 02 to <6 yrs, 40 mg/kg BID; n=7, 02 to <6 yrs, 23/3 mg/kg BID; n=0, 146 to <12 yrs, 15/3 mg/kg BID; n=0, 96 to <12 yrs, 18/3 mg/kg BID; n=0, 126 to <12 yrs, 700/100 mg BID; n=0, 312 to 18 yrs, 15/3 mg/kg BID; n=0, 412 to 18 yrs, 18/3 mg BID; n=0, 312 to 18 yrs, 700/100 mg BID; n=0, 13
FPV Treatment Group22.324.1NANANANANANANA
FPV/RTV Treatment GroupNANA55.332.348.437.621.841.735.3

[back to top]

Percentage of Total Lymphocytes (TLs) That Are CD4+ Cells at Baseline and Weeks 2, 12, 24, and 48

Blood samples of participants were collected for the measurement of the percentage of total lymphocytes that are CD4+ cells. Observed analysis was used for the summary of proportion endpoints using viral load data. (NCT00089583)
Timeframe: Baseline and Weeks 2, 12, 24, and 48

,
InterventionPercentage of TLs that are CD4+ cells (Median)
PI-naïve, Baseline; n=19, 49PI-naïve, Week 2; n=13, 41PI-naïve, Week 12; n=19, 46PI-naïve, Week 24; n=18, 44PI-naïve, Week 48; n=18, 42PI-exp, Baseline; n=0, 40PI-exp, Week 2; n=0, 32PI-exp, Week 12; n=0, 31PI-exp, Week 24; n=0, 34PI-exp, Week 48; n=0, 29
FPV Treatment Group1924273132NANANANANA
FPV/RTV Treatment Group21232528292422232324

[back to top]

Number of Participants With at Least a 1.0 log10 HIV-1 RNA Decrease From Baseline at Weeks 2, 12, 24, and 48 (Observed Analysis)

Blood samples of participants were collected to assess the decrease in the number of HIV-1 RNA. (NCT00089583)
Timeframe: Baseline and Weeks 2, 12, 24, and 48

,
Interventionparticipants (Number)
PI-naïve, Week 2; n=14, 39PI-exp, Week 2; n=0, 32PI-naïve, Week 12; n=19, 46PI-exp, Week 12; n=0, 33PI-naïve, Week 24; n=18, 44PI-exp, Week 24; n=0, 35PI-naïve, Week 48; n=18, 44PI-exp, Week 48; n=0, 33
FPV Treatment Group13NA19NA18NA17NA
FPV/RTV Treatment Group3522412641294024

[back to top]

Number of Participants Reporting Perfect Adherence Over the 3 Days Prior to the Study Visits at Weeks 2, 12, 24, and 48 as Assessed by Study Coordinator Using the Pediatric AIDS Clinical Trials Group (PACTG) Adherence Questionnaire

The PACTG Adherence Questionnaire records individual study drugs, the expected number of doses/24 hour period, and the number of doses missed in the 3 days prior to the study visit. Responses were summarized by age cohort, study drug, treatment regimen, and visit for exploratory analysis only. (NCT00089583)
Timeframe: Weeks 2, 12, 24, and 48

,
Interventionparticipants (Number)
Week 2, Total Population; n= 17, 59Week 2, 2 to <6 years (yrs); n= 17, 16Week 2, 6 to <12 yrs; n= 0, 25Week 2, 12 to 18 yrs; n= 0, 18Week 12, Total Population; n= 16, 55Week 12, 2 to <6 yrs; n= 16, 16Week 12, 6 to <12 yrs; n= 0, 24Week 12, 12 to 18 yrs; n= 0, 15Week 24, Total Population; n= 16, 54Week 24, 2 to <6 yrs; n= 16, 14Week 24, 6 to <12 yrs; n= 0, 24Week 24, 12 to 18 yrs; n= 0, 16Week 48, Total Population; n= 15, 53Week 48, 2 to <6 yrs; n= 15, 14Week 48, 6 to <12 yrs; n= 0, 23Week 48, 12 to 18 yrs; n= 0, 16
FPV Treatment Group1515NANA1515NANA1616NANA1313NANA
FPV/RTV Treatment Group491522124514201143112394213209

[back to top]

Number of Participants (Par.) With Virological Outcome (Plasma HIV-1 Ribonucleic Acid [RNA] <400 Copies/mL) at Week 48

Blood samples of participants were collected to measure plasma HIV-1 RNA concentrations. PI-exp = PI-experienced.Virologic success was defined as plasma HIV-1 RNA <400 copies/mL. Virologic failure: (1) HIV-1 RNA >=400 copies/mL, (2) change of background antiretroviral treatment (ART), (3) discontinued study due to lack of efficacy, (4) discontinued study with last HIV-1 >=400 copies/mL. No virologic data at Week 48 window: (a) discontinued study due to an adverse event or death, (b) discontinued study due to other reasons, (c) missing data during window but still on study. (NCT00089583)
Timeframe: Week 48

,
Interventionparticipants (Number)
PI-naïve, virological (V) success; n=20, 49PI-exp, V success; n=0, 40PI-naïve, V failure (1); n=20, 49PI-exp, V failure (1); n=0, 40PI-naïve, V failure (2); n=20, 49PI-exp, V failure (2); n=0, 40PI-naïve, V failure (3); n=20, 49PI-exp, V failure (3); n=0, 40PI-naïve;, V failure (4); n=20, 49PI-exp, V failure (4); n=0, 40PI-naïve, No V data at Week 48 (a); n=20, 49PI-exp, No V data at Week 48 (a); n=0, 40PI-naïve, No V data Week 48 (b); n=20, 49PI-exp, No V data at Week 48 (b); n=0, 40PI-naïve, No V data at Week 48 (c); n=20, 49PI-exp, No V data at Week 48 (c); n=0, 40
FPV Treatment Group12NA4NA2NA1NA1NA0NA0NA0NA
FPV/RTV Treatment Group361938470033111012

[back to top]

Change in CD4 Counts Cells/mm^3 From Week 36 for IT Arm and From Week 0 for DT Arm

(NCT00090779)
Timeframe: IT arm (weeks 36, 60, 72, 84 and 96) and DT arm (weeks 0, 24, 36, 48 and 60)

,
InterventionChange in Log10 transformed CD4 Counts (Mean)
IT arm (wk 60- wk 36) vs. DT arm (wk 24- wk 0)IT arm (wk 72- wk 36) vs. DT arm (wk 36- wk 0)IT arm (wk 84- wk 36) vs. DT arm (wk 48- wk 0)IT arm (wk 96- wk 36) vs. DT arm (wk 60- wk 0)
DT Arm-0.02-0.03-0.06-0.02
IT Arm-0.11-0.10-0.10-0.12

[back to top]

Time From Study Entry in DT Arm Participants or From Week 36 in IT Arm Participants to Meeting the Clinical, Virologic, or Immunologic Criteria for Treatment Initiation or Re-initiation

5th, 10th, 25th, 50th, 75th and 90th percentiles in weeks from randomization for DT arm or from week 36 for IT arm to meeting the criteria for treatment initiation or re-initiation which include two consecutive CD4 count below 350 cells/mm^3 at least 4 weeks apart, at least 12 weeks into the study or 12 weeks post-treatment discontinuation, confirmed CD4 count below 200 cells/mm^3 or CD4 percent below 14% at any time on study, confirmed HIV-1 RNA level above 750,000 copies/mL 4 weeks into the study or above 200,000 copies/mL 12 weeks or more into the study, or CDC Category B or C diagnosis. (NCT00090779)
Timeframe: 96 weeks since randomization

,
Interventionweeks (Number)
5th percentile10th percentile25th percentile50th percentile75th percentile90th percentile
DT Arm6.912.326.360.096.096.0
IT Arm5.110.422.758.1NANA

[back to top]

Time to Meeting the Clinical, Virologic, or Immunologic Criteria for Treatment Initiation or Re-initiation

5th, 10th, 25th, 50th, 75th and 90th percentiles in weeks from randomization to meeting the criteria for treatment initiation or re-initiation which include CD4 count below 350 cells/mm^3 on two consecutive measurements at least 4 weeks apart, at least 12 weeks into the study or 12 weeks post-treatment discontinuation, confirmed CD4 count below 200 cells/mm^3 or CD4 percent below 14% at any time on study, confirmed HIV-1 RNA level above 750,000 copies/mL 4 weeks into the study or above 200,000 copies/mL 12 weeks or more into the study, or CDC Category B or C diagnosis. (NCT00090779)
Timeframe: 96 weeks since randomization

,
Interventionweeks (Number)
5th percentile10th percentile25th percentile50th percentile75th percentile90th percentile
DT Arm6.912.326.360.096.096.0
IT Arm6.313.036.472.0NANA

[back to top]

Time to Treatment Initiation or Death

5th, 10th, 25th, 50th and 75th percentiles in weeks from randomization to treatment initiation or death (NCT00090779)
Timeframe: 5 years since randomization

,
Interventionweeks (Number)
5th percentile10th percentile25th percentile50th percentile75th percentile
DT Arm13.920.943.797.3157.7
IT Arm3636.967.196.4163.3

[back to top]

Number of Participants Experiencing Either a CDC Category B or C Diagnosis, CD4<200 Cells/mm^3 or CD4 Percent <14%.

(NCT00090779)
Timeframe: 96 weeks since randomization

Interventionparticipants (Number)
IT Arm2
DT Arm8

[back to top]

Number of Participants in IT Arm Off Treatment Before 36 Weeks

The study provided fixed-dose combination emtricitabine/tenofovir DF 200/300 mg orally once daily and lopinavir/ritonavir 200/50 mg administered either as two tablets twice daily or four tablets once daily, for the first 36 weeks for individuals in the IT arm. (NCT00090779)
Timeframe: At Week 36

Interventionparticipants (Number)
IT Arm8

[back to top]

Ranked Log10 HIV-1 RNA Viral Load (log10 Copies/mL) Averaged at 72 and 76 Weeks for the IT Arm and DT Arm

"The primary endpoint is (i) the average of log10 viral loads (VL) at wks 72 and 76 for participants who continued to wk 72 off ARV for the DT arm, (ii) average wk 72 and 76 VL for those who continued to wk 36 off ARV for the IT arm and (iii) an assigned VL rank for the failures who needed ARVs or met criteria for entry into Step 2 prior to these study visits. The assigned rank for the failures was either the last observed rank carried forward or the worst rank relative to the other possible outcomes. This approach was designed to, if anything, bias against finding a treatment effect. To illustrate, consider five participants who enter the study (A, B, C, D, and E), 4 of whom (A, B, C, D) make it to 72 wks off therapy with RNA levels that increase from A to D. Participant E enters Step 2 at wk 12, at which time his RNA is in the 50th percentile. This rank would be carried forward, so the rank order of the log10 HIV-1 RNA endpoints would be A B E C D." (NCT00090779)
Timeframe: At Weeks 72 and 76

Interventionrank (Median)
IT Arm26.0
DT Arm49.3

[back to top]

Ranked log10 HIV-1 RNA Viral Load (log10 Copies/mL) Averaged at Weeks 72 and 76 for the IT Arm and Ranked log10 HIV-1 RNA Viral Load (log10 Copies/mL) Averaged at Weeks 36 and 40 for the DT Arm

"The primary endpoint is (i) average wk 36 and 40 VL for those who continued to wk 36 off ARV for the DT arm, (ii) average wk 72 and 76 VL for those who continued to wk 36 off ARV for the IT arm and (iii) an assigned VL rank for the failures who needed ARVs or met criteria for entry into Step 2 prior to these study visits. The assigned rank for the failures was either the last observed rank carried forward or the worst rank relative to the other possible outcomes. This approach was designed to, if anything, bias against finding a treatment effect. To illustrate, consider five participants who enter the study (A, B, C, D, and E), 4 of whom (A, B, C, D) make it to 72 wks off therapy with RNA levels that increase from A to D. Participant E enters Step 2 at wk 12, at which time his RNA is in the 50th percentile. This rank would be carried forward, so the rank order of the log10 HIV-1 RNA endpoints would be A B E C D." (NCT00090779)
Timeframe: IT arm (weeks 72 and 76) and DT arm ( weeks 36 and 40)

Interventionrank (Median)
IT Arm26.0
DT Arm48.5

[back to top]

Number of Participants Meeting Clinical, Virologic, or Immunologic Criteria for Treatment Initiation or Re-initiation

The clinical, virologic, or immunologic criteria for treatment initiation or re-initiation include CD4 count below 350 cells/mm^3 on two consecutive determinations at least 4 weeks apart, at least 12 weeks into the study or 12 weeks post-treatment discontinuation, (2) confirmed CD4 count below 200 cells/mm^3 or CD4 percent below 14% at any time on study, (3) confirmed HIV-1 RNA level above 750,000 copies/mL 4 weeks into the study or above 200,000 copies/mL 12 weeks or more into the study, or (4) CDC Category B or C diagnosis. (NCT00090779)
Timeframe: 96 weeks since randomization

InterventionParticipants (Number)
IT Arm7
DT Arm23

[back to top]

Severe (Grade 3) and Higher Adverse Events and Any Grade Adverse Event That Leads to a Treatment Change From First Day of Study Treatment to Week 12

"Grade 3 or higher signs and symptoms, laboratory abnormalities, events that are reported through the EAE system, and any grade event that leads to a treatment change from first day of study treatment to week 12.~Grade 3 = Severe Grade 4 = Life threatening Grade 5 = Death" (NCT00099632)
Timeframe: From first day of study treatment to week 12

Interventionparticipants (Number)
7-day Lamivudine/Zidovudine (3TC/ZDV)5
21-day Lamivudine/Zidovudine (3TC/ZDV)1
7-day Emtricitabine/Tenofovir Disoproxil Fumarate (FTC/TDF)1
21-day Emtricitabine/Tenofovir Disoproxil Fumarate (FTC/TDF)0
7-day Lopinavir/Ritonavir (LPV/r)2
21-day Lopinavir/Ritonavir (LPV/r)2

[back to top]

Number of Participants With New PI-resistant Variants as Detected by Standard Composite (Bulk) Genotyping.

For the 7-day treatment duration group, only the genotype results from weeks 3 and 7 contributed; For the 21-day treatment duration groups, only the genotype results from weeks 5 and 9 contributed. (NCT00099632)
Timeframe: 2 and 6 weeks after completion of treatment

Interventionparticipants (Number)
7-day Lamivudine/Zidovudine (3TC/ZDV)0
21-day Lamivudine/Zidovudine (3TC/ZDV)0
7-day Emtricitabine/Tenofovir Disoproxil Fumarate (FTC/TDF)0
21-day Emtricitabine/Tenofovir Disoproxil Fumarate (FTC/TDF)0
7-day Lopinavir/Ritonavir (LPV/r)0
21-day Lopinavir/Ritonavir (LPV/r)0

[back to top]

Number of Participants With New Circulating NRTI-resistant Variants Detected by Standard Composite (Bulk) Genotyping.

For the 7-day treatment duration group, only the genotype results from weeks 3 and 7 contributed; For the 21-day treatment duration groups, only the genotype results from weeks 5 and 9 contributed. (NCT00099632)
Timeframe: 2 and 6 weeks after completion of treatment

Interventionparticipants (Number)
7-day Lamivudine/Zidovudine (3TC/ZDV)0
21-day Lamivudine/Zidovudine (3TC/ZDV)1
7-day Emtricitabine/Tenofovir Disoproxil Fumarate (FTC/TDF)1
21-day Emtricitabine/Tenofovir Disoproxil Fumarate (FTC/TDF)1
7-day Lopinavir/Ritonavir (LPV/r)1
21-day Lopinavir/Ritonavir (LPV/r)0

[back to top]

Number of Participants Who Discontinued Study Treatment Prematurely

participants assigned to 7-day treatment arm and 21-day treatment arm were supposed to stay in study treatment for 7 days and 21 days respectively. (NCT00099632)
Timeframe: From first day of study treatment to last day of study treatment (up to 21 days)

Interventionparticipants (Number)
7-day Lamivudine/Zidovudine (3TC/ZDV)0
21-day Lamivudine/Zidovudine (3TC/ZDV)2
7-day Emtricitabine/Tenofovir Disoproxil Fumarate (FTC/TDF)0
21-day Emtricitabine/Tenofovir Disoproxil Fumarate (FTC/TDF)0
7-day Lopinavir/Ritonavir (LPV/r)0
21-day Lopinavir/Ritonavir (LPV/r)5

[back to top]

Number of Participants With New Circulating Nonnucleoside Reverse Transcriptase Inhibitor (NNRTI)-Resistant Variants as Detected by Standard Composite (Bulk) Genotyping

"For the 7-day treatment duration group, only the genotype results from weeks 3 and 7 contributed to the primary endpoint; For the 21-day treatment duration groups, only the genotype results from weeks 5 and 9 contributed to primary endpoint.~10 participants who did not have resistance samples available were excluded from the primary endpoint analysis." (NCT00099632)
Timeframe: 2 and 6 weeks after completion of treatment

Interventionparticipants (Number)
7-day Lamivudine/Zidovudine (3TC/ZDV)1
21-day Lamivudine/Zidovudine (3TC/ZDV)0
7-day Emtricitabine/Tenofovir Disoproxil Fumarate (FTC/TDF)0
21-day Emtricitabine/Tenofovir Disoproxil Fumarate (FTC/TDF)0
7-day Lopinavir/Ritonavir (LPV/r)3
21-day Lopinavir/Ritonavir (LPV/r)1

[back to top]

Time to First Hospitalization

To compare time to first hospitalization in the three randomized arms (infants who received early ART in Arms 2 and 3 and those who received deferred ART in Arm 1). Not all participants were hospitalized and thus the upper limits could not be evaluated. (NCT00102960)
Timeframe: From randomization up to 4.8 years

InterventionWeeks (Median)
Deferred Therapy73.1
Early Therapy 40 WeeksNA
Early Therapy 96 WeeksNA

[back to top]

Total Occurrence of Grade 3 or 4 Clinical Events

This was a secondary outcome measure that assessed the total count of Grade 3 or 4 (clinical or laboratory) adverse events. (NCT00102960)
Timeframe: 4.8 years

InterventionCount of events (Number)
Deferred Therapy170
Early Therapy 40 Weeks118
Early Therapy 96 Weeks88

[back to top]

Total Occurrence of Grade 3 or 4 Laboratory Events

(NCT00102960)
Timeframe: From randomization up to 4.8 years

InterventionCount of events (Number)
Deferred Therapy35
Early Therapy 40 Weeks44
Early Therapy 96 Weeks33

[back to top]

Number of Children Experiencing Severe CDC Stage B or Stage C Disease or Death (Cumulative After 3.5 Years)

The outcome measure is defined as a number because it represents the number of children that experienced severe CDC Stage B or Stage C disease or death as defined in the outcome measure title above (NCT00102960)
Timeframe: Occurrence of severe CDC Stage B or Stage C disease or death (cumulative after 3.5 years), whichever came first, was assessed from randomization up to at least 3.5 years.

InterventionParticipants (Count of Participants)
Deferred Therapy41
Early Therapy 40 Weeks28
Early Therapy 96 Weeks21

[back to top]

Duration of Hospitalisation

This is the total number of days spent in hospital by the participants and is reported per arm (NCT00102960)
Timeframe: 4.8 years, the study duration

InterventionDays (Number)
Deferred Therapy1018
Early Therapy 40 Weeks533
Early Therapy 96 Weeks414

[back to top]

Hospitalization Rates

Hospitalisation rates in the three arms enrolled in the CHER study (NCT00102960)
Timeframe: 4.8 years

InterventionEvents per 100 person years (Number)
Deferred Therapy27.6
Early Therapy 40 Weeks16.4
Early Therapy 96 Weeks14.2

[back to top]

Number of Participants Who Experienced Clinical Failure (Defined as Development of Severe CDC Stage B or Stage C Disease.) on Therapy.

This included development of severe CDC Stage B or Stage C disease.This was part of the primary outcome measure that was a composite endpoint (NCT00102960)
Timeframe: Clinical failure on therapy was assessed at each visit for the entire study duration of 4.8 years.

InterventionParticipants (Count of Participants)
Deferred Therapy8
Early Therapy 40 Weeks6
Early Therapy 96 Weeks5

[back to top]

Number of Participants Who Experienced Immunological Failure Defined as Failure of CD4% to Reach 20% or CD4% Falls Below 20% on Two Occasions, Within 4 Weeks, at Any Time After the First 24 Weeks of Therapy (Initial Therapy or Restart)

This was part of the primary outcome measure above. The primary outcome was a composite endpoint. The primary outcome analysis only considered the initially enrolled children that were 377 in total (ART-Deferred n=125, Early therapy 40 weeks n=126 and Early therapy 96 weeks n=126). This was part of the primary outcome measure that was a composite endpoint. (NCT00102960)
Timeframe: This outcome was assessed from the date of randomization to immunological failure. Immunological failure was assessed in the entire study duration of 4.8 years.

InterventionParticipants (Count of Participants)
Deferred Therapy9
Early ART for 40 Weeks14
Early Therapy for 96 Weeks11

[back to top]

Number of Participants Who Experienced Regimen-limiting ART Drug Toxicity

Development of toxicity requiring more than one drug substitution within the same class or a switch to a new class of drugs (regimen-limiting toxicity failure) or requiring a permanent treatment discontinuation. This was part of the primary outcome measure that was a composite endpoint. (NCT00102960)
Timeframe: Regimen limiting drug toxicity was monitored from randomization up to the entire study duration of 4.8 years.

InterventionParticipants (Count of Participants)
Deferred Therapy0
Early Therapy 40 Weeks0
Early Therapy 96 Weeks0

[back to top]

Number of Participants Who Experienced Virological Failure Defined as Confirmed HIV-1 RNA Value of at Least 10,000 Copies Per/ml Recorded on Two Consecutive Separate Occasions After 24 Weeks of Treatment (Initial Therapy or Restart)

This was part of the primary outcome measure that was a composite endpoint that included confirmed HIV-1 RNA value of at least 10,000 copies per/ml recorded on two consecutive separate occasions after 24 weeks of treatment (initial therapy or restart). (NCT00102960)
Timeframe: Virological failure was assessed from randomization through the entire study duration of 4.8 years.

InterventionParticipants (Count of Participants)
Deferred Therapy10
Early Therapy 40 Weeks1
Early Therapy 96 Weeks1

[back to top]

Time From Randomization to Starting or Needing to Start Continuous Therapy

Time from randomization to starting (deferred therapy Arm) or needing to start continuous therapy (early therapy 40 or 96 weeks) (NCT00102960)
Timeframe: 4.8 years

InterventionWeeks (Median)
Deferred Therapy20
Early Therapy 40 Weeks33
Early Therapy 96 Weeks70

[back to top]

Time to Death Alone or Death Plus Life Threatening Stage C Events or HIV Events Associated With Permanent End-organ Damage.

This was a composite endpoint in which the number of children experiencing the events is reported. The number of participants experiencing the events did not reach the 50% survival and thus median time-to-event is not be presented. Therefore, we report the number of participants experiencing the events per Arm. (NCT00102960)
Timeframe: 4.8 years

InterventionParticipants (Count of Participants)
Deferred Therapy34
Early Therapy 40 Weeks18
Early Therapy 96 Weeks13

[back to top]

Time to Failure of First Line Therapy or Death

To compare time to failure of first line ART (due to clinical, virological or immunological disease progression, or regimen-limiting ART toxicities) or death among three randomized arms (infants who receive early ART in Arms 2 and 3 and infants in whom ART is deferred until clinical or immunological disease progression in Arm 1) during the study (up to 4.8 years). The number of participants experiencing the events did not reach the 50% survival and thus median time-to-event is not be presented. Therefore we report the number of participants experiencing the events per Arm. (NCT00102960)
Timeframe: From date of randomization up to failure of first-line therapy or death from any cause, whichever came first, assessed up to 4.8 years

InterventionParticipants (Count of Participants)
Deferred Therapy48
Early Therapy up to 40 Weeks32
Early Therapy up to 96 Weeks26

[back to top]

Number of Participants Assessed for Adverse Events (AEs)

Detailed information for Adverse Events and Serious Adverse Events will be represented in the SAE/AE section of PRS. (NCT00105079)
Timeframe: reported up to 28 days after the last dose of study treatment. (Up to 52 weeks)

Interventionparticipants (Number)
Saquinavir/Ritonavir163
Lopinavir/Ritonavir168

[back to top]

Number of Patients Who Discontinued Treatment Due to Abnormal Laboratory Parameters

Routine clinical testing, including hematology and standard chemistry panel was performed at all study visits. Laboratory tests for a fasting lipid profile and fasting insulin determination were obtained at baseline, weeks 24 and 48, and the 4-week follow-up visit. The number of participants who discontinued treatment due to an abnormal laboratory result at any visit is reported. (NCT00105079)
Timeframe: baseline and all study visits (Up to Week 52)

Interventionparticipants (Number)
Saquinavir/Ritonavir0
Lopinavir/Ritonavir0

[back to top]

Change From Baseline in Cluster Differentiation Antigen 4 Positive (CD4+) Lymphocyte Count

Summary statistics for change from baseline in CD4+ lymphocyte count were presented by treatment arm. Change from baseline in CD4+ lymphocyte count was derived as follows: Change from baseline = (CD4+ count at week x) - (CD4+ count at baseline). (NCT00105079)
Timeframe: Baseline to Week 48

,
Interventioncells/mm^3 (Median)
Baseline (n=166,169)Week 48 (n=122,131)Change from Baseline to Week 48 (n=121,130)
Lopinavir/Ritonavir142.0348.0204.0
Saquinavir/Ritonavir141.5319.0178.0

[back to top]

Change From Baseline in HIV-1 RNA Viral Load

Descriptive statistics for change from baseline in log10 transformed plasma HIV-1 RNA load (copies/mL) were presented by treatment arm. Logarithmic transformation (base 10) was applied to HIV-1 RNA viral load at baseline and at each study visit. Change from baseline in plasma HIV-1 RNA was derived as follows: Change from baseline = Log10 (HIV-1 RNA at week x) - Log10 (HIV-1 RNA at baseline) (NCT00105079)
Timeframe: Baseline to Week 48

,
Interventioncopies/mL (Mean)
BaselineWeek 48 (n=126,133)Change from Baseline to Week 48 (n=126,133)
Lopinavir/Ritonavir5.171.83-3.36
Saquinavir/Ritonavir5.201.80-3.39

[back to top]

Number of Patients With HIV-1 RNA Viral Load <50 and <400 Copies/mL

"The secondary objectives of the study were to evaluate the safety, adherence, and tolerability of saquinavir/ritonavir BID plus emtricitabine/tenofovir QD versus lopinavir/ritonavir BID plus emtricitabine/tenofovir QD in treatment-naïve HIV-1 infected adults.~Blood samples for HIV-1 RNA viral load measurement were collected at the Week 48 clinic visit. The number of participants with HIV-1 RNA results <50 copies/mL and the number of participants with HIV-1 RNA results <400 copies/mL are reported." (NCT00105079)
Timeframe: Week 48

,
Interventionparticipants (Number)
Patients with <50 Copies/mLPatients with <400 Copies/mL
Lopinavir/Ritonavir108127
Saquinavir/Ritonavir108121

[back to top]

Number of Patients With Human Immunodeficiency Virus Type 1 (HIV-1) Ribonucleic Acid (RNA) Viral Load <50 Copies/mL

"The primary objective of this study was to evaluate the efficacy of saquinavir/ritonavir BID plus emtricitabine/tenofovir QD versus lopinavir/ritonavir BID plus emtricitabine/tenofovir QD in treatment-naïve HIV-1 infected adults.~Blood samples for HIV-1 RNA viral load measurement were collected at the Week 48 clinic visit. The number of participants with HIV-1 RNA results <50 copies/mL is reported." (NCT00105079)
Timeframe: Week 48

,
Interventionparticipants (Number)
Pts. with HIV-1 RNA Viral Load <50 copies/mL - YESPts. with HIV-1 RNA Viral Load <50 copies/mL - NO
Lopinavir/Ritonavir10862
Saquinavir/Ritonavir10859

[back to top]

Pre-dose Concentration Pharmacokinetic Outcome for LPV/r (Cpredose ug/mL).

Data was analyzed with WinNonLin (Version 5.2, Pharsight, USA) using non-compartmental methods. The pharmacokinetic parameters were calculated using the linear-trapezoidal rule. Cpredose and C4hour at the two measurement times were compared within-subject using the Wilcoxon signed-rank test. (NCT00109590)
Timeframe: Within 72 hours postpartum and during the first 30 days postpartum

Interventionug/mL (Median)
Within 72 Hrs Ppm6.08
At Day 30 Ppm9.17

[back to top]

Median HIV-1 Viral Load at 24 Weeks Postpartum in Women

(NCT00109590)
Timeframe: at 24 weeks postpartum

Interventionlog10 copies/mL (Median)
Arm A : LPV/r x 7d4.3
Arm B : no LPV/r3.9
Arm C: LPV/r x 30d4.0

[back to top]

Number of Women With Grade >=3 Events After Start of Study Treatment

Adverse events were graded using the Division of AIDS (DAIDS) Table for Grading > the Severity of Adult and Pediatric Adverse Events (December 2004). All grade 3 and higher signs, symptoms, and laboratory toxicities (and events of any grade that led to a change in study treatment) were included. (NCT00109590)
Timeframe: After start of study Treatment (postpartum)

Interventionparticipants (Number)
Arm A : LPV/r x 7d2
Arm B : no LPV/r0
Arm C: LPV/r x 30d2

[back to top]

Four (4) Hour Concentration Pharmacokinetic Outcome for LPV/r (C4hour ug/mL).

Data was analyzed with WinNonLin (Version 5.2, Pharsight, USA) using non-compartmental methods. The pharmacokinetic parameters were calculated using the linear-trapezoidal rule. Cpredose and C4hour at the two measurement times were compared within-subject using the Wilcoxon signed-rank test. (NCT00109590)
Timeframe: Within 72 hours postpartum and during the first 30 days postpartum

Interventionug/mL (Median)
Within 72 Hrs Ppm10.78
At Day 30 Ppm12.96

[back to top]

Resistance Mutations in HIV Infected Infants

Resistance mutations as identified by consensus sequencing or OLA (NCT00109590)
Timeframe: 24 weeks postpartum

Interventionparticipants (Number)
Arm B : no LPV/r0
Arm C: LPV/r x 30d0

[back to top]

The Proportion of Women in Each Randomized Arm Who Have One or More New NVP Resistance Mutations as Identified by Consensus Sequencing or Oligonucleotide Ligation Assay (OLA) in Plasma

The incidence of new NVP resistance mutations at day 10 or week 6 postpartum in each randomized arm. Samples with viral load <500 copies/mL were considered free of mutations. If a resistance result was missing for reasons other than VL <500 copies/ml it was conservatively imputed as resistant in the primary analysis. (NCT00109590)
Timeframe: at Day 10 or Week 6 postpartum.

Interventionpercent of participants (Number)
Arm A : LPV/r x 7d3.6
Arm B : no LPV/r7.1
Arm C : LPV/r x 30d5.3

[back to top]

The Proportion of Women in Each Randomized Arm Who Have One or More New NVP Resistance Mutations for the Subgroup of Women With Plasma HIV RNA >= 500 Copies/ml At Entry

The incidence of new NVP resistance mutations at day 10 or week 6 postpartum in each randomized arm. Samples with viral load <500 copies/mL were considered free of mutations. If a resistance result was missing for reasons other than VL <500 copies/ml it was conservatively imputed as resistant in the primary analysis. (NCT00109590)
Timeframe: at Day 10 or Week 6 postpartum.

Interventionpercent of participants (Number)
Arm A: LPV/r x 7d4.9
Arm B: no LPV/r9.5
Arm C : LPV/r x 30d7.0

[back to top]

The Proportion of Women Who Develop One or More New NVP Resistance Mutations as Identified by Consensus Sequencing or Oligonucleotide Ligation Assay in Plasma (Sampling Was Done at Days 10,21,30, and Weeks 5,6, and 8 Postpartum).

The incidence of new NVP resistance mutation in plasma HIV within 8 weeks postpartum in each randomized arm was estimated using an exact binomial confidence interval. If a resistance mutation was detected at any of the timepoints then an endpoint was met. Samples with VL <500 copies/mL were considered free of mutations. If a resistance result was missing for reasons other than VL <500 copies/ml (e.g.missed visit), it was conservatively imputed as resistant in the primary analysis. (NCT00109590)
Timeframe: within 8 weeks postpartum.

Interventionpercent of participants (Number)
Arm A : LPV/r x 7d7.1
Arm B : no LPV/r12.5
Arm C: LPV/r x 30d5.3

[back to top]

Proportion of Women With New NVP Resistance Mutation Within 8 Weeks Postpartum Who Had a NVP Resistance Mutation Detected at 72 Weeks Postpartum.

Resistance mutations as identified by OLA in plasma samples or PBMC at 72 weeks postpartum amongst women who had new NVP resistance mutations within 8 weeks postpatrum. These results were based on the 13 women who developed a new NVP resistance mutation in the first 8 weeks postpartum. For the primary outcome measure 1, one particpant in arm A was unavailable for follow-up after week 5 and was conservatively imputed to have developed resistance mutation. (NCT00109590)
Timeframe: within 72 weeks postpartum

,,
Interventionparticipants (Number)
OLA in plasma samplesOLA in PBMC
Arm A : LPV/r x 7d00
Arm B : no LPV/r00
Arm C: LPV/r x 30d01

[back to top]

The Proportion of Women With Any New ZDV, ddI, or LPV/r Resistance Mutations.

(NCT00109590)
Timeframe: At Week 5 postpartum (ZDV) and at the first timepoint with viral load >=500 copies/ml after treatment discontinuation (ddI and LPV/r).

,,
Interventionpercent of participants (Number)
The proportion of women with new ZDV resistanceThe proportion of women with new ddI resistanceThe proportion of women with new LPV/r resistance
Arm A : LPV/r x 7d000
Arm B : no LPV/r1.7800
Arm C: LPV/r x 30d000

[back to top]

Maximum Concentration Pharmacokinetic Outcome for LPV/r (Cmax ug/mL) .

Data was analyzed with WinNonLin (Version 5.2, Pharsight, USA) using non-compartmental methods. The pharmacokinetic parameters were calculated using the linear-trapezoidal rule. Cpredose and C4hour at the two measurement times were compared within-subject using the Wilcoxon signed-rank test. (NCT00109590)
Timeframe: Within 72 hours postpartum and during the first 30 days postpartum

Interventionug/mL (Median)
Within 72 Hrs Ppm11.2
At Day 30 PpmNA

[back to top]

Area Under the Curve Pharmacokinetic Outcome for LPV/r. (AUC ug*hr/mL)

Data was analyzed with WinNonLin (Version 5.2, Pharsight, USA) using non-compartmental methods. The pharmacokinetic parameters were calculated using the linear-trapezoidal rule. Cpredose and C4hour at the two measurement times were compared within-subject using the Wilcoxon signed-rank test. (NCT00109590)
Timeframe: Within 72 hours postpartum and during the first 30 days postpartum

Interventionug*hr/mL (Median)
Within 72 Hrs Ppm99.7
At Day 30 PpmNA

[back to top]

Time From Treatment Dispensation to Regimen Failure (First Occurrence of Virologic Failure or Treatment Modification)

Blood samples for determining virologic failure were obtained at 16 and 24 weeks, and every 12 weeks thereafter. Virologic failure was defined as a confirmed plasma HIV-1 RNA level >= 1000 copies/mL at or after 16 weeks and before 24 weeks or >=200 copies/mL at or after 24 weeks. Treatment modification was defined as the 1st modification of the regimen, including a permanent discontinuation, switch, or substitution. (NCT00118898)
Timeframe: Follow-up time was variable,median follow-up was 138 weeks; see 'Amount of study follow-up' outcome for details

,,,
InterventionWeeks (Number)
5th percentile time to regimen failure10th percentile time to regimen failure25th percentile time to regimen failure
EFV, FTC/TDF, and Placebo ABC/3TC41672
EFV, Placebo FTC/TDF, and ABC/3TC4424
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC41684
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC4436

[back to top]

Time From Treatment Dispensation to Treatment Modification

Treatment modification is defined as the 1st modification of the regimen, including a permanent discontinuation, switch, or substitution. (NCT00118898)
Timeframe: Follow-up time was variable,median follow-up was 138 weeks; see 'Amount of study follow-up' outcome for details

,,,
InterventionWeeks (Number)
5th percentile time to treatment modification10th percentile time to treatment modification25th percentile time to treatment modification
EFV, FTC/TDF, and Placebo ABC/3TC3.415.083.7
EFV, Placebo FTC/TDF, and ABC/3TC1.42.127.4
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC7.924.9108.9
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC1.65.043.6

[back to top]

Number of Participants With Virologic Failure

Blood samples for determining virologic failure were obtained at 16 and 24 weeks, and every 12 weeks thereafter. Virologic failure was defined as a confirmed plasma HIV-1 RNA level >= 1000 copies/mL at or after 16 weeks and before 24 weeks or >=200 copies/mL at or after 24 weeks. (NCT00118898)
Timeframe: Follow-up time was variable, median follow-up was 138 weeks; see 'Amount of study follow-up' outcome for details

Interventionparticipants (Number)
EFV, FTC/TDF, and Placebo ABC/3TC57
EFV, Placebo FTC/TDF, and ABC/3TC72
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC57
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC83

[back to top]

Number of Participants With Virologic Failure and Emergence of Major Resistance

Emergence of resistant virus was assessed by genotypic testing performed at Stanford University for all participants who met criteria for virologic failure and retrospectively on baseline samples from these participants. Major mutations were defined by International AIDS Society-United States of America (2008), as well as T69D, L74I, G190C/E/Q/T/V for reverse transcriptase and L24I, F53L, I54V/A/T/S, G73C/S/T/A, N88D for protease. (NCT00118898)
Timeframe: Follow-up time was variable,median follow-up was 138 weeks; see 'Amount of study follow-up' outcome for details

Interventionparticipants (Number)
EFV, FTC/TDF, and Placebo ABC/3TC27
EFV, Placebo FTC/TDF, and ABC/3TC41
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC5
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC12

[back to top]

Change in CD4 Count (Cells/mm3) From Baseline

Change was calculated as the CD4 count at Week 48 (or at Week 96) minus the baseline CD4 count (mean of pre-entry and entry values). (NCT00118898)
Timeframe: At Weeks 48 and 96

,,,
InterventionCells/mm3 (Median)
Week 48Week 96
EFV, FTC/TDF, and Placebo ABC/3TC163220.5
EFV, Placebo FTC/TDF, and ABC/3TC188250.5
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC175251.5
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC177.5250.3

[back to top]

Change in Fasting High-density Lipoprotein (HDL) Cholesterol Level From Baseline

Only fasting results are included. The protocol did not require that samples be collected fasting. (NCT00118898)
Timeframe: At Weeks 48 and 96

,,,
Interventionmg/dL (Median)
Week 48Week 96
EFV, FTC/TDF, and Placebo ABC/3TC89
EFV, Placebo FTC/TDF, and ABC/3TC1011
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC54
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC87

[back to top]

Change in Fasting Non-high Density Lipoprotein (Non-HDL) Cholesterol Level From Baseline

Only fasting results are included. The protocol did not require that samples be collected fasting. (NCT00118898)
Timeframe: At Weeks 48 and 96

,,,
Interventionmg/dL (Median)
Week 48Week 96
EFV, FTC/TDF, and Placebo ABC/3TC1413.5
EFV, Placebo FTC/TDF, and ABC/3TC2318
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC810
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC2018

[back to top]

Change in Fasting Total Cholesterol Level From Baseline

Only fasting results are included. The protocol did not require that samples be collected fasting. (NCT00118898)
Timeframe: At Weeks 48 and 96

,,,
Interventionmg/dL (Median)
Week 48Week 96
EFV, FTC/TDF, and Placebo ABC/3TC2223
EFV, Placebo FTC/TDF, and ABC/3TC3533
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC1114
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC3025

[back to top]

Change in Fasting Triglyceride Level From Baseline

Only fasting results are included. The protocol did not require that samples be collected fasting. (NCT00118898)
Timeframe: At Weeks 48 and 96

,,,
Interventionmg/dL (Median)
Week 48Week 96
EFV, FTC/TDF, and Placebo ABC/3TC109
EFV, Placebo FTC/TDF, and ABC/3TC1514
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC1411
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC2433

[back to top]

Time From Treatment Dispensation to a Grade 3/4 Safety Event

Grade 3/4 safety event is defined as a grade 3 or 4 sign, symptom, or laboratory abnormality that is at least one grade higher than at baseline, total bilirubin and creatine kinase (CPK) were excluded. Grading used the Division of AIDS (DAIDS) 2004 Severity of Adverse Events Tables. (NCT00118898)
Timeframe: All follow-up while on initially assigned regimen; the median (25th, 75th percentile) follow-up while on initial regimen was 120 (54, 156) weeks and the range was 0 to 205 weeks.

,,,
InterventionWeeks (Number)
5th percentile time to a grade 3/4 safety event10th percentile time to a grade 3/4 safety event25th percentile time to a grade 3/4 safety event
EFV, FTC/TDF, and Placebo ABC/3TC2.67.959.3
EFV, Placebo FTC/TDF, and ABC/3TC1.32.016.0
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC3.08.181.4
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC1.33.944.4

[back to top]

Cumulative Probability of Not Experiencing a Grade 3/4 Safety Event

Kaplan-Meier estimate of the cumulative survival probability at week 48 and 96. Grade 3/4 safety event is defined as a grade 3 or 4 sign, symptom, or laboratory abnormality that is at least one grade higher than at baseline, total bilirubin and creatine kinase (CPK) were excluded. Grading used the Division of AIDS (DAIDS) 2004 Severity of Adverse Events Tables. As-treated analysis censored at 1st modification of initially assigned regimen, participants who never started treatment were excluded. (NCT00118898)
Timeframe: At week 48 and 96

,,,
Interventionpercentage of participants (Number)
Week 48Week 96
EFV, FTC/TDF, and Placebo ABC/3TC7870
EFV, Placebo FTC/TDF, and ABC/3TC6458
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC7973
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC7366

[back to top]

Cumulative Probability of Not Experiencing Regimen Failure

Kaplan-Meier estimate of the cumulative survival probability at week 48 and 96. Blood samples for determining virologic failure were obtained at 16 and 24 weeks, and every 12 weeks thereafter. Virologic failure was defined as a confirmed plasma HIV-1 RNA level >= 1000 copies/mL at or after 16 weeks and before 24 weeks or >=200 copies/mL at or after 24 weeks. Treatment modification was defined as the 1st modification of the regimen, including a permanent discontinuation, switch, or substitution. (NCT00118898)
Timeframe: At week 48 and 96

,,,
Interventionpercentage of participants (Number)
Week 48Week 96
EFV, FTC/TDF, and Placebo ABC/3TC7970
EFV, Placebo FTC/TDF, and ABC/3TC6454
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC8073
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC6657

[back to top]

Cumulative Probability of Not Experiencing Treatment Modification

Kaplan-Meier estimate of the cumulative survival probability at week 48 and 96. Treatment modification is defined as the 1st modification of the regimen, including a permanent discontinuation, switch, or substitution. (NCT00118898)
Timeframe: At week 48 and 96

,,,
Interventionpercentage of participants (Number)
Week 48Week 96
EFV, FTC/TDF, and Placebo ABC/3TC8073
EFV, Placebo FTC/TDF, and ABC/3TC6756
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC8677
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC7362

[back to top]

Cumulative Probability of Not Experiencing Virologic Failure

Kaplan-Meier estimate of the cumulative survival probability at week 48 and 96. Blood samples for determining virologic failure were obtained at 16 and 24 weeks, and every 12 weeks thereafter. Virologic failure was defined as a confirmed plasma HIV-1 RNA level >= 1000 copies/mL at or after 16 weeks and before 24 weeks or >=200 copies/mL at or after 24 weeks. (NCT00118898)
Timeframe: At week 48 and 96

,,,
Interventionpercentage of participants (Number)
Week 48Week 96
EFV, FTC/TDF, and Placebo ABC/3TC9490
EFV, Placebo FTC/TDF, and ABC/3TC8885
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC9289
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC8883

[back to top] [back to top]

Number of Participants With HIV-1 RNA Levels Less Than 200 Copies/mL

(NCT00118898)
Timeframe: At Weeks 48 and 96

,,,
InterventionParticipants (Number)
Number of Participants with RNA data at Week 48Number with HIV-1 RNA <200 copies/ml at Week 48Number of Participants with RNA data at Week 96Number with HIV-1 RNA <200 copies/ml at Week 96
EFV, FTC/TDF, and Placebo ABC/3TC415398379362
EFV, Placebo FTC/TDF, and ABC/3TC400377361342
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC416391384368
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC411372374346

[back to top]

The Number of Participants With HIV-1 RNA Levels Less Than 50 Copies/mL

(NCT00118898)
Timeframe: At Weeks 48 and 96

,,,
InterventionParticipants (Number)
Number of Participants with RNA data at Week 48Number with HIV-1 RNA <50 copies/ml at Week 48Number of Participants with RNA data at Week 96Number with HIV-1 RNA <50 copies/ml at Week 96
EFV, FTC/TDF, and Placebo ABC/3TC415372379345
EFV, Placebo FTC/TDF, and ABC/3TC400346361328
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC416348384345
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC411322374317

[back to top]

Amount of Study Follow-up

Participants were to be followed for 96 weeks after the last enrollment. Accrual was expected to take 96 weeks, thus the planned follow-up time was 96 to 192 weeks, dependent on when in the study the participant enrolled. This outcome summarizes that total amount of actual follow-up in weeks from randomization to last contact. (NCT00118898)
Timeframe: Follow-up time was variable, median follow-up was 138 weeks

InterventionWeeks (Median)
EFV, FTC/TDF, and Placebo ABC/3TC141.4
EFV, Placebo FTC/TDF, and ABC/3TC133.3
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC141.6
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC137.3

[back to top]

Time From Randomization to Virologic Failure

Blood samples for determining virologic failure were obtained at visit weeks 16 and 24 , and every 12 weeks thereafter. Virologic failure was defined as a confirmed plasma HIV-1 RNA level >= 1000 copies/mL at or after 16 weeks after randomization and before 24 weeks, or >=200 copies/mL at or after 24 weeks. The 5th percentile for time to virologic failure is the time (in weeks) at which 5% of the participants have experienced virologic failure. (NCT00118898)
Timeframe: Follow-up time was variable,median follow-up was 138 weeks; see 'Amount of study follow-up' outcome for details

,,,
InterventionWeeks (Number)
5th percentile time to virologic failure10th percentile time to virologic failure
EFV, FTC/TDF, and Placebo ABC/3TC3696
EFV, Placebo FTC/TDF, and ABC/3TC2436
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC2484
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC2436

[back to top]

Number of Participants With Regimen Failure

Blood samples for determining virologic failure were obtained at 16 and 24 weeks, and every 12 weeks thereafter. Virologic failure was defined as a confirmed plasma HIV-1 RNA level >= 1000 copies/mL at or after 16 weeks and before 24 weeks or >=200 copies/mL at or after 24 weeks. Treatment modification was defined as the 1st modification of the regimen, including a permanent discontinuation, switch, or substitution. (NCT00118898)
Timeframe: Follow-up time was variable, median follow-up was 138 weeks; see 'Amount of study follow-up' outcome for details

Interventionparticipants (Number)
EFV, FTC/TDF, and Placebo ABC/3TC162
EFV, Placebo FTC/TDF, and ABC/3TC246
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC157
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC233

[back to top]

Number of Participants With Treatment Modification

Treatment modification is defined as the 1st modification of the regimen, including a permanent discontinuation, switch, or substitution. (NCT00118898)
Timeframe: Follow-up time was variable, median follow-up was 138 weeks; see 'Amount of study follow-up' outcome for details

Interventionparticipants (Number)
EFV, FTC/TDF, and Placebo ABC/3TC152
EFV, Placebo FTC/TDF, and ABC/3TC239
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC138
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC216

[back to top]

Number of Participants With a Grade 3/4 Safety Event

Grade 3/4 safety event is defined as a grade 3 or 4 sign, symptom, or laboratory abnormality that is at least one grade higher than at baseline, total bilirubin and creatine kinase (CPK) were excluded. Grading used the Division of AIDS (DAIDS) 2004 Severity of Adverse Events Tables. As-treated analysis censored at 1st modification of initially assigned regimen, participants who never started treatment were excluded. (NCT00118898)
Timeframe: Over all study follow-up while on initially assigned treatment, median follow-up was 120 weeks

Interventionparticipants (Number)
EFV, FTC/TDF, and Placebo ABC/3TC145
EFV, Placebo FTC/TDF, and ABC/3TC182
RTV-boosted ATV, FTC/TDF, and Placebo ABC/3TC137
RTV-boosted ATV, Placebo FTC/TDF, and ABC/3TC156

[back to top]

Mean Percent Change From Baseline in Visceral Adipose Tissue (VAT) Area by Computed Tomography (CT) Scans and in Trunk Fat by DEXA.

The mean percent change from baseline in physical signs of lipohypertrophy, as assessed objectively by changes in visceral adipose tissue (VAT) area (cm2) by computed tomography (CT) scans and by changes in trunk fat (kg) by DEXA. Clinical improvement is associated with a decrease in values. (Baseline values can be found in the Baseline Characteristics section.) (NCT00135356)
Timeframe: Baseline, Week 48, Week 96

,
InterventionPercent change (Mean)
Week 48 VAT (n=98; n=53)Week 96 VAT (n=101; n=53)Week 48 Trunk Fat (n=112; n=57)Week 96 Trunk Fat (n=112; n=57)
ATV/RTV Switch Arm6.54.32.61.6
PI/RTV Control Arm-0.42.1-1.8-3.6

[back to top]

Mean Percent Changes From Baseline in Fasting Lipids

Mean percent changes from baseline in fasting total, low-density lipoprotein (LDL), high-density lipoprotein (HDL), and non-HDL cholesterol, triglycerides, and apolipoprotein B (NCT00135356)
Timeframe: Baseline, Week 48, Week 96

,
InterventionPercent change (Number)
Week 48 - Total CholesterolWeek 48 - HDL CholesterolWeek 48 - Non-HDL CholesterolWeek 48 - LDL CholesterolWeek 48 - TriglyceridesWeek 48 - Apolipoprotein BWeek 96 - Total CholesterolWeek 96 - HDL CholesterolWeek 96 - Non-HDL CholesterolWeek 96 - LDL CholesterolWeek 96 - TriglyceridesWeek 96 - Apolipoprotein B
ATV/RTV Switch Arm-13.0-6.2-14.8-10.4-23.8-7.6-12.5-6.8-14.0-8.4-25.0-8.3
PI/RTV Control Arm-1.0-2.6-0.62.6-11.71.1-0.1-4.61.23.6-12.28.3

[back to top]

Percentage of Participants With Abnormal Liver Function Tests

Percentage of participants with Abnormal Alanine Aminotransferase (ALT), Aspartate Aminotransferase (AST), and Total Bilirubin (TBILI) measurements. Values for liver tests are graded using the modified World Health Organization (WHO) criteria. Grade 1 is mild, grade 2 is moderate, grade 3 is severe, grade 4 is life threatening or disabling. (NCT00135356)
Timeframe: Week 48, Week 96

,
InterventionPercentage of Participants (Number)
Wk 48 ALT Grades 1-4Wk 48 ALT Grades 3-4Wk 48 ALT Grade 4Wk 96 ALT Grades 1-4Wk 96 ALT Grades 3-4Wk 96 ALT Grade 4Wk 48 AST Grade 1-4Wk 48 AST Grades 3-4Wk 48 AST Grade 4Wk 96 AST Grades 1-4Wk 96 AST Grades 3-4Wk 96 AST Grade 4Wk 48 TBILI Grades 1-4Wk 48 TBILI Grades 3-4Wk 48 TBILI Grade 4Wk 96 TBILI Grades 1-4Wk 96 TBILI Grades 3-4Wk 96 TBILI Grade 4
ATV/RTV Switch Arm3410372019102410945313956017
PI/RTV Control Arm230032001500190019002100

[back to top]

Percentage of Participants With Adverse Events (AEs) Leading to Discontinuation

Percentage of Participants with AEs leading to discontinuation of study therapy. An AE is defined as any new untoward medical occurrence or worsening of a pre-existing medical condition. All events listed in this table were SAEs, except for renal impairment and hypertriglycerideamia, which were an AEs (and did not meet the 5 percent threshold reported in Adverse Event module of this record). (NCT00135356)
Timeframe: Through Week 96

,
InterventionPercent of Participants (Number)
Any adverse experience leading to discontinuationHyperbilirubinemiaJaundiceDrug abuseRenal impairmentStevens-Johnson syndromeHypertriglyceridemiaSquamous cell carcinoma
ATV/RTV Switch Arm52111100
PI/RTV Control Arm30000011

[back to top]

Percentage of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs), Deaths, and AEs Leading to Discontinuation

Percentage of Participants with AEs, Serious AEs (SAEs), Deaths, and AEs leading to discontinuation. An AE is defined as any new untoward medical occurrence or worsening of a pre-existing medical condition. An SAE is any untoward medical occurrence that at any dose results in death, is life-threatening, requires inpatient hospitalization or causes prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a cancer, is a congenital anomaly/birth defect, results in the development of drug dependency or drug abuse, is an important medical event. (NCT00135356)
Timeframe: Through Week 96 of study therapy

,
InterventionPercentage of Participants (Number)
DeathSAEAE Leading to DiscontinuationAny AEs (all grades) through Week 96
ATV/RTV Switch Arm08590
PI/RTV Control Arm07383

[back to top]

Mean Changes From Baseline in Body Weight at Week 48 and Week 96

(NCT00135356)
Timeframe: Baseline, Week 48, Week 96

,
Interventionkg (Mean)
Week 48 (n=130; n=68)Week 96 (n=130; n=68)
ATV/RTV Switch Arm10
PI/RTV Control Arm-1-1

[back to top]

Change From Baseline in Trunk-to-limb Fat Ratio as Measured by DEXA at Week 96

Mean changes from baseline in trunk-to-limb fat ratio as measured by DEXA, an x-ray scan used to measure bone mineral density. Clinical improvement is associated with a decrease in values.(Baseline trunk-to-limb fat ratio values can be found in the Baseline Characteristics section.) (NCT00135356)
Timeframe: Baseline, Week 96

,
Interventionratio (Mean)
LOCF Population (n=112; n=54)OC Population (n=94; n=45)
ATV/RTV Switch Arm0.040.04
PI/RTV Control Arm0.020.05

[back to top]

Change From Baseline in Trunk-to-limb Fat Ratio as Measured by Dual Energy X-Ray Absortiometry (DEXA) at Week 48

Mean changes from Baseline in trunk-to-limb fat ratio as measured by DEXA, an x-ray scan used to measure bone mineral density. Clinical improvement is associated with a decrease in values. (Baseline trunk-to-limb fat ratio values can be found in the Baseline Characteristics section.) (NCT00135356)
Timeframe: Baseline, Week 48

,
Interventionratio (Mean)
LOCF Population (n=112; n=54)OCPopulation (n=105; n=51)
ATV/RTV Switch Arm0.020.02
PI/RTV Control Arm-0.02-0.01

[back to top]

Kaplan-Meier Cumulative Proportion of Participants Without Virologic Rebound (HIV RNA ≥400 c/mL) at Timepoints up to Week 96 in Treated Participants With HIV RNA <400 c/mL at Baseline

Virologic rebound was measured from the first dose of study therapy to the first of the 2 consecutive measurements ≥400 c/mL. Time to virologic rebound was analyzed using life tables. Measured Values show the Kaplan-Meier cumulative proportion of participants without virologic rebound up to the end of the respective interval. (NCT00135356)
Timeframe: Weeks 8-12, Weeks 20-24, Weeks 32-36, Weeks 44-48, Weeks 56-60, Weeks 68-72, Weeks 80-84, Weeks 92-96

,
InterventionProportion of participants (Number)
By Weeks 8-12By Weeks 20-24By Weeks 32-36By Weeks 44-48By Weeks 56-60By Weeks 68-72By Weeks 80-84By Weeks 92-96
ATV/RTV Switch Arm0.98370.98370.97530.96690.95850.95850.95850.9585
PI/RTV Control Arm1.0001.0000.98410.98410.98410.98410.96740.9309

[back to top]

Mean Change From Baseline in CD4 Count

Mean change from baseline in CD4 count among treated subjects (NCT00135356)
Timeframe: Baseline, Week 48, Week 96

,
Interventioncells/mm3 (Mean)
Week 48 (n=114; n=56)Week 96 (n=96; n=54)
ATV/RTV Switch Arm143
PI/RTV Control Arm4482

[back to top]

Mean Changes From Baseline in Body Mass Index at Week 48 and Week 96

(NCT00135356)
Timeframe: Baseline, Week 48, Week 96

,
Interventionkg/m2 (Mean)
Week 48 (n=130; n=67)Week 96 (n=130; n=67)
ATV/RTV Switch Arm0.30.2
PI/RTV Control Arm-0.2-0.5

[back to top]

Mean Changes From Baseline in Fasting Glucose at Week 48 and Week 96

(NCT00135356)
Timeframe: Baseline, Week 48, Week 96

,
Interventionmg/dL (Mean)
Week 48 (n=124; n=63)Week 96 (n=124; n=64)
ATV/RTV Switch Arm3.61.2
PI/RTV Control Arm-3.33.0

[back to top]

Mean Changes From Baseline in Fasting Homeostasis Model Assessment of Insulin Resistance (HOMA-IR)

HOMA-IR is an index used in evaluation of obese patients at risk for type 2 diabetes which requires fasting glucose and insulin concentrations. It is a mathematical model based on the theory of a negative feedback loop between the liver and β-cells that regulates both fasting glucose and insulin concentrations and can be used to estimate pancreatic β-cell function and degree of insulin resistance. HOMA-IR normal values are between 2 and 2.5. HOMA-IR ≥ 2.5 indicates insulin-resistance. (NCT00135356)
Timeframe: Baseline, Week 48, Week 96

,
Interventionmg/dL x uU/mL (Mean)
Baseline (n=115; n=59)Week 48 (n=115; n=57)Week 96 (n=115; n=57)
ATV/RTV Switch Arm3.420.740.28
PI/RTV Control Arm5.43-1.731.00

[back to top]

Mean Changes From Baseline in Fasting Insulin at Week 48 and Week 96

(NCT00135356)
Timeframe: Baseline, Week 48, Week 96

,
Interventionmicrounits per milliliter (Mean)
Baseline (n=124; n=62)Week 48 (n=124; n=59)Week 96 (n=124; n=59)
ATV/RTV Switch Arm14.11.30.0
PI/RTV Control Arm21.8-4.10.3

[back to top]

Mean Changes From Baseline in Waist Circumference at Week 48 and Week 96

(NCT00135356)
Timeframe: Baseline, Week 48, Week 96

,
Interventioncm (Mean)
Week 48 (n=123; n=66)Week 96 (n=124; n=66)
ATV/RTV Switch Arm-1-1
PI/RTV Control Arm-1-1

[back to top]

Mean Changes From Baseline in Waist-to-Hip Ratio at Week 48 and Week 96

Mean changes from baseline in proportion of waist to hip measurements. (NCT00135356)
Timeframe: Baseline, Week 48, Week 96

,
Interventionratio (Mean)
Week 48 (n=123; n=65)Week 96 (n=124; n=65)
ATV/RTV Switch Arm-0.01-0.01
PI/RTV Control Arm-0.01-0.01

[back to top]

Mean Percent Change From Baseline in Peripheral Adipose Tissue (Limb Fat) by DEXA and by Changes in Subcutaneous Adipose Tissue (SAT) Area by CT Scans

The mean percent change from baseline in physical signs of lipoatrophy, as assessed objectively by changes in peripheral adipose tissue (ie, limb fat (kg) by DEXA and in subcutaneous adipose tissue (SAT) area by CT scans. Clinical improvement is associated with stable values, or an increase in values. (Baseline values can be found in the Baseline Characteristics section.) (NCT00135356)
Timeframe: Baseline, Week 48, Week 96

,
Interventionpercent change (Mean)
Week 48 SAT (n=108; n=59)Week 96 SAT (n=111; n=59)Week 48 Limb Fat (n=112; 54)Week 48 Limb Fat (n=112; n=54)
ATV/RTV Switch Arm-2.2-3.50.9-0.8
PI/RTV Control Arm-5.9-9.7-3.6-6.1

[back to top]

Mean Percent Change From Baseline in Total Body Fat by DEXA and in Total Adipose Tissue (TAT) Area by CT Scans

The mean percent change from baseline in total body fat by DEXA and in total adipose tissue (TAT) area by CT scans. Total body fat and TAT are both associated many factors (trunk fat + limb fat + other [weight, etc]), and thus clinical improvement cannot be predicted based solely an increase or decrease of these values. (Baseline values can be found in the Baseline Characteristics section.) (NCT00135356)
Timeframe: Baseline, Week 48, Week 96

,
Interventionpercent change (Mean)
Week 48 TAT (n=108; n= 59)Week 96 TAT (n=111; n=59)Week 48 Total Body Fat (n=105; n=51)Week 96 Total Body Fat (n=94; n=45)
ATV/RTV Switch Arm0.0-0.91.90.5
PI/RTV Control Arm-3.5-5.0-3.7-7.4

[back to top]

Virologic Response at Week 96

Virologic response defined as Viral Load<50 copies/mL (NCT00144170)
Timeframe: Week 96

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)20
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)8.9

[back to top]

Virologic Response at Week 32

Virologic response defined as Viral Load<50 copies/mL (NCT00144170)
Timeframe: Week 32

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)22.1
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)12.1

[back to top]

Virologic Response at Week 32

Virologic response is defined as: Log(baseline viral load (VL))-Log(on-treatment VL)>=1 (NCT00144170)
Timeframe: week 32

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)38.6
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)19.4

[back to top]

Treatment Response at Week 72

Patients who experienced treatment response. Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00144170)
Timeframe: week 72

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)30.1
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)12.1

[back to top]

Virologic Response at Week 4

Virologic response defined as Viral Load<400 copies/mL (NCT00144170)
Timeframe: Week 4

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)26.2
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)15.2

[back to top]

Virologic Response at Week 4

Virologic response defined as Viral Load<50 copies/mL (NCT00144170)
Timeframe: Week 4

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)4.4
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)3.5

[back to top]

Virologic Response at Week 4

Virologic response is defined as: Log(baseline viral load (VL))-Log(on-treatment VL)>=1 (NCT00144170)
Timeframe: week 4

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)62.3
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)33.4

[back to top]

Virologic Response at Week 40

Virologic response defined as Viral Load<400 copies/mL (NCT00144170)
Timeframe: Week 40

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)31.5
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)15

[back to top]

Median Change From Baseline in Viral Load (Week 8)

(NCT00144170)
Timeframe: Baseline to Week 8

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-1.4
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)-0.33

[back to top]

Median Change From Baseline in Viral Load (Week 80)

(NCT00144170)
Timeframe: Baseline to Week 80

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.55
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)-0.2

[back to top]

Median Change From Baseline in Viral Load (Week 88)

(NCT00144170)
Timeframe: Baseline to Week 88

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.56
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)-0.2

[back to top]

Median Change From Baseline in Viral Load (Week 96)

(NCT00144170)
Timeframe: Baseline to Week 96

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.56
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)-0.2

[back to top]

Time to Confirmed Virologic Failure Through 48 Weeks of Treatment

Time to virologic failure is defined as the time from the start of treatment to the last measurement where the Log(baseline viral load)-Log(on-treatment viral load)>1 before a 2 consecutive measurements where Log(baseline viral load)-Log(on-treatment viral load)<1. (NCT00144170)
Timeframe: after 48 weeks of treatment

InterventionDays (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)117
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)0

[back to top]

Time to Confirmed Virologic Failure Through 96 Weeks of Treatment

Time to virologic failure is defined as the time from the start of treatment to the last measurement where the Log(baseline viral load)-Log(on-treatment viral load)>1 before a 2 consecutive measurements where Log(baseline viral load)-Log(on-treatment viral load)<1. (NCT00144170)
Timeframe: after 96 weeks of treatment

InterventionDays (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)117
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)0

[back to top]

Time to New Centers for Disease Control (CDC) Class C Progression Event or Death.

"Time to death or occurrence of AIDS-defining condition according to the US Centers for Disease Control and Prevention case definition.~The median and quartiles are underestimated since more than 92% of the observations (in both treatment arms) were censored and the estimation was restricted to the largest observed event time." (NCT00144170)
Timeframe: up to 75 weeks of treatment

InterventionDays (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)528
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)332

[back to top]

Time to Treatment Failure Through 48 Weeks of Treatment

Time to treatment failure is defined as 0 for patients who never achieve TR otherwise time to treatment failure is the earliest time of death, discontinuation of the study drug or introduction of a new anti-retroviral drug to the regimen if it is not solely related to either toxicity or intolerance clearly attributable to a background, or the first of two consecutive visits with Log(baseline Viral Load) - Log(on-treatment Viral Load) < 1. (NCT00144170)
Timeframe: after 48 weeks of treatment

InterventionDays (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)112
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)0

[back to top]

Mean Change From Baseline in CD4+ Cell Count (Week 56)

(NCT00144170)
Timeframe: Baseline to Week 56

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)39
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)16

[back to top]

Mean Change From Baseline in CD4+ Cell Count (Week 64)

(NCT00144170)
Timeframe: Baseline to Week 64

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)42
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)13

[back to top]

Mean Change From Baseline in CD4+ Cell Count (Week 72)

(NCT00144170)
Timeframe: Baseline to Week 72

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)42
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)18

[back to top]

Mean Change From Baseline in CD4+ Cell Count (Week 8)

(NCT00144170)
Timeframe: Baseline to Week 8

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)52
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)25

[back to top]

Mean Change From Baseline in CD4+ Cell Count (Week 80)

(NCT00144170)
Timeframe: Baseline to Week 80

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)39
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)19

[back to top]

Mean Change From Baseline in CD4+ Cell Count (Week 88)

(NCT00144170)
Timeframe: Baseline to Week 88

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)41
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)19

[back to top]

Mean Change From Baseline in CD4+ Cell Count (Week 96)

(NCT00144170)
Timeframe: Baseline to Week 96

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)41
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)20

[back to top]

Median Change From Baseline in Viral Load (Week 16)

(NCT00144170)
Timeframe: Baseline to Week 16

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-1.03
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)-0.3

[back to top]

Virologic Response at Week 40

Virologic response defined as Viral Load<50 copies/mL (NCT00144170)
Timeframe: Week 40

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)22.3
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)11.9

[back to top]

Virologic Response at Week 40

Virologic response is defined as: Log(baseline viral load (VL))-Log(on-treatment VL)>=1 (NCT00144170)
Timeframe: week 40

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)37
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)17.3

[back to top]

Virologic Response at Week 48

Virologic response defined as Viral Load<400 copies/mL (NCT00144170)
Timeframe: Week 48

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)30.1
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)13.6

[back to top]

Virologic Response at Week 48

Virologic response defined as Viral Load<50 copies/mL (NCT00144170)
Timeframe: Week 48

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)22.8
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)10.5

[back to top]

Virologic Response at Week 96

Virologic response is defined as: Log(baseline viral load (VL))-Log(on-treatment VL)>=1 (NCT00144170)
Timeframe: Week 96

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)30.1
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)11.4

[back to top]

Virologic Response at Week 48

Virologic response is defined as: Log(baseline viral load (VL))-Log(on-treatment VL)>=1 (NCT00144170)
Timeframe: week 48

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)36.8
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)16.6

[back to top]

Virologic Response at Week 96

Virologic response defined as Viral Load<400 copies/mL (NCT00144170)
Timeframe: week 96

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)27.1
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)10.3

[back to top]

Time to Treatment Failure Through 96 Weeks of Treatment

Time to treatment failure is defined as 0 for patients who never achieve TR otherwise time to treatment failure is the earliest time of death, discontinuation of the study drug or introduction of a new anti-retroviral drug to the regimen if it is not solely related to either toxicity or intolerance clearly attributable to a background, or the first of two consecutive visits with Log(baseline Viral Load) - Log(on-treatment Viral Load) < 1. (NCT00144170)
Timeframe: after 96 weeks of treatment

InterventionDays (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)115
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)0

[back to top]

Virologic Response at Week 88

Virologic response is defined as: Log(baseline viral load (VL))-Log(on-treatment VL)>=1 (NCT00144170)
Timeframe: Week 88

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)31
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)12.1

[back to top]

Median Change From Baseline in Viral Load (Week 2)

(NCT00144170)
Timeframe: Baseline to Week 2

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-1.27
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)-0.56

[back to top]

Virologic Response at Week 88

Virologic response defined as Viral Load<50 copies/mL (NCT00144170)
Timeframe: Week 88

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)20.7
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)8.9

[back to top]

Virologic Response at Week 88

Virologic response defined as Viral Load<400 copies/mL (NCT00144170)
Timeframe: week 88

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)27.8
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)11.2

[back to top]

Virologic Response at Week 80

Virologic response is defined as: Log(baseline viral load (VL))-Log(on-treatment VL)>=1 (NCT00144170)
Timeframe: Week 80

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)32.6
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)13.6

[back to top]

Virologic Response at Week 80

Virologic response defined as Viral Load<50 copies/mL (NCT00144170)
Timeframe: Week 80

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)21.8
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)8.9

[back to top]

Virologic Response at Week 80

Virologic response defined as Viral Load<400 copies/mL (NCT00144170)
Timeframe: Week 80

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)28.7
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)11

[back to top]

Virologic Response at Week 8

Virologic response is defined as: Log(baseline viral load (VL))-Log(on-treatment VL)>=1 (NCT00144170)
Timeframe: week 8

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)56.3
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)26.6

[back to top]

Virologic Response at Week 8

Virologic response defined as Viral Load<50 copies/mL (NCT00144170)
Timeframe: Week 8

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)11
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)6.8

[back to top]

Virologic Response at Week 8

Virologic response defined as Viral Load<400 copies/mL (NCT00144170)
Timeframe: Week 8

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)33.3
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)17.5

[back to top]

Virologic Response at Week 72

Virologic response is defined as: Log(baseline viral load (VL))-Log(on-treatment VL)>=1 (NCT00144170)
Timeframe: Week 72

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)32.2
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)13.8

[back to top]

Virologic Response at Week 72

Virologic response defined as Viral Load<50 copies/mL (NCT00144170)
Timeframe: Week 72

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)21.1
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)8.2

[back to top]

Virologic Response at Week 72

Virologic response defined as Viral Load<400 copies/mL (NCT00144170)
Timeframe: Week 72

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)28
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)11.4

[back to top]

Virologic Response at Week 64

Virologic response is defined as: Log(baseline viral load (VL))-Log(on-treatment VL)>=1 (NCT00144170)
Timeframe: week 64

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)33.6
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)14

[back to top]

Virologic Response at Week 64

Virologic response defined as Viral Load<50 copies/mL (NCT00144170)
Timeframe: Week 64

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)21.6
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)8.2

[back to top]

Median Change From Baseline in Viral Load (Week 24)

(NCT00144170)
Timeframe: Baseline to Week 24

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.69
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)-0.21

[back to top]

Median Change From Baseline in Viral Load (Week 32)

(NCT00144170)
Timeframe: Baseline to Week 32

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.63
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)-0.24

[back to top]

Median Change From Baseline in Viral Load (Week 4)

(NCT00144170)
Timeframe: Baseline to Week 4

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-1.47
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)-0.42

[back to top]

Median Change From Baseline in Viral Load (Week 40)

(NCT00144170)
Timeframe: Baseline to Week 40

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.65
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)-0.2

[back to top]

Median Change From Baseline in Viral Load (Week 48)

(NCT00144170)
Timeframe: Baseline to Week 48

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.65
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)-0.2

[back to top]

Mean Change From Baseline in CD4+ Cell Count (Week 48)

(NCT00144170)
Timeframe: Baseline to Week 48

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)43
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)16

[back to top]

Median Change From Baseline in Viral Load (Week 56)

(NCT00144170)
Timeframe: Baseline to Week 56

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.64
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)-0.22

[back to top]

Median Change From Baseline in Viral Load (Week 64)

(NCT00144170)
Timeframe: Baseline to Week 64

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.63
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)-0.21

[back to top]

Median Change From Baseline in Viral Load (Week 72)

(NCT00144170)
Timeframe: Baseline to Week 72

InterventionLog(Copies/mL) (Median)
Tipranavir(TPV)/Low Dose Ritonavir(r)-0.52
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)-0.2

[back to top]

Mean Change From Baseline in CD4+ Cell Count (Week 40)

(NCT00144170)
Timeframe: Baseline to Week 40

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)47
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)19

[back to top]

Mean Change From Baseline in CD4+ Cell Count (Week 4)

(NCT00144170)
Timeframe: Baseline to Week 4

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)39
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)19

[back to top]

Mean Change From Baseline in CD4+ Cell Count (Week 32)

(NCT00144170)
Timeframe: Baseline to Week 32

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)46
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)17

[back to top]

Treatment Response at Week 16

Patients who experienced treatment response. Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00144170)
Timeframe: week 16

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)47.1
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)19.9

[back to top]

Treatment Response at Week 2

Patients who experienced treatment response. Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00144170)
Timeframe: week 2

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)51
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)25.9

[back to top]

Mean Change From Baseline in CD4+ Cell Count (Week 24)

(NCT00144170)
Timeframe: Baseline to Week 24

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)51
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)21

[back to top]

Treatment Response at Week 24

Patients who experienced treatment response. Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00144170)
Timeframe: Week 24

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)40.9
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)18.0

[back to top]

Virologic Response at Week 64

Virologic response defined as Viral Load<400 copies/mL (NCT00144170)
Timeframe: Week 64

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)28.7
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)12.1

[back to top]

Virologic Response at Week 56

Virologic response defined as Viral Load<50 copies/mL (NCT00144170)
Timeframe: Week 56

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)22.5
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)8.9

[back to top]

Virologic Response at Week 56

Virologic response is defined as: Log(baseline viral load (VL))-Log(on-treatment VL)>=1 (NCT00144170)
Timeframe: week 56

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)35.2
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)15.2

[back to top]

Virologic Response at Week 56

Virologic response defined as Viral Load<400 copies/mL (NCT00144170)
Timeframe: Week 56

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)30.6
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)12.4

[back to top]

Treatment Response at Week 32

Patients who experienced treatment response. Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00144170)
Timeframe: week 32

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)37.2
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)17.3

[back to top]

Treatment Response at Week 4

Patients who experienced treatment response. Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00144170)
Timeframe: week 4

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)59.1
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)29.9

[back to top]

Treatment Response at Week 40

Patients who experienced treatment response. Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00144170)
Timeframe: week 40

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)36.3
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)16.6

[back to top]

Treatment Response at Week 48

Patients who experienced treatment response. Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00144170)
Timeframe: after 48 weeks of treatment

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)34.5
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)15

[back to top]

Mean Change From Baseline in CD4+ Cell Count (Week 2)

(NCT00144170)
Timeframe: Baseline to Week 2

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)26
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)13

[back to top]

Mean Change From Baseline in CD4+ Cell Count (Week 16)

(NCT00144170)
Timeframe: Baseline to Week 16

InterventionCells/mm3 (Mean)
Tipranavir(TPV)/Low Dose Ritonavir(r)57
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)25

[back to top]

Treatment Response at Week 56

Patients who experienced treatment response. Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00144170)
Timeframe: week 56

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)32.6
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)14.5

[back to top]

Treatment Response at Week 64

Patients who experienced treatment response. Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00144170)
Timeframe: week 64

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)31
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)13.3

[back to top]

Treatment Response at Week 8

Patients who experienced treatment response. Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00144170)
Timeframe: week 8

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)54.3
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)24.5

[back to top]

Treatment Response at Week 80

Patients who experienced treatment response. Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00144170)
Timeframe: week 80

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)28.5
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)11.4

[back to top]

Treatment Response at Week 88

Patients who experienced treatment response. Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00144170)
Timeframe: week 88

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)27.8
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)11

[back to top]

Treatment Response at Week 96

Patients who experienced treatment response. Treatment response (TR) is defined as two consecutive VL ≥ 1 log10 below baseline without discontinuation of study drug, change in anti-retroviral background, or rebound (NCT00144170)
Timeframe: after 96 weeks of treatment

Interventionpercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)26.2
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)10

[back to top]

Virologic Response

Virologic response defined as Viral Load<50 copies/mL (NCT00144170)
Timeframe: Week 2 through Week 96 (at any point during trial)

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)36.6
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)16.8

[back to top]

Virologic Response

Virologic response is defined as: Log(baseline viral load (VL))-Log(on-treatment VL)>=1 (NCT00144170)
Timeframe: Week 2 through Week 96 (at any point during trial)

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)75.6
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)48.6

[back to top]

Virologic Response at Viral Load Nadir During Study Treatment Through 96 Weeks

Virologic response defined as Viral Load<400 copies/mL (NCT00144170)
Timeframe: Week 2 through Week 96 (at any point during trial)

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)52.2
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)25.7

[back to top]

Virologic Response at Week 16

Virologic response defined as Viral Load<400 copies/mL (NCT00144170)
Timeframe: Week 16

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)35.2
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)17.5

[back to top]

Virologic Response at Week 16

Virologic response defined as Viral Load<50 copies/mL (NCT00144170)
Timeframe: Week 16

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)20.5
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)10.5

[back to top]

Virologic Response at Week 16

Virologic response is defined as: Log(baseline viral load (VL))-Log(on-treatment VL)>=1 (NCT00144170)
Timeframe: week 16

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)47.1
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)20.8

[back to top]

Virologic Response at Week 2

Virologic response defined as Viral Load<400 copies/mL (NCT00144170)
Timeframe: Week 2

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)14.5
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)8.2

[back to top]

Virologic Response at Week 2

Virologic response defined as Viral Load<50 copies/mL (NCT00144170)
Timeframe: Week 2

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)0.9
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)1.4

[back to top]

Virologic Response at Week 2

Virologic response is defined as: Log(baseline viral load (VL))-Log(on-treatment VL)>=1 (NCT00144170)
Timeframe: Week 2

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)60.5
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)36

[back to top]

Virologic Response at Week 24

Viral Load < 50 copies/mL (NCT00144170)
Timeframe: Week 24

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)21.1
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)11.9

[back to top]

Virologic Response at Week 24

Virologic response defined as Viral Load<400 copies/mL (NCT00144170)
Timeframe: Week 24

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)32.6
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)15.4

[back to top]

Virologic Response at Week 24

Virologic response is defined as: Log(baseline viral load (VL))-Log(on-treatment VL)>=1 (NCT00144170)
Timeframe: week 24

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)42.8
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)19.2

[back to top]

Virologic Response at Week 32

Virologic response defined as Viral Load<400 copies/mL (NCT00144170)
Timeframe: Week 32

InterventionPercentage of participants (Number)
Tipranavir(TPV)/Low Dose Ritonavir(r)32.4
Comparator Protease Inhibitor(CPI)/Low Dose Ritonavir(r)16.1

[back to top]

Rates of Virologic Failure

Virologic failure defined as HIV RNA > 2,000 copies/mL (NCT00145795)
Timeframe: 6 months

Interventionpercentage of randomized subjects (Number)
Kaletra + Current Dual NRTI Backbone0
Current Regimen0

[back to top] [back to top]

Immune Reconstitution [3 Months]

Immune reconstitution is defined as the absolute CD4+ lymphocyte count after 3 months of therapy. Absolute CD4+ T cell count, our measure of immune recovery, was assessed in the clinical laboratory using fluorescent labeled monoclonal antibodies to the CD4 on lymphocytes. This is the main target cell for HIV infection. The absolute CD4+ T cell count is also the only clinically validated surrogate marker of immune dysfunction in HIV. CD4+ count is also our best predictor of morbidity and mortality outcomes. (NCT00145795)
Timeframe: 3 months

Interventioncells per cubic millimeter (Mean)
Kaletra + Current Dual NRTI Backbone41.56
Current Regimen49.40

[back to top]

Immune Reconstitution [6 Months]

Immune reconstitution is defined as the absolute CD4+ lymphocyte count after 6 months of therapy. Absolute CD4+ T cell count, our measure of immune recovery, was assessed in the clinical laboratory using fluorescent labeled monoclonal antibodies to the CD4 on lymphocytes. This is the main target cell for HIV infection. The absolute CD4+ T cell count is also the only clinically validated surrogate marker of immune dysfunction in HIV. CD4+ count is also our best predictor of morbidity and mortality outcomes. (NCT00145795)
Timeframe: 6 months

Interventioncells per cubic millimeter (Mean)
Kaletra + Current Dual NRTI Backbone116
Current Regimen32

[back to top]

Rates of ex Vivo T Cell Apoptosis: CD8+ Cell Population [3 Months]

(NCT00145795)
Timeframe: 3 months

Interventionpercent apoptosis (Mean)
Kaletra + Current Dual NRTI Backbone20.92
Current Regimen16.74

[back to top]

Rates of ex Vivo T Cell Apoptosis: CD8+ Cell Population [6 Months]

(NCT00145795)
Timeframe: 6 months

Interventionpercent apoptosis (Mean)
Kaletra + Current Dual NRTI Backbone17.07
Current Regimen19.01

[back to top]

Rates of ex Vivo T Cell Apoptosis: CD4+ naïve Cell Population [3 Months]

Ex vivo T cell apoptosis can be assessed many different ways. The use of propidium iodide staining to determine the proportion of isolated cells that have undergone apoptosis after ex vivo incubation is a standard method that has been used by many investigators. Apoptotic cells intercalate less PI into their DNA, and on flow cytometry, this cell population is identified by a decrease in mean fluorescence (shift to the left). We have experience with this assay, and we have published on the use of method for determining rates of ex vivo apoptosis for different immune effector cells. (NCT00145795)
Timeframe: 3 months

Interventionpercent apoptosis (Mean)
Kaletra + Current Dual NRTI Backbone16.60
Current Regimen22.53

[back to top]

Rates of ex Vivo T Cell Apoptosis: CD4+ naïve Cell Population [6 Months]

(NCT00145795)
Timeframe: 6 months

Interventionpercent apoptosis (Mean)
Kaletra + Current Dual NRTI Backbone10.03
Current Regimen18.92

[back to top]

Rates of ex Vivo T Cell Apoptosis: CD4+ Memory Cell Population [3 Months]

Ex vivo T cell apoptosis can be assessed many different ways. The use of propidium iodide staining to determine the proportion of isolated cells that have undergone apoptosis after ex vivo incubation is a standard method that has been used by many investigators. Apoptotic cells intercalate less PI into their DNA, and on flow cytometry, this cell population is identified by a decrease in mean fluorescence (shift to the left). We have experience with this assay, and we have published on the use of method for determining rates of ex vivo apoptosis for different immune effector cells. (NCT00145795)
Timeframe: 3 months

Interventionpercent apoptosis (Mean)
Kaletra + Current Dual NRTI Backbone15.27
Current Regimen24.53

[back to top]

Rates of ex Vivo T Cell Apoptosis: CD4+ Memory Cell Population [6 Months]

(NCT00145795)
Timeframe: 6 months

Interventionpercent apoptosis (Mean)
Kaletra + Current Dual NRTI Backbone14.10
Current Regimen17.94

[back to top]

Change From Baseline in HIV-1 RNA at Week 24 of the Induction Phase

Change From Baseline in HIV-1 RNA at Week 24 of the Induction Phase. Change=Week 24 Induction Phase value - Baseline value; a decrease signifies improvement. (NCT00207142)
Timeframe: Baseline, Week 24 of Induction Phase

Interventionlog10 c/mL (Mean)
Randomized Subjects-3.16
Nonrandomized Subjects-3.35
Total-3.21

[back to top]

Time to Suppression (Confirmed HIV-1 RNA < 400 c/mL) During Treatment Phase

Time to suppression was measured from the first dose of Induction Phase study therapy to the first of the 2 consecutive measurements <400 c/mL. Time to suppression was analyzed using life tables. Measured Values show the Kaplan-Meier cumulative number of treated participants without suppression up to the end of the respective interval. (NCT00207142)
Timeframe: Week 16-18, Week 24-26, Week 30-32

,
InterventionParticipants (Number)
Number of Participants Suppressed by Week 16-18Number of Participants Suppressed by Wk 24-26Number of Participants Suppressed by Wk 30-32
Nonrandomized Subjects395455
Randomized Subjects168171172

[back to top]

Time to Suppression (Confirmed HIV-1 RNA < 50 c/mL) During Treatment Phase

Description: Time to suppression was measured from the first dose of Induction Phase study therapy to the first of the 2 consecutive measurements < 50 c/mL. Time to suppression was analyzed using life tables. Measured Values show the Kaplan-Meier cumulative number of treated participants without suppression up to the end of the respective interval. (NCT00207142)
Timeframe: Week 16-18, Week 24-26, Week 38-40, Week 64-66

,
InterventionParticipants (Number)
Number of Participants Suppressed by Wk 16-18Number of Participants Suppressed by Wk 24-26Number of Participants Suppressed by Wk 38-40Number of Participants Suppressed by Wk 64-66
Nonrandomized Subjects353142
Randomized Subjects131171172172

[back to top]

Change From Baseline in CD4 Cell Count at Week 24 of Induction Phase

Change From Baseline in CD4 Count at Week 24 of Induction Phase. Change=Week 24 Induction Phase value - Baseline value; a decrease signifies worsening. (NCT00207142)
Timeframe: Baseline, Week 24 of Induction Phase

Interventioncells/mm3 (Mean)
Randomized Subjects170
Nonrandomized Subjects201
Total177

[back to top]

Change From Baseline in CD4 Cell Count at Week 48 of Rescue Phase

Change From Baseline in CD4 Count at Week 48 of Rescue Phase. Change=Week 48 Rescue Phase value - Baseline value; a decrease signifies worsening. (NCT00207142)
Timeframe: Baseline, Week 48 of Rescue Phase

Interventioncells/mm3 (Mean)
Rescue Treatment313

[back to top]

Kaplan-Meier Cumulative Proportion for Treatment Failure (HIV-1 RNA ≥50 c/mL) at Different Time Points Through Week 48 of the Maintenance Phase

Treatment failure based on HIV-1 RNA ≥ 50 c/mL was defined as virologic rebound on or before Week 48 or discontinuation of study therapy before Week 48 for any reason. Time to treatment failure was analyzed using life tables. Measured Values shows the Kaplan-Meier cumulative proportion of participants without treatment failure up to the end of the respective interval. (NCT00207142)
Timeframe: Weeks 6-8, Weeks 14-16, Weeks 22-24, Weeks 30-32, Weeks 38-40, Weeks 46-48

,
InterventionProportion of participants (Number)
Proportion by Week 6-8Proportion by Week 14-16Proportion by Week 22-24Proportion by Week 30-32Proportion by Week 38-40Proportion by Week 46-48
Continuation Regimen0.94120.91760.90590.88240.83430.8176
Switch Regimen0.96550.96550.95400.93100.85060.8273

[back to top]

Treatment Outcomes Based on Viral Loads (HIV-1 RNA ≥50 c/mL) Through the End of Rescue Phase

Treatment outcome is based on the first reason of failure. These analyses were performed using HIV-1 RNA of 50 c/mL to define suppression and virologic rebound. (NCT00207142)
Timeframe: Through Week 48 of Rescue Phase. Measurements were included from the end of Induction Phase through the last dose of Rescue Phase study therapy plus 4 days.

InterventionParticipants (Number)
Suppression Maintained Through Week 48Suppression, then Virologic Rebound at/before Wk48Discontinuation Before Week 48Viral Suppression Never Achieved
Rescue Treatment29786

[back to top]

Change From End of Induction Phase in CD4 Cell Count at Week 48 of Maintenance Phase

Change in CD4 Cell Count From End of Induction Phase at Week 48 of Maintenance Phase. Change=Week 48 maintenance Phase value - end of Induction Phase value; a decrease signifies worsening. (NCT00207142)
Timeframe: End of Induction Phase (Week 26 to Week 30 of Induction Phase treatment), Week 48 of Maintenance Phase

Interventioncells/mm3 (Mean)
Switch Regimen100
Continuation Regimen92

[back to top]

Treatment Outcomes Based on Viral Loads (HIV-1 RNA ≥400 c/mL) Through the End of Rescue Phase

Treatment outcome is based on the first reason of failure. These analyses were performed using HIV-1 RNA of 400 c/mL to define suppression and virologic rebound. (NCT00207142)
Timeframe: Baseline, Week 48 of Rescue Phase

InterventionParticipants (Number)
Suppression Maintained Through Week 48Suppression, then Virologic Rebound at/before Wk48Discontinuation Before Week 48Viral Suppression Never Achieved
Rescue Treatment39290

[back to top]

Change From Baseline in HIV-1 RNA at Week 48 of the Rescue Phase

Change From Baseline in HIV-1 RNA at Week 48 of the Rescue Phase. Change=Week 48 Rescue Phase value - Baseline value; a decrease signifies improvement. (NCT00207142)
Timeframe: Baseline, Week 48 of Rescue Phase

Interventionlog10 c/mL (Mean)
Rescue Treatment-3.71

[back to top]

Percentage of Participants With HIV-1 RNA <400 c/mL Through Week 48 of the Maintenance Phase

Participants were considered successes unless they experienced treatment failure, or had missing Week 48 HIV-1 RNA. Treatment failure: virologic rebound (ie, 2 consecutive on-treatment HIV-1 RNA ≥ 400 c/mL, or last HIV-1 RNA ≥ 400 c/mL followed by discontinuation), or discontinuation before Week 48. Denominator included all randomized participants. (NCT00207142)
Timeframe: From the end of Induction Phase (Week 26 to Week 30 of Induction Phase treatment) through Week 48 of Maintenance Phase

InterventionPercentage of participants (Number)
Switch Regimen86
Continuation Regimen81

[back to top]

Percentage of Participants With HIV-1 RNA <50 Copies/mL (c/mL) Through Week 48 of the Maintenance Phase

Participants were considered successes unless they experienced treatment failure, or had missing Week 48 HIV-1 RNA. Treatment failure: virologic rebound (ie, 2 consecutive on-treatment HIV-1 RNA ≥ 50 c/mL, or last HIV-1 RNA ≥ 50 c/mL followed by discontinuation), or discontinuation before Week 48. Denominator included all randomized participants. (NCT00207142)
Timeframe: From the end of Induction Phase (Week 26 to Week 30 of Induction Phase treatment) through Week 48 of Maintenance Phase

InterventionPercentage of participants (Number)
Switch Regimen78
Continuation Regimen75

[back to top]

Kaplan-Meier Cumulative Proportion for Treatment Failure (HIV-1 RNA ≥400 c/mL) at Different Time Points Through Week 48 of the Maintenance Phase

Treatment failure based on HIV-1 RNA ≥ 400 c/mL was defined as virologic rebound on or before Week 48 or discontinuation of study therapy before Week 48 for any reason. Time to treatment failure was analyzed using life tables. Measured Values shows the Kaplan-Meier cumulative proportion of participants without treatment failure up to the end of the respective interval. (NCT00207142)
Timeframe: Weeks 6-8, Weeks 14-16, Weeks 22-24, Weeks 30-32, Weeks 38-40, Weeks 46-48

,
InterventionProportion of participants (Number)
Proportion by Week 6-8Proportion by Week 14-16Proportion by Week 22-24Proportion by Week 30-32Proportion by Week 38-40Proportion by Week 46-48
Continuation Regimen0.98850.98850.97700.95400.89660.8823
Switch Regimen0.94120.91760.90590.88240.85850.8585

[back to top]

Percent Change From End of Induction Phase in Fasting Lipids at Week 48 of Maintenance Phase

Percent change in fasting lipids from end of Induction Phase to Week 48 of Maintenance Phase.Percent changes were calculated on the log scale and then back transformed to the original scale.Change=Week 48 maintenance Phase value - end of Induction Phase value; a decrease signifies worsening for HDL cholesterol and improvement for all other lipds. (NCT00207142)
Timeframe: Measurements were included from the end of Induction Phase (Week 26 to Week 30 of Induction therapy) through Week 48 of Maintenance Phase.

,
Interventionpercent change (Mean)
Total CholesterolHDL CholesterolLDL CholesterolTriglyceridesNon-HDL Cholesterol
Continuation Regimen1.40.8-2.19.81.1
Switch Regimen-4.73.7-0.7-27.0-7.4

[back to top]

Summary of Adverse Events During Induction Phase

Summary of Adverse Events (AEs), Deaths, Serious AEs (SAEs), and AEs leading to study discontinuation. An AE is defined as any new untoward medical occurrence or worsening of a pre-existing medical condition. An SAE is any untoward medical occurrence that at any dose results in death, is life-threatening, requires inpatient hospitalization or causes prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a cancer, is a congenital anomaly/birth defect, results in the development of drug dependency or drug abuse, is an important medical event. (NCT00207142)
Timeframe: Measurements are included through the earlier of the last dose of Induction Phase study therapy plus 30 days or the first dose of Maintenance/Rescue Phase therapy (ie, up until 26 to 31 weeks + 30 days).

,,
InterventionParticipants (Number)
DeathsSAEsSAEs related to study therapyAEs leading to discontinuation
Nonrandomized Subjects3929
Randomized Subjects0700
Total31629

[back to top]

Summary of Adverse Events During Maintenance Phase

Summary of Adverse Events (AEs), Deaths, Serious AEs (SAEs), and AEs leading to study discontinuation. An AE is defined as any new untoward medical occurrence or worsening of a pre-existing medical condition. An SAE is any untoward medical occurrence that at any dose results in death, is life-threatening, requires inpatient hospitalization or causes prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a cancer, is a congenital anomaly/birth defect, results in the development of drug dependency or drug abuse, is an important medical event. (NCT00207142)
Timeframe: Measurements are included from the end of Induction Phase (26 to 30 weeks after first dose) through the last dose of Maintenance Phase study therapy plus 30 days.

,
InterventionParticipants (Number)
DeathsSerious Adverse Events (SAEs)SAEs related to study therapyAEs leading to discontinuation
Continuation Regimen0304
Switch Regimen0401

[back to top]

Summary of Adverse Events During Rescue Phase

Summary of Adverse Events (AEs), Deaths, Serious AEs (SAEs), and AEs leading to study discontinuation. An AE is defined as any new untoward medical occurrence or worsening of a pre-existing medical condition. An SAE is any untoward medical occurrence that at any dose results in death, is life-threatening, requires inpatient hospitalization or causes prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a cancer, is a congenital anomaly/birth defect, results in the development of drug dependency or drug abuse, is an important medical event. (NCT00207142)
Timeframe: Measurements are included from the end of Induction Phase (26 to 30 weeks after the first dose therapy) through the last dose of Rescue Phase study therapy plus 30 days.

InterventionParticipants (Number)
DeathsSAEsSAEs related to study therapyAEs leading to discontinuation
Rescue Treatment1201

[back to top]

Proportion of Patient With Viral Load Less Than 400 Copies/mL

(NCT00242216)
Timeframe: 24 weeks

Interventionpercentage (Number)
Atazanavir89
Fosamprenavir73

[back to top]

CD4 Cell Count Change From Baseline During Treatment.

(NCT00242216)
Timeframe: 24 weeks.

Interventioncell/mm3 (Mean)
Atazanavir139
Fosamprenavir117

[back to top]

Cumulative Treatment Failure Rate of Participants on First Line Antiretroviral Therapy Monitored by Primary Health Care Nurses (Investigative Arm)is Not Inferior to the Cumulative Treatment Failure Rate of Participants Monitored by Doctors (Control Arm).

Cumulative treatment failure is a composite endpoint made up of death, virological failure, toxicity failure and protocol-defined loss to follow-up failure. (NCT00255840)
Timeframe: 96 weeks

InterventionPercentage of participants (Number)
Antiretroviral Therapy Monitored by Medical Officer44
Antiretroviral Therapy Managed by Primary Health Care Nurse48

[back to top]

Percentage of Subjects With Adverse Events of Diarrhea During the First 8 Weeks

(NCT00262522)
Timeframe: Week 8

InterventionPercentage of Subjects (Number)
LPV/r 800/200 mg QD Tablet49.1
LPV/r 800/200 mg QD SGC (Through Week 8)54.8
LPV/r 400/100 mg BID Tablet44.6
LPV/r 400/100 mg BID SGC (Through Week 8)49.7

[back to top]

Mean Change From Baseline to Week 96 in CD4+ T Cell Counts

(NCT00262522)
Timeframe: Week 96 (End of Study)

Interventioncells/microliter (Mean)
LPV/r 800/200 mg QD Tablet238.4
LPV/r 400/100 mg BID Tablet254.0

[back to top]

Percentage of Subjects With Plasma Human Immunodeficiency Virus Type 1 (HIV-1) Ribonucleic Acid (RNA) Levels < 50 Copies/mL at Week 96

(NCT00262522)
Timeframe: Week 96 (End of Study)

InterventionPercentage of Subjects (Number)
LPV/r 800/200 mg QD Tablet64.9
LPV/r 400/100 mg BID Tablet69.2

[back to top]

Percentage of Subjects With Plasma Human Immunodeficiency Virus Type 1 (HIV-1) Ribonucleic Acid (RNA) Levels < 50 Copies/mL at Week 48

(NCT00262522)
Timeframe: Week 48

InterventionPercentage of Subjects (Number)
LPV/r 800/200 mg QD Tablet77.2
LPV/r 400/100 mg BID Tablet75.8

[back to top]

Number of HIV+ Infants

Number of infants with HIV-positive status (NCT00270296)
Timeframe: Throughout study, including breastfeeding, assessed up to 24 months

InterventionInfants (Number)
TZV Arm6
Kaletra Arm1
NVP Arm1

[back to top]

Number of Participants With Virologic Suppression

Suppression of the plasma HIV-1 RNA level to less than 400 copies per milliliter (NCT00270296)
Timeframe: Throughout study, including breastfeeding, assessed up to 24 months

InterventionParticipants (Count of Participants)
TZV Arm274
Kaletra Arm256
NVP Arm160

[back to top]

Median Plasma HIV-1 RNA at Baseline and Weeks 12, 24, 48, 72, 96, 132, and 168

Blood samples of participants were collected for the assessment of plasma HIV-1 RNA. (NCT00296504)
Timeframe: Baseline and Weeks 12, 24, 48, 72, 96, 132, and 168

,,,
Interventionlog 10 copies per milliliter (Median)
Baseline, n=73, 78, 92, 82Week 12, n=69, 78, 92, 79Week 24, n=73, 75, 93, 79Week 48, n=73, 77, 85, 68Week 72, n=65, 73, 80, 53Week 96, n=60, 62, 75, 51Week 132, n=50, 58, 65, 46Week 168, n=44, 48, 20, 39
FPV/RTV BID Population (APV30003)4.061.811.691.691.691.691.691.69
FPV/RTV QD Population (APV30003)3.961.71.691.711.721.691.691.69
PI-Experienced Population (Other Studies)1.812.031.91.71.71.71.71.7
PI-Naïve Population (Other Studies)1.71.71.71.71.71.71.71.7

[back to top]

Median Plasma HIV-1 RNA at Baseline and Weeks 24, 48, 72, 96, 120, 144, 168, 180, 204, and 216

Blood samples of participants were collected for the assessment of plasma HIV-1 RNA. (NCT00296504)
Timeframe: Baseline and Weeks 24, 48, 72, 96, 120, 144, 168, 180, 204, and 216

,,,
Interventionlog 10 copies per milliliter (Median)
Baseline, n=119, 18, 221, 54Week 24, n=119, 17, 217, 51Week 48, n=118, 17, 218, 49Week 72, n=113, 15, 204, 46Week 96, n=106, 15, 192, 43Week 120, n=96, 15, 183, 40Week 144, n=89, 15, 173, 39Week 168, n=81, 14, 160, 38Week 180, n=82, 12, 159, 34Week 204, n=78, 12, 153, 27Week 216, n=80, 0, 143, 4
FPV Population (APV30001)4.821.691.691.691.691.691.691.691.691.691.69
FPV Population (APV30002)4.821.691.691.691.691.691.691.691.691.691.69
NFV Population (APV30001)1.691.691.691.691.691.691.691.691.691.69NA
NFV Population (APV30002)1.781.691.691.691.691.691.691.691.691.691.69

[back to top]

Median Plasma HIV-1 RNA at Weeks 180, 240, 300, 360, 420, and 432

Blood samples of participants were collected for the assessment of plasma HIV-1 RNA. (NCT00296504)
Timeframe: Weeks 180, 240, 300, 360, 420, and 432

Interventionlog 10 copies per milliliters (Median)
Week 180, n=14Week 240, n=101Week 300, n=82Week 360, n=53Week 420, n=52Week 432, n=41
Final Analysis Population (APV30005)1.71.71.71.71.71.7

[back to top]

Median Value of the Total Cholesterol/HDL Ratio at Weeks 120, 180, 204, 216, and 432

Fasting blood samples of participants were collected for the assessment of the total cholesterol/HDL ratio. The ratio of total cholesterol/HDL was calculated by dividing the value of total cholesterol by the value of HDL. (NCT00296504)
Timeframe: Weeks 120, 180, 204, 216, and 432

Interventionratio (Median)
Week 120, n=14Week 180, n=14Week 204, n=106Week 216, n=102Week 432, n=40
Final Analysis Population (APV30005)5.55.53.73.54.0

[back to top]

Number of Participants With Any Adverse Event (AE): Interim Analysis

"An AE is defined as any untoward medical occurrence in a participant or clinical investigation participant administered a pharmaceutical product and which does not necessarily have a causal relationship with this treatment. A list of all adverse events is reported in the Other (Non-Serious) Adverse Events section." (NCT00296504)
Timeframe: Baseline (Day 1) up to 31 January 2006 (up to Week 264)

Interventionparticipants (Number)
FPV Population (APV30001)117
NFV Population (APV30001)16
FPV Population (APV30002)215
NFV Population (APV30002)52
FPV/RTV QD Population (APV30003)72
FPV/RTV BID Population (APV30003)75
PI-Naïve Population (Other Studies)95
PI-Experienced Population (Other Studies)68

[back to top]

Change From Baseline in Aspartate Aminotransferase (AST), Alanine Transaminase (ALT), and Serum Lipase at Weeks 48, 120, 180, 204, and 216

Blood samples of participants were collected for the assessment of AST, ALT, and serum lipase. Change from Baseline at Weeks 48, 120, 180, 204, and 216 was calculated as the value at that particular week minus the value at Baseline (Day 1). (NCT00296504)
Timeframe: Baseline (Day 1) and Weeks 48, 120, 180, 204, and 216

,,,
Interventionunits per liter (U/L) (Median)
AST; Week 48, n=117, 17, 215, 47AST; Week 120, n=96, 15, 182, 40AST; Week 180, n=80, 12, 154, 33AST; Week 204, n=77, 12, 151, 27AST; Week 216, n=79, 0, 142, 4ALT; Week 48, n=117, 17, 216, 47ALT; Week 120, n=96, 15, 182, 40ALT; Week 180, n=81, 12, 155, 33ALT; Week 204, n=77, 12, 152, 27ALT; Week 216, n=79, 0, 142, 4Serum lipase; Week 48, n=118, 17, 217, 47Serum lipase; Week 120, n=95, 15, 184, 40Serum lipase; Week 180, n=81, 12, 156, 33Serum lipase;Week 204, n=78, 12, 153, 27Serum lipase; Week 216, n=80, 0, 143, 4
FPV Population (APV30001)-7-8-10-7-9-5-9-10-10-1044435
FPV Population (APV30002)-7-8-7-7-7-8-8-6-5-60-1110
NFV Population (APV30001)-2023NA0-132NA14-0.50NA
NFV Population (APV30002)01-2-21241-1-3-2212-3

[back to top]

Change From Baseline in Aspartate Aminotransferase (AST), Alanine Transaminase (ALT), and Serum Lipase at Weeks 48, 96, 132, and 168

Blood samples of participants were collected for the assessment of AST, ALT, and serum lipase. Change from Baseline at Weeks 48, 96, 132, and 168 was calculated as the value at that particular week minus the value at Baseline (Day 1). (NCT00296504)
Timeframe: Baseline (Day 1) and Weeks 48, 96, 132, and 168

,,,
Interventionunits per liter (U/L) (Median)
AST; Week 48, n=73, 78, 83, 65AST; Week 96, n=57, 63, 73, 49AST; Week 132, n=50, 57, 64, 46AST; Week 168, n=45, 48, 20, 36ALT; Week 48, n=70, 78, 83, 65ALT; Week 96, n=59, 63, 73, 49ALT; Week 132, n=51, 58, 64, 46ALT; Week 168, n=45, 48, 20, 37Serum lipase; Week 48, n=72, 78, 57, 64Serum lipase; Week 96, n=59, 63, 52, 49Serum lipase; Week 132, n=52, 58, 47, 46Serum lipase; Week 168, n=45, 48, 7, 37
FPV/RTV BID Population (APV30003)-5-4-4-6-6-3-4-52442
FPV/RTV QD Population (APV30003)-6-6-6-6-2-4-2-51320
PI-Experienced Population (Other Studies)0-11-1012-31323
PI-Naïve Population (Other Studies)11222226101-5

[back to top]

Change From Baseline in the Indicated Clinical Chemistry Parameters at Weeks 48, 96, 120, 132, 168, 180, 204, and 216

Fasting blood samples of participants were collected for the assessment of triglycerides (Tri.), cholesterol (Chol.), high density cholesterol (HDL), low density cholesterol (LDL), and fasting blood glucose (FBG). Change from Baseline at Weeks (W) 48, 96, 120, 132, 168, 180, 204, and 216 was calculated as the value at that particular week minus the value at Baseline (Day 1). (NCT00296504)
Timeframe: Baseline (Day 1) and Weeks 48, 96, 120, 132, 168, 180, 204, and 216

,,,,,,,
Interventionmilligrams per deciliter (mg/dl) (Median)
Tri.; W 48, n=54, 14, 159, 38, 61, 65, 77, 59Tri.; W 96, n=92, 13, 132 36, 47, 52, 67, 44Tri.; W 120, n=84, 14, 137, 33, 45, 49, 61, 40Tri.; W 132, n=82, 12, 128, 32, 40, 44, 61, 40Tri.; W 168, n=71, 14, 123, 31, 31, 40, 20, 33Tri.; W 180, n=73, 12, 114, 30, 33, 38, 61, 40Tri.; W 204, n=73, 12, 113, 23, 20, 26, 61, 40Tri.; W 216, n=76, 0, 111, 2, 5, 11, 61, 40Chol.; W 48, n=54, 14, 159, 38, 61, 65, 77, 59Chol.; W 96, n=92, 13, 132, 36, 47, 52, 67, 44Chol.; W 120, n=84, 14, 137, 33, 45, 49, 61, 40Chol.; W 132, n=82, 13, 128, 32, 40, 44, 61, 40Chol.; W 168, n=71, 14, 123, 31, 31, 40, 20, 33Chol.; W 180, n=73, 12, 114, 30, 33, 38, 61, 40Chol.; W 204, n=73, 12, 113, 23, 20, 26, 61, 40Chol.; W 216, n=76, 0, 111, 2, 5, 11, 61, 40HDL Chol.; W 48, n=54, 14, 159, 38, 61, 65, 77 59,HDL Chol.; W 96, n=92, 13, 132, 36, 47, 52, 67, 44HDL Chol.; W 120, n=84, 14, 137, 33, 45, 49, 61,40HDL Chol.; W 132, n=82, 13, 128, 32, 40, 44, 61,40HDL Chol.; W 168, n=71, 14, 123, 31, 31, 40, 0, 0HDL Chol.; W 180, n=73, 12, 114, 30, 33, 38, 61,40HDL Chol.; W 204, n=73, 10, 113, 23, 20, 26, 61,40HDL Chol.; W 216, n=76, 0, 111, 2, 5, 11, 61, 40LDL Chol.; W 48, n=50, 13, 145, 35, 53, 50, 67, 48LDL Chol.; W 96, n=87, 12, 121, 34, 43, 43, 63, 37LDL Chol.; W 120, n=78, 13, 120, 29, 40, 39, 61,40LDL Chol.; W 132, n=78, 12, 117, 29, 36, 37, 58,34LDL Chol.; W 168, n=63, 13, 115, 30, 28, 34, 0, 0LDL Chol.; W 180, n=67, 11, 111, 27, 29, 30, 61,40LDL Chol.; W 204, n=70, 11, 105, 22, 19, 19, 61,40LDL Chol.; W 216, n=69, 0, 105, 2, 4, 10, 61, 40FBG; W 48, n=54, 14, 158, 36, 61, 65, 77, 59FBG; W 96, n=92, 13, 132, 34, 47, 52, 67, 44FBG; W 120, n=84, 14, 136, 31, 44, 48, 61, 40FBG; W 132, n=82, 13, 128, 30, 40, 44, 61, 40FBG; W 168, n=71, 14, 123, 29, 31, 40, 0, 0FBG; W 180, n=73, 12, 112, 28, 33, 37, 61, 40FBG; W 204, n=73, 12, 115, 23, 20, 26, 61, 40FBG; W 216, n=75, 0, 110, 2, 5, 11, 61, 40
FPV Population (APV30001)109152382752394243565452521012121317171616322527253727353374504220
FPV Population (APV30002)5147434727374339534650474753534781212121715171730212124182322213.61.81.8002.71.85.4
FPV/RTV BID Population (APV30003)143422184284966131022-499112822452426-1012-2-64137420-32511
FPV/RTV QD Population (APV30003)916131429134-714121114183-3-656686876323-6-4-3-2-14-3920102200
NFV Population (APV30001)-41-48-33-2-12-41-36NA3-3-9146-45NA-0.285613410NA10-2-411-515NA-20-201-0.94NA
NFV Population (APV30002)2015-4184181298108913211922303479121015401-2452135-4-5-9-4-2-1.802
PI-Experienced Population (Other Studies)-547-73-129-167-170NANA221932122843NANA851016518NANA171538144854NANA0-5-2-140-1NANA
PI-Naïve Population (Other Studies)5-5-465-212NA81313265-5NA345310123NA3730-1-7-45NA0007212021NA

[back to top]

Change From Baseline in the Total Cholesterol/HDL Ratio at Weeks 48, 120, 180, 204, and 216

blood samples of participants were collected for the assessment of the total cholesterol/HDL ratio. The ratio of total cholesterol/HDL was calculated by dividing the value of total cholesterol by the value of HDL. Change from Baseline at Weeks 48, 120, 180, 204, and 216 was calculated as the value at that particular week minus the value at Baseline (Day 1). (NCT00296504)
Timeframe: Baseline (Day 1) and Weeks 48, 120, 180, 204, and 216

,,,
Interventionratio (Median)
Week 48, n=54, 14, 159, 38Week 120, n=84, 14, 137, 33Week 180, n=73, 12, 114, 30Week 204, n=73, 12, 113, 23Week 216, n=76, 0, 111, 2
FPV Population (APV30001)0.1-0.2-0.4-0.3-0.5
FPV Population (APV30002)0.4-0.1-0.3-0.4-0.5
NFV Population (APV30001)-0.4-0.6-0.6-0.6NA
NFV Population (APV30002)0.1-0.2-0.3-0.40.3

[back to top]

Median Values of the Indicated Clinical Chemistry Parameters at Weeks 120, 180, 204, 216, and 432

Fasting blood samples of participants were collected for the assessment of triglycerides, cholesterol, high density cholesterol (HDL), low density cholesterol (LDL), and fasting blood glucose (FBG). (NCT00296504)
Timeframe: Weeks 120, 180, 204, 216, and 432

Interventionmilligrams per deciliter (mg/dl) (Median)
Triglycerides; Week 120, n=14Triglycerides; Week 180, n=14Triglycerides; Week 204, n=106Triglycerides; Week 216, n=102Triglycerides; Week 432, n=40Cholesterol; Week 120, n=14Cholesterol; Week 180, n=14Cholesterol; Week 204, n=106Cholesterol; Week 216, n=102Cholesterol; Week 432, n=40HDL Cholesterol; Week 120, n=14HDL Cholesterol; Week 180, n=14HDL Cholesterol; Week 204, n=106HDL Cholesterol; Week 216, n=102HDL Cholesterol; Week 432, n=40LDL Cholesterol; Week 120, n=14LDL Cholesterol; Week 180, n=14LDL Cholesterol; Week 204, n=106LDL Cholesterol; Week 216, n=102LDL Cholesterol; Week 432, n=40FBG; Week 120, n=14FBG; Week 180, n=14FBG; Week 204, n=106FBG; Week 216, n=104FBG Cholesterol; Week 432, n=40
Final Analysis Population (APV30005)237248137120132202206201205192403652515140405251518688929092

[back to top]

Number of Participants Enrolled in Studies APV30001 and APV300002 With the Indicated HIV-associated Conditions

The number of participants with the indicated HIV-associated conditions were assessed, excluding recurrences. (NCT00296504)
Timeframe: Baseline (Day 1) up to 31 January 2006 (up to Week 264)

,,,
Interventionparticipants (Number)
Candidiasis, oropharyngealHerpes zosterDiarrhea lasting >1 monthCandidiasis, vulvovaginalCervical dysplasiaFever lasting >1 monthHairy leukoplakia, oralPeripheral neuropathyCytomegalovirus retinitisHerpes simplexMycobacterium tuberculosis, any siteEncepathopathy, HIV-relatedKaposis's sarcoma, cutaneousLymphoma,immunoblasticMycobacterium avium complex/ M kansaiiPneumocystis carinii pneumoniaProgressive multifocal leukoencephalopathyToxoplasmosis of brainNon-CDC HIV-associated conditions
FPV Population (APV30001)4011111111000000010
FPV Population (APV30002)7310001201211111108
NFV Population (APV30001)0000000000000000000
NFV Population (APV30002)0320000000100000000

[back to top]

Number of Participants Enrolled in Study APV30003 and Other Studies With the Indicated HIV-associated Conditions

The number of participants with the indicated HIV-associated conditions were assessed. (NCT00296504)
Timeframe: Baseline (Day 1) up to 31 January 2006 (up to Week 264)

,,,
Interventionparticipants (Number)
Candidiasis, oropharyngealHairy leukoplakia, oralPeripheral neuropathyDiarrhea lasting >1 monthHerpes ZosterMycobacterium tuberculosis, any siteOther, Non-CDC HIV associated condition
FPV/RTV BID Population (APV30003)0011000
FPV/RTV QD Population (APV30003)5110111
PI-Experienced Population (Other Studies)0100000
PI-Naïve Population (Other Studies)0000101

[back to top]

Cluster of Differentiation Antigen 4 (CD4+) Cell Count at Baseline and Weeks 48, 120, 168, 180, 204, and 216: Observed Analysis

Blood samples of participants were collected for the assessment of CD4+ cell count. CD4+ cells are white blood cells that are important in fighting infection. HIV infects CD4+ cells, replicates in them, and destroys them. CD4+ cell count provides a measure of the status of the immune system and to what extent it is affected by HIV. (NCT00296504)
Timeframe: Baseline and Weeks 48, 120, 168, 180, 204, and 216

,,,
Interventioncells per millimeters cubed (cells/mm^3) (Median)
Baseline, n=119, 18, 219, 52Week 48, n=117, 17, 211, 49Week 120, n=95, 15, 177, 40Week 168, n=81, 12, 160, 37Week 180, n=78, 9, 153, 33Week 204, n=75, 11, 145, 27Week 216, n=77, 0, 138, 4
FPV Population (APV30001)211426512594541535608
FPV Population (APV30002)167396475506494494534
NFV Population (APV30001)505499613608447563NA
NFV Population (APV30002)297377439422413459548

[back to top]

Number of Participants With HIV-1 Disease Progression to CDC Class C, or New CDC Class C or Death, From Baseline

The number of participants with progression of HIV-1 disease were assessed using the CDC classification of HIV-1: class A, asymptomatic or lymphadenopathy; class B: symptomatic, but not AIDS; class C, AIDS. A participant is considered to have had a disease progression if they report a CDC Class C event for the first time, if they report a new CDC Class C event, or if they experience any fatal adverse event during the study. (NCT00296504)
Timeframe: Baseline (Day 1) up to 31 January 2006 (up to Week 264)

,,,,,,,
Interventionparticipants (Number)
CDC class A to CDC class CCDC class B to CDC class CCDC class C to new CDC class CCDC class A, B, or C to death (on treatment)
FPV Population (APV30001)0310
FPV Population (APV30002)3523
FPV/RTV BID Population (APV30003)1000
FPV/RTV QD Population (APV30003)0100
NFV Population (APV30001)0000
NFV Population (APV30002)0321
PI-Experienced Population (Other Studies)0110
PI-Naïve Population (Other Studies)0000

[back to top]

Percentage of Participants With Plasma HIV-1RNA <400 and <50 Copies Per Milliliter at Baseline and Weeks 12, 24, 48, 60, 96, and 132 (MD=F and Observed)

Blood samples of participants were collected for the assessment of HIV-1RNA copies in plasma. Viral load, measured in RNA copies per milliliter of plasma, is an efficacy measure for antiretroviral drugs. In the MD=F analysis, participants who had missing data at or had discontinued the study prior to a certain time point are classified as non-responders. In the observed analysis (OA), data are presented for the number of participants still enrolled in the study at a certain time point. (NCT00296504)
Timeframe: Baseline and Weeks 12, 24, 48, 60, 96, and 132

,,,
Interventionpercentage of participants (Number)
Baseline <400 copies, n=73, 78, 104, 86; MD=FWeek 12 <400 copies, n=73, 78, 104, 86; MD=FWeek 24 <400 copies, n=73, 78, 104, 86; MD=FWeek 48 <400 copies, n=73, 78, 104, 86; MD=FWeek 60 <400 copies, n=73, 78, 104, 86; MD=FWeek 96 <400 copies, n=73, 78, 104, 86; MD=FWeek 132 <400 copies, n=73, 78, 104, 86; MD=FBaseline <400 copies, n=73, 78, 92, 82; observedWeek 12 <400 copies, n=69, 78, 92, 79; observedWeek 24 <400 copies, n=73, 75, 93, 79; observedWeek 48 <400 copies, n=73, 77, 85, 68; observedWeek 60 <400 copies, n=71, 76, 81, 58; observedWeek 96 <400 copies, n=60, 62, 75, 51; observedWeek 132 <400 copies,n=50, 58, 65, 46; observedBaseline <50 copies, n=73, 78, 104, 86; MD=FWeek 12 <50 copies, n=73, 78, 104, 86; MD=FWeek 24 <50 copies, n=73, 78, 104, 86; MD=FWeek 48 <50 copies, n=73, 78, 104, 86; MD=FWeek 60 <50 copies, n=73, 78, 104, 86; MD=FWeek 96 <50 copies, n=73, 78, 104, 86; MD=FWeek 132 <50 copies, n=73, 78, 104, 86; MD=FBaseline <50 copies, n=73, 78, 92, 82; observedWeek 12 <50 copies, n=69, 78, 92, 79; observedWeek 24 <50 copies, n=73, 75, 93, 79; observedWeek 48 <50 copies, n=73, 77, 85, 68; observedWeek 60 <50 copies, n=71, 76, 81, 58; observedWeek 96 <50 copies, n=60, 62, 75, 51; observedWeek 132 <50 copies, n=50, 58, 65, 46; observed
FPV/RTV BID Population (APV30003)1827978776260182837979778104764605951450476761616560
FPV/RTV QD Population (APV30003)0757971705849080797172707204763495145370496349525554
PI-Experienced Population (Other Studies)59526050514541625766637676764440424241374046434653606374
PI-Naïve Population (Other Studies)84828578746760959295959593955978786968615366888785888485

[back to top]

Percentage of Participants With Plasma HIV-1RNA <50 Copies Per Milliliter at Baseline and Weeks 120, 180, 240, 300, 360, 420, and 432 (Observed)

Blood samples of participants were collected for the assessment of HIV-1RNA copies in plasma. Viral load, measured in RNA copies per milliliter of plasma,is an efficacy measure for antiretroviral drugs. (NCT00296504)
Timeframe: Baseline and Weeks 120, 180, 240, 300, 360, 420, and 432

Interventionpercentage of participants (Number)
Week 120 <50 copies, n=14; observedWeek 180 <50 copies, n=14; observedWeek 240 <50 copies, n=101; observedWeek 300 <50 copies, n=82; observedWeek 360 <50 copies, n=53; observedWeek 420 <50 copies, n=52; observedWeek 432 <50 copies, n=41; observed
Final Analysis Population (APV30005)93868793899293

[back to top]

Cluster of Differentiation Antigen 4 (CD4+) Cell Count at Baseline and Weeks 24, 48, 96, 132, and 168: Observed Analysis

Blood samples of participants were collected for the assessment of CD4+ cell count. CD4+ cells are white blood cells that are important in fighting infection. HIV infects CD4+ cells, replicates in them, and destroys them. CD4+ cell count provides a measure of the status of the immune system and to what extent it is affected by HIV. (NCT00296504)
Timeframe: Baseline and Weeks 24, 48, 96, 132, and 168

,,,
Interventioncells per millimeters cubed (cells/mm^3) (Median)
Baseline, n=72, 78, 94, 76Week 24, n=71, 73, 92, 75Week 48, n=73, 77, 85, 68Week 96, n=58, 62, 73, 50Week 132, n=50, 56, 64, 47Week 168, n=40, 47, 20, 39
FPV/RTV BID Population (APV30003)304348386417386409
FPV/RTV QD Population (APV30003)311367374409395513
PI-Experienced Population (Other Studies)361399418463486460
PI-Naïve Population (Other Studies)477520582567642619

[back to top]

Number of Participants With Any Adverse Event (AE): Final Analysis

"An AE is defined as any untoward medical occurrence in a participant or clinical investigation participant administered a pharmaceutical product and which does not necessarily have a causal relationship with this treatment. A list of all adverse events is reported in the Other (Non-Serious) Adverse Events section." (NCT00296504)
Timeframe: Post January 2006; for up to 241 weeks

Interventionparticipants (Number)
Final Analysis Population (APV30005)95

[back to top]

Median Aspartate Aminotransferase (AST), Alanine Transaminase (ALT), and Serum Lipase Values at Weeks 120, 180, 204, 216, and 432

Blood samples of participants were collected for the assessment of AST, ALT, and serum lipase. (NCT00296504)
Timeframe: Weeks 120, 180, 204, 216, and 432

Interventionunits per liter (U/L) (Median)
AST; Week 120, n=14AST; Week 180, n=14AST; Week 204, n=107AST; Week 216, n=106AST; Week 432, n=41ALT; Week 120, n=14ALT; Week 180, n=14ALT; Week 204, n=107ALT; Week 216, n=106ALT; Week 432, n=41Serum lipase;Week 120, n=14Serum lipase; Week 180, n=14Serum lipase; Week 204, n=107Serum lipase; Week 216, n=106Serum lipase; Week 432, n=41
Final Analysis Population (APV30005)182120212118321919194739343636

[back to top]

Change From Baseline in the Total Cholesterol/HDL Ratio at Weeks 48, 96, 132, and 168

Fasting blood samples of participants were collected for the assessment of the total cholesterol/HDL ratio. The ratio of total cholesterol/HDL was calculated by dividing the value of total cholesterol by the value of HDL. Change from Baseline at Weeks 48, 96, 132, and 168 was calculated as the value at that particular week minus the value at Baseline (Day 1). (NCT00296504)
Timeframe: Baseline (Day 1) and Weeks 48, 96, 132, and 168

,,,
Interventionratio (Median)
Week 48, n=61, 65 , 45, 7Week 96, n=47, 52, 42, 5Week 132, n=40, 44, 41, 4Week 168, n=31, 40, 3, 5
FPV/RTV BID Population (APV30003)-0.30.1-0.60.2
FPV/RTV QD Population (APV30003)-0.4-0.5-0.4-0.3
PI-Experienced Population (Other Studies)-0.8-0.3-1.5-1.4
PI-Naïve Population (Other Studies)-0.1-0.1-0.2-0.6

[back to top]

Percentage of Participants With Plasma HIV-1 Ribonucleic Acid (RNA) <400 and <50 Copies Per Milliliter at Baseline and Weeks 48, 120, 180, and 216 (MD=F and Observed)

Blood samples of participants were collected for the assessment of HIV-1RNA copies in plasma. Viral load, measured in RNA copies per milliliter of plasma, is an efficacy measure for antiretroviral drugs. In the MD=F analysis, participants who had missing data at or had discontinued the study prior to a certain time point are classified as non-responders. In the observed analysis (OA), data are presented for the number of participants still enrolled in the study at a certain time point. Participants in the NFV populations had received antiretroviral therapy prior to Baseline. (NCT00296504)
Timeframe: Baseline and Weeks 48, 120, 180, and 216

,,
Interventionpercentage of participants (Number)
Baseline <400 copies, n=119, 219, 54; MD=FWeek 48 <400 copies, n=119, 219, 54; MD=FWeek 120 <400 copies, n=119, 219, 54; MD=FWeek 180 <400 copies, n=119, 219, 54; MD=FWeek 216 <400 copies, n=119, 219, 0; MD=FBaseline <400 copies, n=119, 219, 54; observedWeek 48 <400 copies, n=118, 216, 49; observedWeek 120 <400 copies, n=96, 181, 40; observedWeek 180 <400 copies, n=82, 158, 34; observedWeek 216 <400 copies, n=80, 142, 0; observedBaseline <50 copies, n=119, 219, 54; MD=FWeek 48 <50 copies, n=119, 219, 54; MD=FWeek 120 <50 copies, n=119, 219, 54; MD=FWeek 180 <50 copies, n=119, 219, 54; MD=FWeek 216 <50 copies, n=119, 219, 0; MD=FBaseline <50 copies, n=119, 219, 54; observedWeek 48 <50 copies, n=118, 216, 49; observedWeek 120 <50 copies, n=96, 181, 40; observedWeek 180 <50 copies, n=82, 158, 34; observedWeek 216 <50 copies, n=80, 142, 0; observed
FPV Population (APV30001)192766662193959593180716158181888886
FPV Population (APV30002)094776861095939494076666358077808889
NFV Population (APV30002)61856954NA61949385NA46596148NA46658376NA

[back to top]

Change in CD4 Percent From Entry to Week 48

Change was calculated as CD4 percent at week 48 minus entry CD4 percent (last CD4 percent before randomization date). Only subjects who reached 48 weeks of follow-up before DSMB decisions to unblind each Cohort were included in summary. (NCT00307151)
Timeframe: 48 weeks if before date of DSMB decision to unblind Cohort results (Coh I: April 20, 2009; Coh II: October 27, 2010)

InterventionPercent of CD4 (Mean)
Coh I: NVP13.9
Coh I: LPV/r12.0
Coh II: NVP15.2
Coh II: LPV/r14.3

[back to top]

Percent of Participants Experiencing Virologic Failure

Virologic failure is defined as a confirmed plasma HIV-1 RNA level that is <1 log10 copies/mL below the study entry value at 12 to 24 weeks after treatment is initiated OR a confirmed plasma HIV-1 RNA level >400 copies/mL at 24 weeks OR death on or before 24 weeks. Results report percent of participants reaching a virologic failure endpoint by week 24 calculated using the Kaplan-Meier method. (NCT00307151)
Timeframe: Earlier of 24 weeks or date of DSMB decision to unblind Cohort results (Coh I: April 20, 2009; Coh II: October 27, 2010)

InterventionPercent of participants (Number)
Coh I: NVP27.4
Coh I: LPV/r10.4
Coh II: NVP28.6
Coh II: LPV/r12.9

[back to top]

Time From Start of Study Treatment to First New Grade >=3 Lab Abnormality, Sign or Symptom Occurring on Study Treatment

Safety events include lab abnormalities, signs or symptoms of grade 3 or higher. Events were graded according to the Division of AIDS Table for Grading Severity of Adult and Pediatric Adverse Events, Version 1.0. Events defined as new if first occurrence was after initiation of study treatment or if severity increased from entry and while on the NNRTI or PI component of study treatment. (NCT00307151)
Timeframe: On randomized NNRTI or PI component of study treatment and until date of DSMB decision to unblind Cohort results (Coh I: April 20, 2009; Coh II: October 27, 2010)

,,,
InterventionWeeks (Number)
10th percentile25th percentile
Coh I: LPV/r836
Coh I: NVP424
Coh II: LPV/r412
Coh II: NVP34

[back to top]

Time From Randomization to Virologic Failure

Virologic failure is defined as the earlier of a confirmed plasma HIV-1 RNA level that is <1 log10 copies/mL below the study entry value at 12 to 24 weeks after treatment is initiated OR a confirmed plasma HIV-1 RNA level >400 copies/mL at 24 weeks OR a confirmed viral rebound >4000 copies/mL after week 24 OR death. (NCT00307151)
Timeframe: Until date of DSMB decision to unblind Cohort results (Coh I: April 20, 2009 - median follow-up 48 weeks and range 0 - 125 weeks; Coh II: October 27, 2010 - median follow-up 72 weeks and range from 0 to 204 weeks)

,,,
InterventionWeeks (Number)
5th percentile10th percentile
Coh I: LPV/r1624
Coh I: NVP1212
Coh II: LPV/r1624
Coh II: NVP1216

[back to top]

Time From Randomization to Treatment Failure, Defined as Virologic Failure or Permanent Discontinuation of the Randomized NNRTI or PI Component of Study Treatment

Treatment failure is defined as a confirmed plasma HIV-1 RNA level that is <1 log10 copies/mL below the study entry value at 12 to 24 weeks after treatment is initiated OR a confirmed plasma HIV-1 RNA level >400 copies/mL at 24 weeks OR a confirmed viral rebound >4000 copies/mL after week 24 OR permanent discontinuation of the randomized NNRTI or PI component of study treatment for any reason including death. (NCT00307151)
Timeframe: Until date of DSMB decision to unblind Cohort results (Coh I: April 20, 2009 - median follow-up 48 weeks and range 0 - 125 weeks; Coh II: October 27, 2010 - median follow-up 72 weeks and range from 0 to 204 weeks)

,,,
InterventionWeeks (Number)
10th percentile25th percentile
Coh I: LPV/r436
Coh I: NVP1216
Coh II: LPV/r1436
Coh II: NVP416

[back to top] [back to top]

Time From Randomization to Death

Results report 2nd percentile of time from randomization to death (NCT00307151)
Timeframe: Until date of DSMB decision to unblind Cohort results (Coh I: April 20, 2009 - median follow-up 48 weeks and range 0 - 125 weeks; Coh II: October 27, 2010 - median follow-up 72 weeks and range from 0 to 204 weeks)

InterventionWeeks (Number)
Coh I: NVP11
Coh I: LPV/r3
Coh II: NVP2
Coh II: LPV/r83

[back to top]

Percent of Participants With Treatment Failure, Defined as a Confirmed Virologic Failure or Permanent Discontinuation of the Randomized NNRTI or PI Component of Study Treatment

Treatment failure is defined as a confirmed plasma HIV-1 RNA level that is <1 log10 copies/mL below the study entry value at 12 to 24 weeks after treatment is initiated OR a confirmed plasma HIV-1 RNA level >400 copies/mL at 24 weeks OR permanent discontinuation of the randomized NNRTI or PI component of study treatment at or prior to 24 weeks of treatment for any reason including death. Results report percent of participants reaching a treatment failure endpoint by week 24 calculated using the Kaplan-Meier method. (NCT00307151)
Timeframe: Earlier of 24 weeks or date of DSMB decision to unblind Cohort results (Coh I: April 20, 2009; Coh II: October 27, 2010)

InterventionPercent of participants (Number)
Coh I: NVP39.6
Coh I: LPV/r21.7
Coh II: NVP40.8
Coh II: LPV/r19.3

[back to top]

Number of Participants Developing New NRTI, NNRTI or PI-resistant Virus

Numbers of participants developing new NRTI, NNRTI or PI-resistant virus after reaching a virologic failure endpoint (NCT00307151)
Timeframe: Until date of DSMB decision to unblind Cohort results (Coh I: April 20, 2009 - median follow-up 48 weeks and range 0 - 125 weeks; Coh II: October 27, 2010 - median follow-up 72 weeks and range from 0 to 204 weeks)

Interventionparticipants (Number)
Coh I: NVP16
Coh I: LPV/r1
Coh II: NVP10
Coh II: LPV/r4

[back to top]

Infant HIV Status

The neonatal HIV-1 status are assessed by the Roche Amplicor HIV-1 DNA Assay Version 1.5 (Roche Molecular Systems). (NCT00326716)
Timeframe: Birth Through 6 Months on Study

,
InterventionParticipants (Number)
HIV PositiveHIV Negative
Infant ATV 300 mg / RTV 100 mg020
Infant ATV 400 mg / RTV 100 mg020

[back to top]

Infant Gender

(NCT00326716)
Timeframe: At the time of delivery

,
InterventionParticipants (Number)
MaleFemale
Mothers ATV 300 mg / 100 mg Third Trimester128
Mothers ATV 400 mg / 100 mg Third Trimester911

[back to top]

Median Change From Baseline to Day of Delivery in Maternal Cluster of Differentiation 4 (CD4) Cell Count

The median CD4 cell count change from baseline was calculated for all treated mothers at the time of delivery ± 2 days. Maternal CD4 cell counts were assessed by the Roche Amplicor® Ultrasensitive Assay Version 1.5. (NCT00326716)
Timeframe: Baseline, Day of Delivery ± 2 Days

Interventioncells / mm^3 (Median)
Mother ATV 300 mg / RTV 100 mg89
Mother ATV 400 mg / RTV 100 mg174

[back to top]

Mean RTV Trough Plasma Concentration (Cmin) 24 Hours Following the Daily Dose

Cmin = plasma concentration 24 hours post dose of ritonavir at specified time points. (NCT00326716)
Timeframe: Pregnancy Weeks 12 to 28, 28 to 36, and 4-6 Weeks Postpartum at 24 hours following the daily dose.

Interventionng•h / mL (Geometric Mean)
Mothers ATV 300 mg / RTV 100 mg Second Trimester50.10
Mothers ATV 300 mg / RTV 100 mg Third Trimester41.12
Mothers ATV 400 mg / RTV 100 mg Third Trimester38.05

[back to top]

Mean RTV Time of Maximum Observed Plasma Concentration (Tmax)

Tmax = time to reach the maximum observed plasma concentration of ritonavir at specified time points. (NCT00326716)
Timeframe: Pregnancy Weeks 12 to 28, Weeks 28 to 36, and 4-6 Weeks Postpartum

InterventionHours (Geometric Mean)
Mothers ATV 300 mg / 100 mg Second Trimester6.11
Mothers ATV 300 mg / 100 mg Third Trimester4.15
Mothers ATV 400 mg / 100 mg Third Trimester4.63

[back to top]

Mean RTV Terminal Elimination Half Life (T 1/2)

T 1/2 = terminal elimination half life of ritonavir at specified time points. (NCT00326716)
Timeframe: Pregnancy Weeks 12 to 28, 28 to 36, and 4-6 Weeks Postpartum

InterventionHours (Geometric Mean)
Mothers ATV 300 mg / 100 mg Second Trimester5.03
Mothers ATV 300 mg / 100 mg Third Trimester5.28
Mothers ATV 400 mg / 100 mg Third Trimester5.10

[back to top]

Mean RTV Maximum Plasma Concentration (Cmax) in One Dosing Interval

Cmax = maximum observed plasma concentration of ritonavir at specified time points. (NCT00326716)
Timeframe: Pregnancy Weeks 12 to 28, 28 to 36, and 4-6 Weeks Postpartum

Interventionng / mL (Geometric Mean)
Mothers ATV 300 mg / RTV 100 mg Second Trimester530.81
Mothers ATV 300 mg / RTV 100 mg Third Trimester587.36
Mothers ATV 400 mg / RTV 100 mg Third Trimester524.48

[back to top]

Mean RTV Area Under the Concentration Curve (AUC TAU)

AUC = area under the concentration curve (AUC [TAU]) of ritonavir in one dosing interval. (NCT00326716)
Timeframe: Pregnancy Weeks 12 to 28, 28 to 36, and 4-6 Weeks Postpartum

Interventionng•h / mL (Geometric Mean)
Mothers ATV 300 mg / RTV 100 mg Second Trimester4,500.03
Mothers ATV 300 mg / RTV 100 mg Third Trimester4,664.93
Mothers ATV 400 mg / RTV 100 mg Third Trimester4,383.30

[back to top]

Mean HIV RNA Level at Baseline

(NCT00326716)
Timeframe: Baseline

Interventionlog10 cm / mL (Mean)
Mothers ATV 300 mg / RTV 100 mg Third Trimester3.520
Mothers ATV 400 mg / RTV 100 mg Third Trimester4.020

[back to top]

Mean CD4 Cell Count at Baseline

(NCT00326716)
Timeframe: Baseline

Interventioncells / mm^3 (Mean)
Mothers ATV 300 mg / RTV 100 mg Third Trimester435.0
Mothers ATV 400 mg / RTV 100 mg Third Trimester390.0

[back to top]

Mean ATV Trough Plasma Concentration (Cmin) 24 Hours Following the Daily Dose

Cmin = plasma concentration 24 hours post dose of atazanavir at specified time points. (NCT00326716)
Timeframe: Pregnancy Weeks 12 to 28, 28 to 36, and 4-6 Weeks Postpartum at 24 hours following the daily dose.

Interventionng•h / mL (Geometric Mean)
Mothers ATV 300 mg / RTV 100 mg Second Trimester663.78
Mothers ATV 300 mg / RTV 100 mg Third Trimester668.48
Mothers ATV 400 mg / RTV 100 mg Third Trimester916.63

[back to top]

Mean ATV Time of Maximum Observed Plasma Concentration (Tmax)

Tmax = time to reach maximum observed plasma concentration of atazanavir at specified time points. (NCT00326716)
Timeframe: Pregnancy Weeks 12 to 28, 28 to 36, and 4-6 Weeks Postpartum

InterventionHours (Geometric Mean)
Mothers ATV 300 mg / 100 mg Second Trimester3.68
Mothers ATV 300 mg / 100 mg Third Trimester2.94
Mothers ATV 400 mg / 100 mg Third Trimester3.30

[back to top]

Mean ATV Terminal Elimination Half Life (T 1/2)

T 1/2 = terminal elimination half life of atazanavir at specified time points. (NCT00326716)
Timeframe: Pregnancy Weeks 12 to 28, 28 to 36, and 4-6 Weeks Postpartum

InterventionHours (Geometric Mean)
Mothers ATV 300 mg / 100 mg Second Trimester10.42
Mothers ATV 300 mg / 100 mg Third Trimester12.10
Mothers ATV 400 mg / 100 mg Third Trimester12.17

[back to top]

Mean ATV Maximum Plasma Concentration (Cmax) in One Dosing Interval

Cmax = maximum observed plasma concentration of atazanavir at specified time points. (NCT00326716)
Timeframe: Pregnancy Weeks 12 to 28, 28 to 36, and 4-6 Weeks Postpartum

Interventionng / mL (Geometric Mean)
Mothers ATV 300 mg / RTV 100 mg Second Trimester3,729.09
Mothers ATV 300 mg / RTV 100 mg Third Trimester3,291.46
Mothers ATV 400 mg / RTV 100 mg Third Trimester4,210.76

[back to top]

Mean ATV Area Under the Concentration Curve (AUC TAU)

AUC = area under the concentration curve (AUC [TAU]) of atazanavir in one dosing interval from time zero to 24 hours. (NCT00326716)
Timeframe: Pregnancy Weeks 12 to 28, 28 to 36, and 4-6 Weeks Postpartum

Interventionng•h / mL (Geometric Mean)
Mothers ATV 300 mg / RTV 100 mg Second Trimester34,399.13
Mothers ATV 300 mg / RTV 100 mg Third Trimester34,251.50
Mothers ATV 400 mg / RTV 100 mg Third Trimester46,602.45

[back to top]

Infant Gestational Age at Delivery

(NCT00326716)
Timeframe: At the time of delivery

InterventionWeeks (Mean)
Mothers ATV 300 mg / 100 mg Third Trimester38
Mothers ATV 400 mg / 100 mg Third Trimester38

[back to top]

Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)

AE=any new untoward medical occurrence or worsening of a pre-existing medical condition which does not necessarily have a causal relationship with this treatment. SAE =any untoward medical occurrence that at any dose: results in death, is life-threatening, requires inpatient hospitalization or causes prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect, results in development of drug dependency or drug abuse, is an important medical event. (NCT00326716)
Timeframe: During study period and 30 days post-study.

,
InterventionParticipants (Number)
Death (n=41, n=40)Serious Adverse Event (n=41, n=40)Total AEs Leading to Discontinuation (n=41, n=40)Anemia Leading to Discontinuation (n=41, n=40)Transaminitis Discontinuation (n=41, n=40)Prematurity Causing Discontinuation (n=41, n=40)All AEs (n=41, n=40)Anemia (n=41, n=40)Diarrhea (n=41, n=40)Nausea (n=41, n=40)Vomiting (n=41, n=40)Jaundice (n=41, n=40)Hyperbilirubinemia (n=41, n=40)Ocular Icterus (n=41, n=40)Skin / subcutaneous tissue disorders (n=41, n=40)
All Infants0142201403405201020
All Treated Mothers016211NA406467101310

[back to top]

Number of Participants With Grade 2 to Grade 4 AEs and SAEs

AEs and SAEs considered possibly, probably, or certainly related to study treatment, were graded according to Common Terminology Criteria for Adverse Events (CTCAE) Version 3.0 (Grade 1=Mild, Grade 2=Moderate, Grade 3=Severe, Grade 4=Life-threatening or disabling, Grade 5=Death). Hyperbilirubinemia (Grade 1=1.1 to 1.5 upper limit of normal [ULN] [mild], Grade 2=1.6 to 2.5 ULN [moderate], Grade 3=2.6 to 5.0 ULN [severe], Grade 4= > 5.0 ULN [potentially life threatening]). (NCT00326716)
Timeframe: During Study Period and 30 Days Post-Study.

,
InterventionParticipants (Number)
Grade 2 to Grade 4 (n=41, n=40)Related Grade 2 to Grade 4 (n=41, n=40)Grade 3 to Grade 4 (n=41, n=40)Grade 3 to Grade 4 Total Bilirubin (n=41, n=40)
All Infants19087
All Treated Mothers32101219

[back to top]

SAEs in Enrolled Infants

SAEs were evaluated for all treated and untreated participants. An SAE was defined as an untoward medical occurrence that results in death, is life-threatening (defined as an event in which the participant was at risk of death at the time of the event); might have caused death if it were more severe, required inpatient hospitalization or prolongation of existing hospitalization, in persistent or significant disability/incapacity, is a congenital anomaly/birth defect, an important medical event that required intervention to prevent serious outcomes. (NCT00326716)
Timeframe: Birth Through Week 16 of Life

,
InterventionParticipants (Number)
Any Adverse ExperienceBlood and Lymphatic System DisordersAnemiaCardiac DisordersCardio-Respiratory ArrestRestrictive CardiomyopathyCongenital, Familial, and Genetic DisordersAtrial Septal DefectGastrointestinal DisordersConstipationVomitingHepatobiliary DisordersHyperbilirubinemiaJaundiceInfections and InfestationsBronchiolitisGastroenteritisMeningitisPneumoniaSepsisSyphilisInjury, Poisoning, and Procedural ComplicationsOverdoseMetabolism and Nutrition DisordersHyperkalemiaHypoglycemiaNervous System DisordersCerebral IschemiaConvulsionPregnancy, Puerperium, and Perinatal ConditionsPremature BabyRespiratory, Thoracic, and Mediastinal DisordersNeonatal Respiratory Distress SyndromeRespiratory Distress
Infant ATV 300 mg / RTV 100 mg10111110011110140110111111011111211
Infant ATV 400 mg / RTV 100 mg4110001100011022001000010100011000

[back to top]

SAEs in Enrolled Mothers

SAEs were evaluated for all treated and untreated participants. An SAE was defined as an untoward medical occurrence that results in death, is life-threatening (defined as an event in which the participant was at risk of death at the time of the event); might have caused death if it were more severe, required inpatient hospitalization or prolongation of existing hospitalization, in persistent or significant disability/incapacity, is a congenital anomaly/birth defect, an important medical event that required intervention to prevent serious outcomes. (NCT00326716)
Timeframe: During Study Period and 30 Days Post-Study.

,
InterventionParticipants (Number)
Any Adverse ExperienceBlood and Lymphatic System DisordersAnemiaCardiac DisordersCardiomyopathySinus ArrhythmiaGastrointestinal DisordersAbdominal HerniaHepatobiliary DisordersHyperbilirubinemiaInfections and InfestationsEndometrial DecidualPneumoniaSepsisInvestigationsTransaminases IncreasedPregnancy, Puerperium, and Perinatal ConditionsAmenorrheaPre-eclampsiaPregnancy Induced HypertensionPremature Rupture of MembranesVascular DisordersHypertensionHemorrhage
Mother ATV 300 mg / RTV 100 mg711110000022000020101101
Mother ATV 400 mg / RTV 100 mg822101111120111121100110

[back to top]

Maternal HIV Ribonucleic Acid (RNA) Level on Day of Delivery

The maternal HIV RNA level is assessed by the Roche Amplicor® Ultrasensitive Assay Version 1.5. (NCT00326716)
Timeframe: Day of Delivery ± 2 Days

,
InterventionParticipants (Number)
HIV RNA < 400 copies/mLHIV RNA < 50 copies/mL
Mothers ATV 300 mg / RTV 100 mg1919
Mothers ATV 400 mg / RTV 100 mg2019

[back to top]

Median Change From Baseline to Day of Delivery in Maternal HIV RNA Level

The maternal HIV RNA level was determined at baseline and the day of delivery ± 2 days using VR-OC. The maternal HIV RNA level is assessed by the Roche Amplicor® Ultrasensitive Assay Version 1.5. (NCT00326716)
Timeframe: Baseline, Day of Delivery ± 2 Days

Interventionlog10 c / mL (Median)
Mother ATV 300 mg / RTV 100 mg-1.8
Mother ATV 400 mg / RTV 100 mg-2.37

[back to top]

Mean Atazanavir Maternal Plasma Concentration and Neonatal Cord Blood Concentration

Mean atazanavir maternal plasma concentration and neonatal cord blood concentration as measured at the time of delivery. (NCT00326716)
Timeframe: At Time of Delivery

,
Interventionng / mL (Mean)
Maternal Serum ConcentrationCord Blood Concentration
Mothers ATV 300 mg / RTV 100 mg1,412.05273.20
Mothers ATV 400 mg / RTV 100 mg1,568.06231.49

[back to top]

Mean Atazanavir Plasma Protein Binding

Atazanavir Plasma Protein Binding Percentage measured at specified time points. (NCT00326716)
Timeframe: Pregnancy Weeks 28 to Delivery at 3 Hours Postdose and 24 Hours Postdose, and Time of Delivery

,
InterventionPercentage Bound (Mean)
Third Trimester 3 Hours Post Dose (n = 20, 20)Third Trimester 24 Hours Post Dose (n = 19, 20)Time of Delivery (n = 15, 12)
Mother ATV 300 mg / RTV 100 mg91.3490.3777.05
Mother ATV 400 mg / RTV 100 mg87.7088.8975.62

[back to top]

Median Infant Total Bilirubin Level

Median infant total bilirubin level as measured at specified time points. (NCT00326716)
Timeframe: Birth (Day 1), Day 3, Day 5, and Day 7 of Life

,
Interventionmg / dL (Median)
Day 1 (Birth)Day 3Day 5Day 7
Infant ATV 300 mg / RTV 100 mg3.208.407.105.10
Infant ATV 400 mg / RTV 100 mg3.259.209.257.30

[back to top]

Multicenter AIDS Cohort Study (MACS) Participant Adherence to Regimen and Drug Components for ATV 300 mg / RTV 100 mg Test Dose

The MACS was administered to evaluate participant adherence to each drug and the adherence to the regimen. The MACS adherence questionnaire asks patients how many medication doses they missed during the previous day, 2 days, 3 days and 4 days. Drug-specific questions included adherence with dose and frequency. Adherence was defined as taking all doses and numbers of pills as prescribed for each medication. This strict adherence cut-off was based on the guidelines stating that anything less than excellent adherence may result in a virus breakthrough and development of resistance. (NCT00326716)
Timeframe: Study Week 2, Pregnancy Weeks 20 to Weeks 28, Pregnancy Weeks 28 to Delivery, Week 2 Postpartum, Week 4 Postpartum

,
InterventionParticipants (Number)
Study Week 2 Regimen (n = 20,18)Study Week 2 ATV (n = 20,18)Study Week 2 RTV (n = 20,18)Study Week 2 ZDV/3TC (n = 20,18)Visit 1 Pregnancy Week 20 to 28 Regimen (n=12, 9)Visit 1 Pregnancy Week 20 to 28 ATV (n=12, 9)Visit 1 Pregnancy Week 20 to 28 RTV (n=12, 9)Visit 1 Pregnancy Week 20 to 28 ZDV/3TC (n=12, 9)Visit 2 Pregnancy Week 20 to 28 Regimen (n=8, 5)Visit 2 Pregnancy Week 20 to 28 ATV (n=8, 5)Visit 2 Pregnancy Week 20 to 28 RTV (n=8, 5)Visit 2 Pregnancy Week 20 to 28 ZDV/3TC (n=8, 5)Visit 3 Pregnancy Week 20 to 28 Regimen (n=6, 2)Visit 3 Pregnancy Week 20 to 28 ATV (n=6, 2)Visit 3 Pregnancy Week 20 to 28 RTV (n=6, 2)Visit 3 Pregnancy Week 20 to 28 ADV/3TC (n=6, 2)Visit 4 Pregnancy Wk 20 to 28 Regimen (n=0, 13)Visit 4 Pregnancy Week 20 to 28 ATV (n=0, 13)Visit 4 Pregnancy Week 20 to 28 RTV (n=0, 13)Visit 4 Pregnancy Wk 20 to 28 ZDV/3TC (n=0, 13)Visit 1 Pregnancy Wk 20 to Birth Regimen (n=20,20)Visit 1 Pregnancy Week 20 to Birth ATV (n=20,20)Visit 1 Pregnancy Week 20 to Birth RTV (n=20,20)Visit 1 Pregnancy Wk 20 to Birth ZDV/3TC (n=20,20)Visit 2 Pregnancy Wk 20 to Birth Regimen (n=19,19)Visit 2 Pregnancy Week 20 to Birth ATV (n=19,19)Visit 2 Pregnancy Week 20 to Birth RTV (n=19,19)Visit 2 Pregnancy Wk 20 to Birth ZDV/3TC (n=19,19)Visit 3 Pregnancy Wk 20 to Birth Regimen (n=15,19)Visit 3 Pregnancy Week 20 to Birth ATV (n=15,19)Visit 3 Pregnancy Week 20 to Birth RTV (n=15,19)Visit 3 Pregnancy Wk 20 to Birth ZDV/3TC (n=15,19)Visit 4 Pregnancy Wk 28 to Birth Regimen (n=5,15)Visit 4 Pregnancy Week 28 to Birth ATV (n=5,15)Visit 4 Pregnancy Week 28 to Birth RTV (n=5,15)Visit 4 Pregnancy Wk 28 to Birth ZDV/3TC (n=5,15)Visit 5 Pregnancy Wk 28 to Birth Regimen (n=1, 2)Visit 5 Pregnancy Week 28 to Birth ATV (n=1, 2)Visit 5 Pregnancy Week 28 to Birth RTV (n=1, 2)Visit 5 Pregnancy Wk 28 to Birth ZDV/3TC (n=1, 2)Visit 6 Pregnancy Wk 28 to Birth Regimen (n=0, 1)Visit 6 Pregnancy Week 28 to Birth ATV (n=0, 1)Visit 6 Pregnancy Week 28 to Birth RTV (n=0, 1)Visit 6 Pregnancy Wk 28 to Birth ZDV/3TC (n=0, 1)Postpartum Week 2 Regimen (n=19, 19)Postpartum Week 2 ATV (n=18, 19)Postpartum Week 2 RTV (n=18, 19)Postpartum Week 2 ZDV/3TC (n =19, 19)Postpartum Week 4 Regimen (n=17, 19)Postpartum Week 4 ATV (n=17, 19)Postpartum Week 4 RTV (n=17, 19)Postpartum Week 4 ZDV/3TC (n=17, 19)
Mother ATV 300 mg / RTV 100 mg202020201112121138744554NANANANA19202019181919181213131255551111NANANANA1416161416171617
Mother ATV 400 mg / RTV 100 mg171717178899354422221313131320202020181919181819191815151515222211111818181818191918

[back to top]

Infant Race

(NCT00326716)
Timeframe: At the time of delivery

,
InterventionParticipants (Number)
BlackWhiteMixed Race
Mothers ATV 300 mg / 100 mg Third Trimester1550
Mothers ATV 400 mg / 100 mg Third Trimester1811

[back to top]

Time-weighted Mean Change From Baseline Plasma HIV-RNA.

(NCT00335322)
Timeframe: 48 weeks

Interventionlog copies/mL (Mean)
TDF/FTC+EFV-2.59
TDF/FTC+r/ATV-2.69
TDF/FTC+AZT+ABC-2.39

[back to top]

Time Weighted Mean Change From Baseline Plasma HIV-RNA

(NCT00335322)
Timeframe: 144 weeks

Interventionlog copies/mL (Mean)
TDF/FTC+EFV-2.77
TDF/FTC+ r/ATV-2.88
TDF/FTC + AZT+ABC-2.54

[back to top]

Number of Participants With Genotype Substitutions for Virologic Rebounds (HIV-RNA ≥ 400 c/mL) Through Week 48

International Aids Society of the United States (IAS-USA)-defined major protease inhibitor (PI) substitutions are V32I, L33F, M46I/L, I47V, G48V, I50L/V, I54M/L, I76V, I82A/F/T/S, I84V, N88S, and L90M. Reverse Transcriptase (RT) are TAMS and M184V. (NCT00337467)
Timeframe: Week 48

Interventionparticipants (Number)
IAS-USA-defined major PI substitutionsRT substitutions
Atazanavir (ATV)/Ritonavir (RTV) Monotherapy.10

[back to top]

Percentage of Participants With Virological Rebound Through Week 96

Virological rebound is defined as confirmed on-treatment HIV RNA >= 400 c/mL at 2 consecutive visits or last on-treatment HIV RNA >=400 c/mL followed by discontinuation of study therapy. In addition, virologic rebound defined based on HIV RNA >=50 c/m, latter analysis performed on subjects with baseline HIV RNA < 50 c/mL. (NCT00337467)
Timeframe: Week 96

Interventionpercentage of participants (Number)
HIV RNA >= 400 c/mL (n=61)HIV RNA>=50 c/mL w/Baseline HIV RNA<50 c/mL (n=60)
Atazanavir (ATV)/Ritonavir (RTV) Monotherapy14.833.3

[back to top]

Percentage of Participants With Treatment Failure Through Week 96

Treatment Failure through Week 96 defined as virologic rebound (HIV RNA >=400 c/mL) on or before Week 96 or study discontinuation before Week 96. In addition, treatment failure defined based on HIV RNA >= 50 c/mL, latter analysis performed on treated subjects with baseline HIV RNA < 50 c/mL. (NCT00337467)
Timeframe: Week 96

Interventionpercentage of participants (Number)
HIV RNA >= 400 c/mL(n=61)HIV RNA>=50 c/mL w/Baseline HIV RNA<50 c/mL (n=60)
Atazanavir (ATV)/Ritonavir (RTV) Monotherapy34.443.3

[back to top]

Percentage of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs), Deaths, and Discontinuations Due to AEs

AE=any new untoward medical occurrence or worsening of a pre-existing medical condition that does not necessarily have a causal relationship to treatment. SAE=any untoward medical occurrence that results in death, is life-threatening, requires inpatient hospitalization or causes prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect, or is an important medical event. AE grades are: mild (1), moderate (2), severe (3), life-threatening (4), and death (5). (NCT00337467)
Timeframe: From Baseline through Week 96

Interventionpercentage of participants (Number)
All Grades AEsAll Grades AEs Related to Study TherapyGrade 3 to Grade 4 AEsGrade 3 to Grade 4 AEs Related to Study TherapyDeathsSAEsSAEs Related to Study TherapyAEs Leading to DiscontinuationLipodystrophy-Related AEs
Atazanavir (ATV)/Ritonavir (RTV) Monotherapy.75.413.1183.33.319.704.91.6

[back to top]

Number of Participants With Genotype Substitutions for Virologic Rebounds (HIV-RNA ≥ 400 c/mL) Through Week 96

International Aids Society of the United States (IAS-USA)-defined major protease inhibitor (PI) substitutions are V32I, L33F, M46I/L, I47V, G48V, I50L/V, I54M/L, I76V, I82A/F/T/S, I84V, N88S, and L90M. Reverse Transcriptase (RT) are TAMS and M184V. (NCT00337467)
Timeframe: Week 96

Interventionparticipants (Number)
IAS-USA-defined major PI substitutionsRT substitutions
Atazanavir (ATV)/Ritonavir (RTV) Monotherapy20

[back to top]

Mean Percent Changes From Baseline in Fasting Total Cholesterol, High Density Lipoprotein (HDL) Cholesterol, Non-HDL Cholesterol, Low Density Lipoprotein (LDL) Cholesterol, and Triglycerides at Week 96

Lipid values after starting lipid-reducing agents are excluded from analyses. Baseline values are provided in Baseline Characteristics. (NCT00337467)
Timeframe: Baseline, Week 96

Interventionpercent change (Mean)
Total Cholesterol (n=29)HDL Cholesterol (n=29)Non-HDL Cholesterol (n=29)LDL Cholesterol (n=28)Triglycerides (n=29)
Atazanavir (ATV)/Ritonavir (RTV) Monotherapy.142192916

[back to top]

Mean Percent Changes From Baseline in Fasting Total Cholesterol, High Density Lipoprotein (HDL) Cholesterol, Non-HDL Cholesterol, Low Density Lipoprotein (LDL) Cholesterol, and Triglycerides at Week 48

Lipid values after starting lipid-reducing agents are excluded from analyses. Baseline values are provided in Baseline Characteristics. (NCT00337467)
Timeframe: Baseline, Week 48

Interventionpercent change (Mean)
Total Cholesterol (n=36)HDL Cholesterol (n=35)Non-HDL Cholesterol (n=35)LDL Cholesterol (n=33)Triglycerides (n=36)
Atazanavir (ATV)/Ritonavir (RTV) Monotherapy.92122017

[back to top]

Mean Change From Baseline in Cluster of Differentiation 4 (CD4) Cell Count at Week 24

(NCT00337467)
Timeframe: Baseline, Week 24

Interventioncells /mm3 (Mean)
Baseline Value (n=61)Change at Week 24 (n=51)
Atazanavir (ATV)/Ritonavir (RTV) Monotherapy.55961

[back to top]

Mean Change From Baseline in CD4 Cell Count at Week 96

(NCT00337467)
Timeframe: Baseline, Week 96

Interventioncells /mm3 (Mean)
Baseline Value (n=61)Change at Week 96 (n=40)
Atazanavir (ATV)/Ritonavir (RTV) Monotherapy.55963

[back to top]

Mean Change From Baseline in CD4 Cell Count at Week 48

(NCT00337467)
Timeframe: Baseline, Week 48

Interventioncells /mm3 (Mean)
Baseline Value (n=61)Change at Week 48 (n=46)
Atazanavir (ATV)/Ritonavir (RTV) Monotherapy.55953

[back to top]

Cumulative Proportion of Participants Without Treatment Failure Through Week 100

This Kaplan-Meier life table reports the cumulative proportion of participants without treatment failure up to the end of the respective time interval. Failure time is measured from the start of study therapy, and is based on the earliest event defining failure (virologic rebound at or before Week 96, or discontinuation prior to Week 96). (NCT00337467)
Timeframe: Through Week 100

Interventionproportion of participants (Number)
Interval Week 4 - 8 (n=61)Interval Week 8 - 12 (n=60)Interval Week 12 - 16 (n=58)Interval Week 16 - 20 (n=55)Interval Week 20 - 24 (n=54)Interval Week 24 - 28 (n=53)Interval Week 32 - 36 (n=52)Interval Week 36 - 40 (n=50)Interval Week 48 - 52 (n=48)Interval Week 56 - 60 (n=46)Interval Week 64 - 68 (n=45)Interval Week 68 - 72 (n=43)Interval Week 72 - 76 (n=42)Interval Week 84 - 88 (n=41)Interval Week 92 - 96 (n=40)Interval Week 96 - 100 (n=31)
Proportion of Participants0.98360.95080.90160.88520.86890.85250.81970.78690.75410.73770.70490.68850.67210.65570.65570.6557

[back to top]

Percentage of Participants With Treatment Failure Through Week 48

Treatment Failure through Week 48 defined as virologic rebound (HIV RNA >=400 c/mL) on or before Week 48 or study discontinuation before Week 48. Virological rebound is defined as confirmed on-treatment HIV ribonucleic acid (RNA) >= 400 c/mL at 2 consecutive visits or last on-treatment HIV RNA >=400 c/mL followed by discontinuation of study therapy. (NCT00337467)
Timeframe: Week 48

InterventionPercentage of Participants (Number)
Atazanavir (ATV)/Ritonavir (RTV) Monotherapy21.3

[back to top]

Percentage of Participants With Virological Rebound Through Week 48

Virological rebound is defined as confirmed on-treatment HIV RNA >= 400 c/mL at 2 consecutive visits or last on-treatment HIV RNA >=400 c/mL followed by discontinuation of study therapy. In addition, virologic rebound defined based on HIV RNA >=50 c/m, latter analysis performed on subjects with baseline HIV RNA < 50 c/mL. (NCT00337467)
Timeframe: Week 48

Interventionpercentage of participants (Number)
HIV RNA >= 400 c/mL (n=61)HIV RNA>=50 c/mL w/Baseline HIV RNA<50 c/mL (n=60)
Atazanavir (ATV)/Ritonavir (RTV) Monotherapy1226.7

[back to top]

Serious Adverse Events

Safety outcomes in four different randomly assigned regimens (NCT00342355)
Timeframe: January 2004 until March 31, 2008

Interventionparticipant (Number)
AZT+ddI+EFV73
AZT + ddI + r/LPV69
d4T + 3TC + EFV64
d4T + 3TC + r/LPV60

[back to top]

Progression to AIDS or Death in tx naïve Pts With Adv HIV dx in the Four Randomly Assigned Regimens.

Progression of disease, AIDS, or death in treatment naive patients with advanced HIV diagnosis will be evaluated in the four randomly assigned regimens. (NCT00342355)
Timeframe: January 2004 until March 31 2008

Interventionparticipants (Number)
AZT+ddI+EFV93
AZT + ddI + r/LPV77
d4T + 3TC + EFV70
d4T + 3TC + r/LPV80

[back to top]

Number of Subjects With at Least One New PI-associated Resistance Mutation at Time of Virologic Failure.

Number of subjects with at least one new PI-associated resistance mutation at time of virologic failure. Resistance interpretations used the May 6, 2009 Stanford algorithm. (NCT00357552)
Timeframe: At time of virologic failure

Interventionparticipants (Number)
Virologic Failures by Week 24.2

[back to top]

Percentage of Subjects Reporting Not Skipping Medications in the Last Month.

The percentage of subjects reporting never missing medications in the last month. (NCT00357552)
Timeframe: Study entry and weeks 2, 4, 8, 12, 16, 20, and 24

Interventionpercentage of subjects with data (Number)
week 2 (N=120)week 4 (N=121)week 8 (N=123)week 12 (N=123)week 16 (N=122)week 20 (N=120)week 24 (N=122)
LPV/r Monotherapy9086.887.886.286.190.989.4

[back to top]

Number of Screened Subjects With at Least One NNRTI, or NRTI-associated Resistance Mutation at A5230 Screening.

Number of screened subjects with at least one NNRTI, or NRTI-associated resistance mutation. Resistance interpretations used the November 30, 2011 Stanford algorithm. (NCT00357552)
Timeframe: Screening

Interventionnumber of screened subjects (Number)
At least one NNRTI-associated mutationAt least one NRTI-associated mutation
All Screened Subjects With Available Sequences201197

[back to top]

Proportion of Participants With Plasma HIV-1 RNA Levels < 400 Copies/mL From Baseline to Week 104

(NCT00357552)
Timeframe: At Weeks 0, 12, 16, 20, 24, 32, 40, 48, 56, 68, 80, 92, 104

Interventionproportion of participants (Number)
week 0 (N=123)week 12 (N=122)week 16 (N=121)week 20 (N=115)week 24 (N=122)week 32 (N=121)week 40 (N=118)week 48 (N=118)week 56 (N=120)week 68 (N=116)week 80 (N=117)week 92 (N=116)week 104 (N=117)
LPV/r Monotherapy0.020.750.870.840.840.830.840.870.860.910.850.870.89

[back to top]

Level of HIV-1 RNA as Ascertained From Paired DBS and Plasma

Proportion of DBS samples with HIV-1 RNA level <= 400 copies/mL, proportion of plasma samples with HIV-1 RNA level <= 400 copies/mL and proportion of paired DBS and plasma samples that are concordant (both <= 400 copies/mL or both > 400 copies/mL). Results are pooled over 4 different storage temperature conditions (-80C, -20C, 4C and room temperature). (NCT00357552)
Timeframe: At study entry and weeks 24 and 48

Interventionproportion of samples (Number)
study entry DBS <= 400 cp/mLstudy entry plasma <= 400 cp/mLstudy entry DBS & plasma concordanceweek 24 DBS <= 400 cp/mLweek 24 plasma <= 400 cp/mLweek 24 DBS & plasma concordanceweek 48 DBS <= 400 cp/mLweek 48 plasma <= 400 cp/mLweek 48 DBS & plasma concordance
LPV/r Monotherapy0.170.000.830.820.800.800.940.910.97

[back to top]

Time to Treatment Failure, Defined as the First Occurrence of Death, Disease Progression, or Virologic Failure.

25th percentile in weeks from study entry to treatment failure, defined as the first occurrence of death, disease progression, or virologic failure. Virologic failure was defined as HIV-1 >= 400 copies/mL after week 24 or 2 consecutive HIV-1 RNA >= 400 copies/mL after week 16 following suppression on LPV/r monotherapy. (NCT00357552)
Timeframe: Study entry to Week 104

Interventionweeks (Number)
LPV/r Monotherapy48.0

[back to top]

Time to First New Grade 3 or 4 Sign or Symptom or Laboratory Toxicity Following LPV/r Intensification

25th percentile in weeks from study entry to first new grade 3 or 4 sign or symptom or laboratory toxicity following LPV/r intensification. Grading of adverse events (signs and symptoms and laboratory toxicities) was according to Division of AIDS Table for Grading the Severity of Adult and Pediatric Adverse Events, Version 1.0, December 2004. (NCT00357552)
Timeframe: From LPV/r intensification to week 104

Interventionweeks (Number)
LPV/r Monotherapy26.0

[back to top]

Probability of Grade 3 or 4 Sign or Symptom, or Laboratory Toxicity Over 24 Weeks on Study.

Probability of Grade 3 or 4 sign or symptom, or laboratory toxicity over 24 weeks on study using Kaplan-Meier estimates of the cumulative probability of Grade 3 or 4 sign or symptom, or laboratory toxicity at week 24. Grading of adverse events (signs and symptoms and laboratory toxicities) was according to Division of AIDS Table for Grading the Severity of Adult and Pediatric Adverse Events, Version 1.0, December 2004. (NCT00357552)
Timeframe: From study entry to week 24

Interventioncumulative probability of grade 3 or 4 (Number)
LPV/r Monotherapy0.23

[back to top]

Percentage of Enrolled Participants With Virologic Success at Week 24 on LPV/r Monotherapy

Virologic success at week 24 on LPV/r monotherapy was defined as remaining on LPV/r monotherapy at week 24 without prior virologic failure. Virologic failure was met with either of these two conditions: (i) failure to suppress HIV-1 RNA to < 400 copies/mL by week 24 or (ii) confirmed HIV-1 RNA >= 400 copies/mL after confirmed HIV-1 RNA < 400 copies/mL. (NCT00357552)
Timeframe: From study entry to week 24

Interventionpercentage of enrolled subjects (Number)
LPV/r Monotherapy87

[back to top]

Number of Participants With Study-targeted Diagnoses and Clinical Events

Cardiac disorders, Infections and infestations, Metabolism and nutrition disorders, Neoplasms benign, malignant and unspecified (including cysts and polyps), Pregnancy, puerperium and perinatal conditions, Vascular disorders, were specified a priori as study-targeted events by the study chair. (NCT00357552)
Timeframe: Study entry to week 104

Interventionparticipants (Number)
LPV/r Monotherapy39

[back to top]

Change in CD4+ Cell Counts From Study Entry to Week 104

(NCT00357552)
Timeframe: Study entry and week 104

Interventioncells/mm^3 (Median)
LPV/r Monotherapy213

[back to top]

Virologic Response (HIV-1 RNA <50 Copies/mL) at Week 48 for Participants With 0-2 Protease Inhibitor Substitutions at Baseline Associated With Reduced Response to Lopinavir/Ritonavir

Substitutions considered in the analysis were L10F/I/R/V, K20M/N/R, L24I, L33F, M36I, I47V, G48V, I54L/T/V, V82A/C/F/S/T, and I84V as defined in the proposed United States Package Insert. (NCT00358917)
Timeframe: Week 48 (End of Study)

InterventionPercentage of Participants (Number)
LPV/r 800/200 mg QD Tablet65.5
LPV/r 400/100 mg BID Tablet61.6

[back to top]

Percentage of Participants With Plasma Human Immunodeficiency Virus Type 1 (HIV-1) Ribonucleic Acid (RNA) Levels < 50 Copies/Milliliter (mL) at Week 48

(NCT00358917)
Timeframe: Week 48 (End of Study)

InterventionPercentage of Participants (Number)
LPV/r 800/200 mg QD Tablet76.0
LPV/r 400/100 mg BID Tablet72.2

[back to top]

Percentage of Participants With New Primary Protease Mutations at Week 48

Emergence of new primary protease inhibitor mutations (i.e., mutations at codons 30, 32, 48, 50, 82, 84, and 90 that were not present at baseline). (NCT00358917)
Timeframe: Week 48 (End of Study)

InterventionPercentage of Participants (Number)
LPV/r 800/200 mg QD Tablet8.0
LPV/r 400/100 mg BID Tablet15.6

[back to top]

Percentage of Participants Responding at Week 48 Based on the Food and Drug Administration (FDA) Time to Loss of Virologic Response (TLOVR) Algorithm

A participant was classified as a responder at the first of 2 consecutive human immunodeficiency virus type 1 (HIV-1) ribonucleic acid (RNA) levels <50 copies/mL. The participant continued to be a responder until 2 consecutive values >=50 copies/mL were reached, until the final value if that value was >=50 copies/mL, or until discontinuation or death. (NCT00358917)
Timeframe: Week 48 (End of Study)

InterventionPercentage of Participants (Number)
LPV/r 800/200 mg QD Tablet55.3
LPV/r 400/100 mg BID Tablet51.8

[back to top]

Mean Change From Baseline to Week 48 in Cluster of Differentiation 4 Single-Positive Thymocyte (CD4+ T) Cell Counts

(NCT00358917)
Timeframe: Week 48 (End of Study)

Interventioncells/microliter (Mean)
LPV/r 800/200 mg QD Tablet135.3
LPV/r 400/100 mg BID Tablet121.5

[back to top]

Steady-State Plasma Levels of Amprenavir (APV) and Ritonavir (RTV) Ctau at Weeks 12 and 24

Blood samples were drawn at weeks 12 and 24 to determine the plasma levels of APV and RTV. Concentration at the end of the dosing interval at steady state (Ctau) was presented. (NCT00363142)
Timeframe: Weeks 12 and 24

,,
Interventionmicrograms/mL (Geometric Mean)
Week 12 APV CtauWeek 24 APV CtauWeek 12 RTV CtauWeek 24 RTV Ctau
FPV/r1001.381.270.0260.022
FPV/r2001.281.490.0500.056
FPV/RTV 700/100 mg BID1.352.380.2280.203

[back to top]

Number of Participants With Grade 2-4 Adverse Events Occurring in Greater Than or Equal to 2% of Subjects Through Week 24

The number of participants who experienced any grades 2 to 4 adverse events was tabulated. Adverse events were graded based on the Division of Acquired Immunodeficiency Syndrome (AIDS) Table for Grading the Severity of Adult and Pediatric Adverse Events. (NCT00363142)
Timeframe: Baseline through Week 24

,
Interventionparticipants (Number)
Any eventDiarrheaBronchitisNauseaSinusitisUpper respiratory tract infectionBlood glucose increasedDepressionHeadacheInfluenzaLow density lipoprotein increasedNasopharyngitisOtitis mediaBack painHypercholesterolemiaInsomnia
FPV/r10048833343111331000
FPV/r20024231100222002222

[back to top]

Percentage of Participants With Plasma HIV-1 RNA <50 Copies/mL at Week 24, TLOVR Analysis

A blood sample was drawn to determine the amount of plasma HIV-1 RNA virus in copies/mL at week 24. The percentage of participants plasma with HIV-1 RNA <50 copies/mL at Week 24 were determined by the TLOVR algorithm with stratification by the six randomization strata. (NCT00363142)
Timeframe: Week 24

,
InterventionPercentage of participants (Number)
Plasma HIV-1 RNA <50 copies/mLHIV-1 RNA greater than or equal to 50 copies/mL
FPV/r10082.917.1
FPV/r20084.615.4

[back to top]

Percentage of Participants With Plasma Human Immunodeficiency Virus, Type 1, Ribonucleic Acid (HIV-1 RNA) <400 Copies/mL at Week 24, Time to Loss of Virologic Response (TLOVR) Analysis

A blood sample was drawn to determine the amount of plasma HIV-1 RNA virus in copies/mL at week 24. The percentage of participants with plasma HIV-1 RNA <400 copies/mL at Week 24 were determined by the TLOVR algorithm with stratification by the six randomization strata. (NCT00363142)
Timeframe: Week 24

,
InterventionPercentage of participants (Number)
Plasma HIV-1 RNA <400 copies/mLHIV-1 RNA greater than or equal to 400 copies/mL
FPV/r100928
FPV/r20094.25.8

[back to top]

Number of Participants Who Discontinued Treatment Due to Adverse Events Through Week 24

The number of participants who prematurely discontinued study drug due to adverse events was tabulated. Data are summarized by individual adverse event. Adverse events were defined as any untoward medical occurrence in a patient or clinical investigation subject, temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product. (NCT00363142)
Timeframe: Baseline through Week 24

,
Interventionparticipants (Number)
Any eventAlanine aminotransferase increasedAspartate aminotransferase increasedMetastatic neoplasmsCentral nervous system lesion
FPV/r10021111
FPV/r20000000

[back to top]

Percentage of Participants Not Meeting the Definition of Virologic Failure at or Prior to Week 24

Virologic failure was defined as two consecutive plasma HIV-1 RNA measures greater than 400 copies/milliliter (mL) separated by at least 2 to 4 week. The percentage of participants not meeting the virologic failure definition was estimated with stratification by the six randomization strata using Mantel-Haenszel weights and the missing/discontinuation equals failure (MD=F) analysis. Missing/discontinuation values were considered failures. (NCT00363142)
Timeframe: Week 24

InterventionPercentage of participants (Number)
FPV/r10092.1
FPV/r20094.2

[back to top]

Percent Change From Baseline in Low Density Lipoprotein (LDL) at Week 24

A blood sample was drawn to determine the LDL level at Week 24. Percent change in LDL was defined as (LDL level at Week 24 minus level at baseline) divided by level at baseline x 100%. (NCT00363142)
Timeframe: Baseline and Week 24

InterventionPercent change (Median)
FPV/r1000
FPV/r2002.1

[back to top]

Number of Participants With Plasma HIV-1 RNA Genotypic Mutations and Phenotypic Resistance at Time of Virologic Failure Not Present at Baseline

A blood sample was drawn for subjects failing to respond to therapy and the mutations present in the virus were identified. For each subject, the mutations found at the time of failure were compared with any mutations found in the blood sample at baseline. New mutations that developed at the time of virologic failure were tabulated by drug class. (NCT00363142)
Timeframe: Baseline through Week 24

InterventionParticipants (Number)
FPV/r2000

[back to top]

Median Change From Baseline of CD4+ Cell Count at Week 24, Observed Analysis

A blood sample was drawn to determine the CD4+ cell count at week 24. Change from baseline was defined as CD4+ cell count at Week 24 minus CD4+ cell count at baseline. (NCT00363142)
Timeframe: Baseline and Week 24

Interventioncells/mm3 (Median)
FPV/r10011.5
FPV/r20015

[back to top]

Mean Change From Baseline of log10 Copies/mL Plasma HIV-1 RNA Levels at Week 24, Observed Analysis

A blood sample was drawn to determine the amount of plasma HIV-1 RNA virus in copies/mL at week 24. Change from baseline was defined as plasma HIV-1 RNA level at Week 24 minus plasma HIV-1 RNA level at baseline. (NCT00363142)
Timeframe: Baseline and Week 24

Interventionlog10 copies/mL (Mean)
FPV/r100-0.015
FPV/r200-0.022

[back to top]

Percent Change From Baseline in Total Cholesterol, High Density Lipoprotein (HDL), and Triglycerides at Week 24

A blood sample was drawn to determine the cholesterol, HDL, triglycerides levels at Week 24. Percent change in total blood cholesterol, HDL, and triglycerides was defined as (lipid level at Week 24 minus level at baseline) divided by level at baseline x 100%. (NCT00363142)
Timeframe: Baseline and Week 24

,
InterventionPercent change (Median)
Total cholesterolHDLTriglycerides
FPV/r100-0.5-2.1-13.5
FPV/r2000.70-0.6

[back to top]

Descriptive Statistics of ETR Subgroup - Change From Baseline in CD4+ Cell Count Using the Imputation Method of LOCF

The Etravirine-TMC125 (ETR Subgroup population was defined as all ITT subjects who took at least 1 dose of ETR). The Last Observation Carried Forward (LOCF) imputation method was applied. (NCT00381303)
Timeframe: Week 48

Interventionx10^6 Cells/L (Mean)
Female107
Male98
Black113
Caucasian84
Hispanic90
Asian45
Other138

[back to top]

Descriptive Statistics of Change From Baseline in CD4+ Cell Count Using Observed Values

Observed obsevations have no imputation methods applied. (NCT00381303)
Timeframe: Baseline, Week 48

Interventionx10^6 cells/L (Mean)
Female152
Male122
Black143
Caucasian151
Hispanic133
Asian45
Other179

[back to top]

Descriptive Statistics of Change From Baseline in CD4+ Cell Count Using the Imputation Method of Last Observation Carried Forward (LOCF)

Last Observation Carried Forward (LOCF) imputation method applied. (NCT00381303)
Timeframe: Week 48

Interventionx10^6 cells/L (Mean)
Female112
Male103
Black109
Caucasian110
Hispanic109
Asian45
Other138

[back to top]

Descriptive Statistics of [TLOVR Non-virologic Failure (VF) Censored] - VL < 50 HIV-1 RNA by Race

TLOVR non-virologic failure (VF) censored. This imputation method differs from the TLOVR algorithm, because subjects that dropped out for reasons other than virologic failure were censored from the time of discontinuation onwards. (NCT00381303)
Timeframe: Week 48

Interventionparticipants (Number)
Black128
Caucasian39
Hispanic59
Asian2
Other1

[back to top]

Descriptive Statistics of ETR Subgroup - Change From Baseline in CD4+ Cell Using Observed Values

The Etravirine-TMC125 (ETR Subgroup population was defined as all ITT subjects who took at least 1 dose of ETR) (NCT00381303)
Timeframe: Week 48

Interventionx10^6 cells/L (Mean)
Female136
Male108
Black135
Caucasian111
Hispanic98
Asian45
Other179

[back to top]

Descriptive Statistics of ETR Subgroup [TLOVR Non-virologic Failure (VF) Censored] - VL < 50 HIV-1 RNA

"The Etravirine-TMC125 (ETR Subgroup population was defined as all ITT subjects who took at least 1 dose of ETR)~TLOVR non-virologic failure(VF) censored. This imputation method differs from the TLOVR algorithm, because subjects that dropped out for reasons other than virologic failure were censored from the time of discontinuation onwards." (NCT00381303)
Timeframe: Week 48

Interventionparticipants (Number)
Female69
Male54
Black74
Caucasian21
Hispanic25
Asian2
Other1

[back to top]

Number of Etravirine-TMC125 (ETR) Subgroup- VL < 50 HIV-1 RNA Copies/mL (TLOVR) Subjects

The Etravirine-TMC125 (ETR Subgroup population was defined as all ITT subjects who took at least 1 dose of ETR) (NCT00381303)
Timeframe: Week 48

Interventionparticipants (Number)
Female69
Male54
Black74
Caucasian21
Hispanic25
Asian2
Other1

[back to top]

Number of TLOVR Non-virologic Failure (VF) Censored - VL < 50 HIV-1 RNA Subjects by Sex

TLOVR non-virologic failure (VF) censored. This imputation method differs from the TLOVR algorithm, because subjects that dropped out for reasons other than virologic failure were censored from the time of discontinuation onwards. (NCT00381303)
Timeframe: Week 48

Interventionparticipants (Number)
Female146
Male83

[back to top]

Number of Viral Load (VL) < 50 HIV-1 RNA Copies/mL (Time to Loss of Virologic Response[TLOVR]) Subjects by Sex

TLOVR - responders/non-responders per FDA TLOVR response algorithm. A subject was considered a responder at that time point and that subsequent. A subject was considered a non-responder at a time point in the following situations: discontinued treatment at that time point, a rebound value at that time point and that subsequent or at that time point and that followed by treatment discontinuation, intermittent missing values were considered a response if the immediately preceding and following visits were a response, rebound at earlier time point, or any new, unplanned ARV except in tolerability (NCT00381303)
Timeframe: Week 48

Interventionparticipants (Number)
Female146
Male83

[back to top]

Number of VL < 50 HIV-1 RNA Copies/mL (TLOVR) Subjects by Race

Intention to Treat population (ITT) (NCT00381303)
Timeframe: Week 48

Interventionparticipants (Number)
Black128
Caucasian39
Hispanic59
Asian2
Other1

[back to top]

Total Bilirubin

6 month mean and standard deviation for total bilirubin. (NCT00413153)
Timeframe: 6 months

Interventionmg/dL (Mean)
Boosted Reyataz (ATV/r)2.8
Continue Kaletra (LPV/r)0.6

[back to top]

Insulin Sensitivity

6 month mean and standard deviation for insulin-stimulated glucose uptake (M) per unit insulin at 120 minutes as measured by euglycemic hyperinsulinemic clamp. (NCT00413153)
Timeframe: 6 months

Interventionumol/kg/min per uU/mL insulin (Mean)
Boosted Reyataz (ATV/r)39.0
Continue Kaletra (LPV/r)49.2

[back to top]

Liver Enzymes -- Aspartate Aminotransferase (AST)

6 month mean and standard deviation for AST. (NCT00413153)
Timeframe: 6 months

InterventionU/L (Mean)
Boosted Reyataz (ATV/r)39
Continue Kaletra (LPV/r)42

[back to top]

Body Composition - Visceral Adipose Tissue

6 month mean and standard deviation for visceral adipose tissue (VAT) as measured by single slice computed tomography (CT) scan at the L4 pedicle (pedicle of 4th lumbar vertebra). (NCT00413153)
Timeframe: 6 months

Interventionsquare centimeters (Mean)
Boosted Reyataz (ATV/r)91
Continue Kaletra (LPV/r)167

[back to top]

Fasting Glucose

6 month mean and standard deviation for fasting glucose. (NCT00413153)
Timeframe: 6 months

Interventionmg/dL (Mean)
Boosted Reyataz (ATV/r)84
Continue Kaletra (LPV/r)90

[back to top]

Glucose Trafficking

"6 month mean and standard deviation for glucose uptake into anterior thigh muscle as measured by FDG/PET scanning during euglycemic hyperinsulinemic clamp. During the hyperinsulinemic conditions of the clamp, glucose and 18-FDG [labeled glucose] are taken up by muscle. The quantity of 18-FDG taken up is measured by the PET scan. Although there are no well-accepted norms for this measurement, a higher value indicates that more glucose is being taken up by (or trafficked to) muscle. Increased uptake of glucose indicates increased muscle insulin sensitivity." (NCT00413153)
Timeframe: 6 months

Interventionumol/kg/min (Mean)
Boosted Reyataz (ATV/r)26.7
Continue Kaletra (LPV/r)24.4

[back to top]

Immune Parameters -- CD4 Count

6 month mean and standard deviation for CD4+ count. (NCT00413153)
Timeframe: 6 months

Interventioncells/microL (Mean)
Boosted Reyataz (ATV/r)432
Continue Kaletra (LPV/r)688

[back to top]

Liver Enzymes -- Alanine Aminotransferase (ALT)

6 month mean and standard deviation for ALT. (NCT00413153)
Timeframe: 6 months

InterventionU/L (Mean)
Boosted Reyataz (ATV/r)61
Continue Kaletra (LPV/r)65

[back to top]

Lipid Metabolism - Serum Triglyceride

6 month mean and standard deviation for serum triglyceride. (NCT00413153)
Timeframe: 6 months

Interventionmg/dL (Mean)
Boosted Reyataz (ATV/r)147
Continue Kaletra (LPV/r)209

[back to top]

Viral Set Point

set point is reached after the immune system has developed HIV antibodies and begins to attempt to fight the virus (NCT00414518)
Timeframe: Throughout study

InterventionLog 10 copies virus/ml (Mean)
12 Week Treatment Folllowed by Treatment Interruption4.8627
CD4 T Cell Guided Therapy4.2434

[back to top]

Number of Participants Experiencing Either an AIDS-defining Event, a Grade 3 or 4 Adverse Event, or Acute Retroviral Syndrome

(NCT00414518)
Timeframe: At Week 24

Interventionparticipants (Number)
Arm A1
Arm B1

[back to top]

Plasma HIV-1 Viral Load (Copies/ml) at Week 24 as Compared Between the Two Arms

(NCT00414518)
Timeframe: At Week 24

InterventionLog 10 copies of virus/ml (Mean)
12 Week Treatment Arm Followed by Treatment Interruption4.8627
CD4 T Cell Guided Therapyh4.2620

[back to top]

Incidence of Severe Adverse Events (Excluding Mortality)

(NCT00427297)
Timeframe: 2 years

Interventionevent (Number)
NVP-containing21
NVP-sparing6

[back to top]

Incidence of Mortality

Death during follow-up (NCT00427297)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
NVP-containing4
NVP-sparing5

[back to top]

Immunologic Failure

Immunologic treatment failure was defined as CD4% dropping below 15%, after a previous result greater than or equal to 15% (following along WHO Guidelines). (NCT00427297)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
NVP-containing2
NVP-sparing1

[back to top]

Viral Failure

Virologic treatment failure was defined as follow-up (at least 24 weeks after enrollment date) viral load > 400 copies. (NCT00427297)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
NVP-containing2
NVP-sparing2

[back to top]

Number Participants With Abnormal Vital Signs

Abnormal Vital signs included are high and low Pulse rate (PR), high and how Temperature (Temp), high and low Systolic Blood Pressure (SBP) and high and low Diastolic Blood Pressure (DBP). Vital signs (SBP, DBP, PR,Temp) were measured after participants were in a semi-supine position for at least 5 minutes. (NCT00435929)
Timeframe: Up to Day 35

,
Interventionparticipants (Number)
High PRLow PRHigh TempLow TempHigh-SBPLow-SBPHigh-DBPLow-DBP
Moderate Hepatic Impairment00001002
Normal Liver Function00001001

[back to top]

Maximum Observed Plasma Concentration (Cmax) of SQV and RTV

The plasma concentration (Cmax) is defined as maximum observed analyte concentration. The pharmacokinetic profile for following 14 days of administration with SQV/RTV 1000/100 mg BID included determining the maximum observed plasma concentration (C max) of SQV and Ritonavir RTV The Cmax was analyzed for SQV and RTV by non-compartmental methods, using WinNonlin. (NCT00435929)
Timeframe: Pre-dose and at 0.5 hr, 1hr, 2hrs, 3hrs, 4hrs, 5hrs, 6hrs, 8hrs, 10hrs, and 12 hrs post-dose on Day 14

,
Interventionng/mL (Mean)
Cmax of SQVCmax of RTV
Moderate Hepatic Impairment36101460
Normal Liver Function43001500

[back to top]

Number of Participants With Abnormal Electrocardiogram (ECG) Findings

Abnormal ECG findings included are high and low Heart Rate (HRT), high and low PQ/PR interval (PQ/PR), high and low QRS interval (QRS), high and low QT interval (QT), high and low QTCB interval (QTcB), high and low QTcF interval (QTcF), high and low RR interval (RR), high and low T Wave, high and low U Wave, high and low ECG. The 12 Lead ECG was recorded after participants were in a semi-supine position for at least 5 minutes. Only participants with abnormal ECG findings are presented in the table below. (NCT00435929)
Timeframe: Up to Day 35

,
Interventionparticipants (Number)
HIGH- HRT ECG ; n=7, 9LOW- HRT ECG; n=7, 9HIGH- PQ (PR); n=7, 9LOW- PQ (PR); n=7, 9HIGH- QRS; n=7, 9LOW- QRS; n=7, 9HIGH- QT; n=7, 9LOW- QT; n=7, 9HIGH- QTcB; n=5, 8LOW- QTcB; n=5, 8HIGH- QTcF; n=5, 8LOW- QTcF; n=5, 8HIGH- RR; n=5, 8LOW- RR; n=5, 8HIGH- T WAVE; n=7, 9LOW- T WAVE; n=7, 9HIGH- U WAVE; n=1, 0LOW- U WAVE; n=1, 0HIGH- ECG; n=7, 9LOW- ECG; n=7, 9
Moderate Hepatic Impairment00100020202000000000
Normal Liver Function10000000000001000000

[back to top]

Number of Participants With the Indicated Grade 3 and Grade 4 Laboratory Parameters

Laboratory parameters specified in Clinical Operating Guidelines (COG) were summarized. AIDS Clinical Trial Group (ACTG) and American Heart Association (AHA) criteria were used to grade COG laboratory test values. Laboratory parameters for which an increase to Grade 3 (G3) or Grade 4 (G4) occurred are presented in the table below. (NCT00435929)
Timeframe: Up to Day 35

,
Interventionparticipants (Number)
ASAT (SGOT), (Hyper) G3ASAT (SGOT), (Hyper) G4Alkaline Phosphatase, (Hyper) G3Alkaline Phosphatase, (Hyper) G4ALAT (SGPT), (Hyper) G3ALAT (SGPT), (Hyper) G4Direct Bilirubin, (Hyper) G3Direct Bilirubin, (Hyper) G4Creatinine, (Hyper) G3Creatinine, (Hyper) G4Albumin, (Hypo) G3Albumin, (Hypo) G4Prothrombin Time seconds(Hyper) G3Prothrombin Time seconds (Hyper) G4Creatine Kinase, (Hyper) G3Creatine Kinase, (Hyper) G4Cholesterol, (Hyper) G3Cholesterol, (Hyper) G4Triglycerides, (Hyper) G3Triglycerides, (Hyper) G4Calcium, (Hypo) G3Calcium, (Hypo) G4Potassium, (Hypo) G3Potassium, (Hypo) G4Sodium, (Hypo) G3Sodium, (Hypo) G4Platelets, (Hypo) G3Platelets, (Hypo) G4Neutrophils, (Hypo) G3Neutrophils, (Hypo) G4Fasting Glucose, (Hyper) G3Fasting Glucose, (Hyper) G4Amylase, (Hyper) G3Amylase, (Hyper) G4Uric Acid, (Hyper) G3Uric Acid, (Hyper) G4Proteinuria 0 to 4+, (Hyper) G3Proteinuria 0 to 4+, (Hyper) G4
Moderate Hepatic Impairment10000011000010100000000000001000102010
Normal Liver Function00000000000000000020000000000000000000

[back to top]

Plasma Clearance After Oral Administration (CL/F) of SQV and RTV

The CL/F is the oral clearance; that is clearance based on oral bioavailability. The Pharmacokinetic profile for following 14 days of administration with SQV/RTV 1000/100 mg BID included determining the plasma clearance after oral administration (CL/F)of SQV and RTV The CL/F was analyzed for SQV and RTV by non-compartmental methods, using WinNonlin. (NCT00435929)
Timeframe: Pre-dose and at 0.5 hr, 1hr, 2hrs, 3hrs, 4hrs, 5hrs, 6hrs, 8hrs, 10hrs, and 12 hrs post dose on Day 14

,
InterventionL/hr (Mean)
CL/F of SQVCL/F of RTV
Moderate Hepatic Impairment84.41612.568
Normal Liver Function47.0529.289

[back to top]

Terminal Half-life (T1/2) of SQV and RTV

Terminal half-life is the time measured for the plasma concentration to decrease by one half. The Pharmacokinetic profile for following 14 days of administration with SQV/RTV 1000/100 mg BID included determining the terminal half-life (T1/2) of SQV and RTV. The T1/2 was analyzed for SQV and RTV by non-compartmental methods, using WinNonlin. (NCT00435929)
Timeframe: Pre-dose and at 0.5 hr, 1hr, 2hrs, 3hrs, 4hrs, 5hrs, 6hrs, 8hrs, 10hrs, and 12 hrs post-dose on Day 14

,
Interventionhour (Mean)
T1/2 of SQVT1/2 of RTV
Moderate Hepatic Impairment4.104.82
Normal Liver Function3.313.80

[back to top]

Minimum Observed Plasma Concentration (Cmin) of SQV and RTV

Cmin is the minimum blood plasma concentration that a drug achieves. The Pharmacokinetic profile for following 14 days of administration with SQV/RTV 1000/100 mg BID included determining the minimum observed plasma concentration (C min) of SQV and RTV The Cmin was analyzed for SQV and RTV by non-compartmental methods, using WinNonlin. (NCT00435929)
Timeframe: Pre-dose and at 0.5 hr, 1hr, 2hrs, 3hrs, 4hrs, 5hrs, 6hrs, 8hrs, 10hrs, and 12 hrs post dose on Day 14

,
Interventionng/mL (Mean)
Cmin of SQVCmin of RTV
Moderate Hepatic Impairment834418
Normal Liver Function965399

[back to top]

Time of Maximum Plasma Concentration (Tmax) of SQV and RTV

The Tmax is defined as actual sampling time to reach maximum observed analyte concentration. The Pharmacokinetic profile for following 14 days of administration with SQV/RTV 1000/100 mg BID included determining the time of maximum plasma concentration of SQV and RTV. The Tmax was analyzed for SQV and RTV by non-compartmental methods, using WinNonlin. (NCT00435929)
Timeframe: Pre-dose and at 0.5 hr, 1hr, 2hrs, 3hrs, 4hrs, 5hrs, 6hrs, 8hrs, 10hrs, and 12 hrs post-dose on Day 14

,
Interventionhour (Mean)
Tmax of SQVTmax of RTV
Moderate Hepatic Impairment5.004.07
Normal Liver Function5.004.29

[back to top]

Area Under the Plasma Concentration-Time Curve From Time of Administration to 12 Hours After Dosing (AUC 0-12h) of Saquinavir (SQV) and Ritonavir (RTV)

Area Under the Plasma Concentration-Time Curve (AUC) is a measure of the plasma concentration of the drug over time. It is used to characterize drug absorption. The pharmacokinetic profile for following 14 days of administration with SQV/RTV 1000/100 mg BID included determining the area under the plasma concentration-time curve from 0 to 12 hours after dosing (AUC (0-12h) of SQV and RTV The AUC (0-12hours) was analyzed for SQV and RTV by non-compartmental methods, using WinNonlin. (NCT00435929)
Timeframe: Pre-dose and at 0.5 hr, 1hr, 2hrs, 3hrs, 4hrs, 5hrs, 6hrs, 8hrs, 10hrs, and 12 hrs post-dose on Day 14

,
Interventionng*hr/mL (Mean)
AUC for SQVAUC for RTV
Moderate Hepatic Impairment243329930
Normal Liver Function2851810985

[back to top]

Cluster of Differentiation 4 (CD4 ) Count

The pharmacodynamic profile for following 14 days of administration with SQV/RTV 1000/100 mg BID included determining the cluster of differentiation 4 (CD4) count in participants in each group. (NCT00435929)
Timeframe: Screening (Day -35 to -1), pre-dose on Day 8, Day 14 and at follow up (Day 28-Day 35)

,
Interventioncells/cubic millimeter (Mean)
Screening; n=7, 9Day 8;n=6, 9Day 14;n=7, 9Follow up; n=6, 8
Moderate Hepatic Impairment451.444558.000566.333567.625
Normal Liver Function540.000638.500683.571692.500

[back to top]

Volume of Distribution (Vd) of SQV and RTV

Vd is defined as the theoretical volume in which the total amount of drug would need to be uniformly distributed to produce the desired blood concentration of a drug. The pharmacokinetic profile for following 14 days of administration with SQV/RTV 1000/100 mg BID included determining the Vd of SQV and RTV The Vd was analyzed for SQV and RTV by non-compartmental methods, using WinNonlin. (NCT00435929)
Timeframe: Pre-dose and at 0.5 hr, 1hr, 2hrs, 3hrs, 4hrs, 5hrs, 6hrs, 8hrs, 10hrs, and 12 hrs post dose on Day 14

,
InterventionLitres (Mean)
Vd of SQVVd of RTV
Moderate Hepatic Impairment464.049102.585
Normal Liver Function213.29450.232

[back to top]

Mean Age at Baseline of Participants Randomized to Treatment for the 48-Week Randomized Phase

The mean age of participants randomized to treatment in the Randomized Phase was calculated at Baseline. (NCT00440947)
Timeframe: Baseline of Randomized Phase

Interventionyears (Mean)
ABC/3TC + ATV: Randomized Phase37.5
ABC/3TC + ATV/r: Randomized Phase39.7

[back to top]

Change From Baseline in HIV-1 RNA at Week 84

Change from baseline was calculated as the Week 84 value minus the baseline value. Blood was drawn to analyze for plasma HIV viral load. (NCT00440947)
Timeframe: Baseline and Week 84

Interventionlog10 c/ml (Mean)
ABC/3TC + ATV: Randomized Phase-3.261
ABC/3TC + ATV/r: Randomized Phase-3.270

[back to top]

Change From Baseline in HIV-1 RNA at Week 36

Change from baseline was calculated as the Week 36 value minus the baseline value. Blood was drawn to analyze for plasma HIV viral load. (NCT00440947)
Timeframe: Baseline and Week 36

Interventionlog10 c/ml (Mean)
ABC/3TC + ATV/r: Induction Phase-3.241

[back to top]

Change From Baseline in HIV-1 RNA at Week 144

Change from baseline was calculated as the Week 144 value minus the baseline value. Blood was drawn to analyze for plasma HIV viral load. (NCT00440947)
Timeframe: Baseline and Week 144

Interventionlog10 c/ml (Mean)
ABC/3TC + ATV: Extension Phase-3.291
ABC/3TC + ATV/r: Extension Phase-3.239

[back to top]

Mean Percent Compliance at Week 84

Percent compliance is defined as the total number of pills taken divided by the total number of pills prescribed. The total number of pills taken was calculated by subtracting any returned pills from the total number of pills that were dispensed to each participant during this period. Compliance was calculated for each medication in the regimen. (NCT00440947)
Timeframe: Week 84

,
Interventionpercent compliance (Mean)
Abacavir/LamivudineRitonavirAtazanavir
ABC/3TC + ATV: Randomized Phase92.092.998.9
ABC/3TC + ATV/r: Randomized Phase91.291.592.4

[back to top]

Change From Baseline in CD4+ Cell Count at Week 84

A CD4+ cell is a T lymphocyte that carries the CD4 antigen. Immunologic response was assessed by CD4+ counts. Change from baseline was calculated as the Week 84 value minus the baseline value. Blood was drawn to analyze for CD4+ cell count. (NCT00440947)
Timeframe: Baseline and Week 84

Interventioncells/millimeters cubed (mm^3) (Mean)
ABC/3TC + ATV: Randomized Phase265.7
ABC/3TC + ATV/r: Randomized Phase282.9

[back to top]

Change From Baseline in CD4+ Cell Count at Week 36

Blood was drawn to analyze for CD4+ cell count. A CD4+ cell is a T lymphocyte that carries the CD4 antigen. Immunologic response was assessed by CD4+ counts. Change from baseline was calculated as the Week 36 value minus the baseline value. (NCT00440947)
Timeframe: Baseline and Week 36

Interventioncells/millimeters cubed (mm^3) (Mean)
ABC/3TC + ATV/r: Induction Phase185.4

[back to top]

Change From Baseline in CD4+ Cell Count at Week 144

A CD4+ cell is a T lymphocyte that carries the CD4 antigen. Immunologic response was assessed by CD4+ counts. Change from baseline was calculated as the Week 144 value minus the baseline value. Blood was drawn to analyze for CD4+ cell count. (NCT00440947)
Timeframe: Baseline and Week 144

Interventioncells/millimeters cubed (mm^3) (Mean)
ABC/3TC + ATV: Extension Phase317.7
ABC/3TC + ATV/r: Extension Phase325.1

[back to top]

Percentage of Participants Who Achieved Plasma HIV-1 RNA <50 c/ml at the Week 84 Visit

A blood sample was drawn to determine the amount of HIV-1 RNA virus in c/ml at Week 84. The percentage of participants with HIV-1 RNA <50 c/ml at Week 84 was tabulated. The secondary analysis methods were: Observed (Obs; uses all visits with data in the analysis period), and missing/discontinuation=failure (M/D=F) analyses. M/D=F: participants with missing data or data collected after study medication DC were considered failures. (NCT00440947)
Timeframe: Week 84

,
Interventionpercentage of participants (Number)
ObsM/D=F
ABC/3TC + ATV: Randomized Phase9285
ABC/3TC + ATV/r: Randomized Phase9282

[back to top]

Percentage of Participants Who Achieved Plasma HIV-1 RNA <50 c/ml at the Week 36 Visit

The percentage of PAR with HIV-1 RNA virus <50 c/ml from a Week 36 blood sample was tabulated. Per TLOVR algorithm, responders were PAR with confirmed viral load <50 c/ml who had not met any non-responder criterion. Non-responders were PAR who never achieved confirmed HIV RNA <50 c/ml, prematurely discontinued (DC) study or study medication (any reason), had confirmed rebound to >=50 c/ml, or had an unconfirmed HIV RNA >=50 c/ml at last visit. ITT-E observed analysis (Obs): all observed data. ITT-E M/D=F analysis: PAR with missing data/data collected after study medication DC were failures. (NCT00440947)
Timeframe: Week 36

Interventionpercentage of participants (Number)
TLOVRObsM/D=F
ABC/3TC + ATV/r: Induction Phase808877

[back to top]

Percentage of Participants Who Achieved Plasma HIV-1 RNA <50 c/ml at the Week 144 Visit

Percentage of PAR with HIV-1 RNA <50 c/ml at Week 144 was tabulated; stratified by baseline HIV-1 RNA (<100,000 and >=100,000 c/ml). Per TLOVR algorithm, responders were PAR with confirmed (CF) HIV RNA <50 c/ml who had not met any non-responder (NR) criterion. NR were PAR who never achieved CF HIV RNA <50 c/ml, prematurely discontinued (DC) study or study medication (Med), had CF rebound to >=50 c/ml, or had an unconfirmed HIV RNA >=50 c/ml at last visit. Observed analysis (Obs): all observed data. M/D=F analysis: PAR with missing data/data collected after study Med DC were failures. (NCT00440947)
Timeframe: Week 144

,
Interventionpercentage of participants (Number)
TLOVRObsM/D=F
ABC/3TC + ATV: Extension Phase779580
ABC/3TC + ATV/r: Extension Phase739278

[back to top]

Percentage of Participants Who Achieved HIV-1 RNA <400 c/ml at the Week 144 Visit

Percentage of PAR with HIV-1 RNA <400 c/ml at Week 144 was tabulated; stratified by baseline HIV-1 RNA (<100,000 and >=100,000 c/ml). Per TLOVR algorithm, responders were PAR with confirmed (CF) HIV-RNA <400 c/ml who had not met any non-responder (NR) criterion. NR were PAR who never achieved CF HIV RNA <400 c/ml, prematurely discontinued (DC) study or study medication (Med), had CF rebound to >=400 c/ml, or had an unconfirmed HIV RNA >=400 c/ml at last visit. Observed analysis (Obs): all observed data. M/D=F analysis: PAR with missing data/data collected after study Med DC were failures. (NCT00440947)
Timeframe: Week 144

,
Interventionpercentage of participants (Number)
TLOVRObsM/D=F
ABC/3TC + ATV: Extension Phase849984
ABC/3TC + ATV/r: Extension Phase809782

[back to top]

Number of Participants Who Met the Protocol-defined Virologic Failure (PDVF) Criteria at Week 36

The number of participants that failed to respond to therapy through 36 weeks on treatment, based on the protocol definition of virologic failure (PDVF), was tabulated. PDVF was defined as (a) failure to achieve plasma HIV-1 RNA <400 c/ml by Week 30 or (b) confirmed HIV-1 RNA rebound >=400 c/ml after achieving HIV-1 <400 c/ml. (NCT00440947)
Timeframe: Week 36

Interventionparticipants (Number)
Protocol-defined virologic failureFailure to achieve <400 c/ml by Week 30Confirmed rebound after achieving <400 c/ml
ABC/3TC + ATV/r: Induction Phase15510

[back to top]

Number of Participants Who Met the PDVF Criteria at Week 84

The number of participants that failed to respond to therapy from the time of treatment randomization through Week 84, based on the protocol definition of virologic failure (PDVF), was tabulated. PDVF was defined as (a) failure to achieve plasma HIV-1 RNA <400 c/ml by Week 30 or (b) confirmed HIV-1 RNA rebound >=400 c/ml after achieving HIV-1 <400 c/ml. (NCT00440947)
Timeframe: Week 84

,
Interventionparticipants (Number)
Protocol-defined virologic failureConfirmed rebound after achieving <400 c/ml
ABC/3TC + ATV: Randomized Phase11
ABC/3TC + ATV/r: Randomized Phase77

[back to top]

Number of Participants Who Met the PDVF Criteria at Week 144

The number of participants enrolled in the extension phase that failed to respond to therapy from Week 84 through Week 144, based on the protocol definition of virologic failure (PDVF) was tabulated,. PDVF was defined as (a) failure to achieve plasma HIV-1 RNA <400 c/ml by Week 30 or (b) confirmed HIV-1 RNA rebound >=400 c/ml after achieving HIV-1 <400 c/ml. (NCT00440947)
Timeframe: Week 144

,
Interventionparticipants (Number)
Protocol-defined virologic failureConfirmed rebound after achieving <400 c/ml
ABC/3TC + ATV: Extension Phase55
ABC/3TC + ATV/r: Extension Phase66

[back to top]

Number of Confirmed Virologic Failure Participants With Treatment-emergent HIV Genotypic Resistance in Reverse Transcriptase and Protease From Week 84 Through Week 144

A blood sample was drawn for participants failing to respond to therapy, and the mutations present in the virus were identified. For each participant, the mutations found at the time of failure were compared with any mutations found in the blood sample at baseline. New International AIDs Society-USA defined resistance mutations that developed at the time of failure were tabulated by drug class. VF, virologic failure; NRTI, nucleoside reverse transcriptase inhibitor; NNRTI, non-nucleoside reverse transcriptase inhibitor; PI, protease inhibitor. (NCT00440947)
Timeframe: Week 84 through Week 144

,
Interventionparticipants (Number)
PAR with paired genotypes at baseline and VFPAR with treatment-emergent mutationsPAR with NRTI mutationsPAR with NNRTI mutationsPAR with major PI mutationsPAR with minor PI mutations
ABC/3TC + ATV: Extension Phase520002
ABC/3TC + ATV/r: Extension Phase511011

[back to top]

Number of Confirmed Virologic Failure Participants With Treatment-emergent HIV Genotypic Resistance in Reverse Transcriptase and Protease From Randomization at Week 36 Through Week 84

A blood sample was drawn for participants failing to respond to therapy, and the mutations present in the virus were identified. For each participant, the mutations found at the time of failure were compared with any mutations found in the blood sample at baseline. New International AIDs Society-USA defined resistance mutations that developed at the time of failure were tabulated by drug class. VF, virologic failure; NRTI, nucleoside reverse transcriptase inhibitor; NNRTI, non-nucleoside reverse transcriptase inhibitor; PI, protease inhibitor. (NCT00440947)
Timeframe: Randomization at Week 36 through Week 84

,
Interventionparticipants (Number)
PAR with paired genotypes at baseline and VFPAR with treatment-emergent mutationsPAR with NRTI mutationsPAR with NNRTI mutationsPAR with major PI mutationsPAR with minor PI mutations
ABC/3TC + ATV: Randomized Phase111000
ABC/3TC + ATV/r: Randomized Phase720002

[back to top]

Percentage of Participants Who Achieved HIV-1 RNA <400 c/ml at the Week 84 Visit

Percentage of PAR with HIV-1 RNA <400 c/ml at Week 84 was tabulated; stratified by baseline HIV-1 RNA (<100,000 and >=100,000 c/ml). Per TLOVR algorithm, responders were PAR with confirmed (CF) HIV-RNA <400 c/ml who had not met any non-responder (NR) criterion. NR were PAR who never achieved CF HIV RNA <400 c/ml, prematurely discontinued (DC) study or study medication (Med), had CF rebound to >=400 c/ml, or had an unconfirmed HIV RNA >=400 c/ml at last visit. Observed analysis (Obs): all observed data. M/D=F analysis: PAR with missing data/data collected after study Med DC were failures. (NCT00440947)
Timeframe: Week 84

,
Interventionpercentage of participants (Number)
TLOVRObsM/D=F
ABC/3TC + ATV: Randomized Phase929992
ABC/3TC + ATV/r: Randomized Phase869887

[back to top]

Number of Confirmed Virologic Failure Participants With Treatment-emergent HIV Genotypic Resistance in Reverse Transcriptase and Protease From Baseline Through Week 36

A blood sample was drawn for participants failing to respond to therapy, and the mutations present in the virus were identified. For each participant, the mutations found at the time of failure were compared with any mutations found in the blood sample at baseline. New resistance-associated mutations (defined by the International AIDS Society-USA guidelines) that developed at the time of failure were tabulated by drug class. PAR, participants; VF, virologic failure; NRTI, nucleoside reverse transcriptase inhibitor; NNRTI, non-nucleoside reverse transcriptase inhibitor; PI, protease inhibitor. (NCT00440947)
Timeframe: Baseline through Week 36

Interventionparticipants (Number)
PAR with paired genotypes at baseline and VFPAR with treatment-emergent mutationsPAR with NRTI mutationsPAR with NNRTI mutationsPAR with major PI mutationsPAR with minor PI mutations
ABC/3TC + ATV/r: Induction Phase1564102

[back to top]

Percentage of Participants Who Achieved Plasma HIV-1 RNA <400 c/ml at the Week 36 Visit

The percentage of PAR with HIV-1 RNA virus <400 c/ml from a Week 36 blood sample was tabulated. Per TLOVR algorithm, responders were PAR with confirmed (CF) HIV RNA <400 c/ml who had not met any non-responder criterion. Non-responders were PAR who never achieved CF HIV RNA <400 c/ml, prematurely discontinued (DC) study or study medication (Med; any reason), had CF rebound to >=400 c/ml, or had an unconfirmed HIV RNA >=400 c/ml at last visit. ITT-E observed analysis (Obs): all observed data. ITT-E M/D=F analysis: PAR with missing data/data collected after study Med DC were failures. (NCT00440947)
Timeframe: Week 36

Interventionpercentage of participants (Number)
TLOVRObsM/D=F
ABC/3TC + ATV/r: Induction Phase829884

[back to top]

Number of Confirmed Virologic Failure Participants From Week 84 Through Week 144 With Treatment-emergent Reductions in HIV Susceptibility to Abacavir, Lamivudine, Atazanavir, or Ritonavir

A blood sample was drawn for participants failing to respond to therapy, and changes in drug susceptibility for HIV isolated from the participants for each drug used in the study were assessed. For each participant, the changes in drug susceptibility detected by phenotypic assay in virus from the sample collected at the time of failure was compared with drug susceptibility in the virus from the blood sample at baseline. PAR, participant. (NCT00440947)
Timeframe: Week 84 through Week 144

,
Interventionparticipants (Number)
PAR with reduced abacavir susceptibilityPAR with reduced lamivudine susceptibilityPAR with reduced atazanavir susceptibilityPAR with reduced ritonavir susceptibility
ABC/3TC + ATV: Extension Phase0100
ABC/3TC + ATV/r: Extension Phase1111

[back to top]

Number of Confirmed Virologic Failure Participants From Randomization at Week 36 Through Week 84 With Treatment-emergent Reductions in HIV Susceptibility to Abacavir, Lamivudine, Atazanavir, or Ritonavir

A blood sample was drawn for participants failing to respond to therapy, and changes in drug susceptibility for HIV isolated from the participants for each drug used in the study were assessed. For each participant, the changes in drug susceptibility detected by phenotypic assay in virus from the sample collected at the time of failure was compared with drug susceptibility in the virus from the blood sample at baseline. PAR, participant. (NCT00440947)
Timeframe: Randomization at Week 36 through Week 84

,
Interventionparticipants (Number)
PAR with reduced abacavir susceptibilityPAR with reduced lamivudine susceptibilityPAR with reduced atazanavir susceptibilityPAR with reduced ritonavir susceptibility
ABC/3TC + ATV: Randomized Phase0100
ABC/3TC + ATV/r: Randomized Phase0000

[back to top]

Number of Confirmed Virologic Failure Participants From Baseline Through Week 36 With Treatment-emergent Reductions in Susceptibility to Abacavir, Lamivudine, Atazanavir, or Ritonavir

A blood sample was drawn for participants failing to respond to therapy, and changes in drug susceptibility for HIV isolated from the participants for each drug used in the study were assessed. For each participant, the changes in drug susceptibility detected by phenotypic assay in virus from the sample collected at the time of failure was compared with drug susceptibility in the virus from the blood sample at baseline. PAR, participant. (NCT00440947)
Timeframe: Baseline through Week 36

Interventionparticipants (Number)
PAR with reduced abacavir susceptibilityPAR with reduced lamivudine susceptibilityPAR with reduced atazanavir susceptibilityPAR with reduced ritonavir susceptibility
ABC/3TC + ATV/r: Induction Phase0100

[back to top]

Mean Percent Compliance at Week 36

Percent compliance is defined as the total number of pills taken divided by the total number of pills prescribed. The total number of pills taken was calculated by subtracting any returned pills from the total number of pills that were dispensed to each participant during this period. Compliance was calculated for each medication in the regimen. (NCT00440947)
Timeframe: Week 36

Interventionpercent compliance (Mean)
Abacavir/LamivudineRitonavirAtazanavir
ABC/3TC + ATV/r: Induction Phase92.292.392.7

[back to top]

Mean Percent Compliance at Week 144

Percent compliance is defined as the total number of pills taken divided by the total number of pills prescribed. The total number of pills taken was calculated by subtracting any returned pills from the total number of pills that were dispensed to each participant during this period. Compliance was calculated for each medication in the regimen. (NCT00440947)
Timeframe: Week 144

,
Interventionpercent compliance (Mean)
Abacavir/LamivudineRitonavirAtazanavir
ABC/3TC + ATV: Extension Phase92.093.399.1
ABC/3TC + ATV/r: Extension Phase90.190.191.4

[back to top]

Percentage of Participants (PAR) Who Achieved Plasma HIV-1 RNA <50 Copies (c) /Milliliter (ml) at the Week 84 Visit

The percentage of PAR with HIV-1 RNA virus <50 c/ml determined from a blood sample drawn at Week 84 was tabulated by treatment arm with stratification by baseline HIV-1 RNA (<100,000 c/ml and >=100,000 c/ml). Per TLOVR algorithm, responders were PAR with confirmed viral load <50 c/ml who had not met any non-responder criterion. Non-responders were PAR who never achieved confirmed HIV RNA <50 c/ml, prematurely discontinued study or study medication for any reason, had confirmed rebound to at least 50 c/ml, or had an unconfirmed HIV RNA of at least 50 c/ml at last visit. (NCT00440947)
Timeframe: Week 84

Interventionpercentage of participants (Number)
ABC/3TC + ATV: Randomized Phase86
ABC/3TC + ATV/r: Randomized Phase81

[back to top]

Number of Patients With Clinical Adverse Experiences (CAEs) Through 24 Weeks

An adverse experience is defined as any unfavorable and unintended change in the structure, function, or chemistry of the body temporally associated with the use of the SPONSOR'S (Merck & Co., Inc.) product, whether or not considered related to the use of the product (NCT00443703)
Timeframe: 24 Week last patient last visit

,
InterventionParticipants (Number)
With CAEsWithout CAEs
KALETRA™ 400/100 mg b.i.d.10668
MK0518 400 mg b.i.d.10965

[back to top]

Mean Percent Change From Baseline in Fasting Serum Cholesterol at Week 12

(NCT00443703)
Timeframe: Baseline and Week 12

InterventionPercent Change (Mean)
MK0518 400 mg b.i.d.-12.83
KALETRA™ 400/100 mg b.i.d.0.70

[back to top]

Mean Percent Change From Baseline in Fasting Serum Cholesterol at Week 24

(NCT00443703)
Timeframe: Baseline and Week 24

InterventionPercent Change (Mean)
MK0518 400 mg b.i.d.-13.81
KALETRA™ 400/100 mg b.i.d.2.70

[back to top]

Mean Percent Change From Baseline in Fasting Serum High-Density Lipoprotein Cholesterol (HDL-C) at Week 12

(NCT00443703)
Timeframe: Baseline and Week 12

InterventionPercent Change (Mean)
MK0518 400 mg b.i.d.-0.86
KALETRA™ 400/100 mg b.i.d.0.78

[back to top]

Mean Percent Change From Baseline in Fasting Serum Low-Density Lipoprotein Cholesterol (LDL-C) at Week 12

(NCT00443703)
Timeframe: Baseline and Week 12

InterventionPercent Change (Mean)
MK0518 400 mg b.i.d.-2.43
KALETRA™ 400/100 mg b.i.d.2.05

[back to top]

Mean Percent Change From Baseline in Fasting Serum Low-Density Lipoprotein Cholesterol (LDL-C) at Week 24

(NCT00443703)
Timeframe: Baseline and Week 24

InterventionPercent Change (Mean)
MK0518 400 mg b.i.d.-1.12
KALETRA™ 400/100 mg b.i.d.8.54

[back to top]

Mean Percent Change From Baseline in Non-High-Density Lipoprotein Cholesterol (Non-HDL-C) at Week 12

(NCT00443703)
Timeframe: Baseline and Week 12

InterventionPercent Change (Mean)
MK0518 400 mg b.i.d.-15.17
KALETRA™ 400/100 mg b.i.d.2.31

[back to top]

Mean Percent Change From Baseline in Non-High-Density Lipoprotein Cholesterol (Non-HDL-C) at Week 24

(NCT00443703)
Timeframe: Baseline and Week 24

InterventionPercent Change (Mean)
MK0518 400 mg b.i.d.-15.03
KALETRA™ 400/100 mg b.i.d.5.53

[back to top]

Median Percent Change From Baseline in Serum Triglyceride at Week 12

Standard Deviation (Robust): calculated as interquartile range (IQR)/1.075, where IQR=3rd quartile-1st quartile. (NCT00443703)
Timeframe: Baseline and Week 12

InterventionPercent Change (Median)
MK0518 400 mg b.i.d.-41.50
KALETRA™ 400/100 mg b.i.d.3.56

[back to top]

Median Percent Change From Baseline in Serum Triglyceride at Week 24

Standard Deviation (Robust): calculated as interquartile range (IQR)/1.075, where IQR=3rd quartile-1st quartile. (NCT00443703)
Timeframe: Baseline and Week 24

InterventionPercent Change (Median)
MK0518 400 mg b.i.d.-44.53
KALETRA™ 400/100 mg b.i.d.6.14

[back to top]

Number of Patients With Plasma Human Immunodeficiency Virus (HIV) RiboNucleic Acid (RNA) <50 Copies/mL at Week 24

(NCT00443703)
Timeframe: Week 24

InterventionParticipants (Number)
MK0518 400 mg b.i.d.139
KALETRA™ 400/100 mg b.i.d.152

[back to top]

Number of Patients That Died by 24 Week Last Patient Last Visit

(NCT00443703)
Timeframe: 24 Week last patient last visit

,
Interventionparticipants (Number)
DiedDid Not Die
KALETRA™ 400/100 mg b.i.d.0174
MK0518 400 mg b.i.d.0174

[back to top]

Number of Patients That Discontinued Due to CAEs Through 24 Weeks

(NCT00443703)
Timeframe: 24 Week last patient last visit

,
Interventionparticipants (Number)
Discontinued with CAEsDid not Discontinue with CAEs
KALETRA™ 400/100 mg b.i.d.4170
MK0518 400 mg b.i.d.4170

[back to top] [back to top]

Number of Patients That Discontinued Due to LAEs Through 24 Weeks

(NCT00443703)
Timeframe: 24 Week last patient last visit

,
Interventionparticipants (Number)
Discontinued with LAEsDid Not Discontinue with LAEs
KALETRA™ 400/100 mg b.i.d.1173
MK0518 400 mg b.i.d.2172

[back to top] [back to top] [back to top] [back to top]

Number of Patients With Serious CAEs Through 24 Weeks

Serious CAEs are any AEs occurring at any dose that; results in death; or is life threatening; or results in a persistent or significant disability/incapacity; or results in or prolongs an existing inpatient hospitalization; or is a congenital anomaly/birth defect; or is a cancer; or is an overdose (NCT00443703)
Timeframe: 24 Week last patient last visit

,
Interventionparticipants (Number)
With Serious CAEsWithout Serious CAEs
KALETRA™ 400/100 mg b.i.d.10164
MK0518 400 mg b.i.d.15159

[back to top]

Number of Patients With Laboratory Adverse Experiences (LAEs) Through 24 Weeks

A laboratory adverse experience (LAE) is defined as any unfavorable and unintended change in the chemistry of the body temporally associated with the use of the SPONSOR'S (Merck & Co., Inc.) product, whether or not considered related to the use of the product (NCT00443703)
Timeframe: 24 Week last patient last visit

,
Interventionparticipants (Number)
With LAEsWithout LAEs
KALETRA™ 400/100 mg b.i.d.7167
MK0518 400 mg b.i.d.11163

[back to top]

Number of Patients With Serious LAEs Through 24 Weeks

Serious LAEs are any LAEs occurring at any dose that; results in death; or is life threatening; or results in a persistent or significant disability/incapacity; or results in or prolongs an existing inpatient hospitalization; or is a congenital anomaly/birth defect; or is a cancer; or is an overdose (NCT00443703)
Timeframe: 24 Week last patient last visit

,
Interventionparticipants (Number)
With LAEsWithout LAEs
KALETRA™ 400/100 mg b.i.d.0174
MK0518 400 mg b.i.d.0174

[back to top]

Mean Percent Change From Baseline in Fasting Serum High-Density Lipoprotein Cholesterol (HDL-C) at Week 24

(NCT00443703)
Timeframe: Baseline and Week 24

InterventionPercent Change (Mean)
MK0518 400 mg b.i.d.-4.42
KALETRA™ 400/100 mg b.i.d.-1.70

[back to top] [back to top]

Mean Percent Change From Baseline in Fasting Serum High-density Lipoprotein Cholesterol (HDL-C) at Week 24

(NCT00443729)
Timeframe: Baseline and Week 24

InterventionPercent Change (Mean)
MK0518 400 mg b.i.d.-1.77
KALETRA™ 400/100 mg b.i.d.-0.15

[back to top]

Mean Percent Change From Baseline in Fasting Serum Low-density Lipoprotein Cholesterol (LDL-C) at Week 12

(NCT00443729)
Timeframe: Baseline and Week 12

InterventionPercent Change (Mean)
MK0518 400 mg b.i.d.3.99
KALETRA™ 400/100 mg b.i.d.0.55

[back to top]

Mean Percent Change From Baseline in Fasting Serum Low-density Lipoprotein Cholesterol (LDL-C) at Week 24

(NCT00443729)
Timeframe: Baseline and Week 24

InterventionPercent Change (Mean)
MK0518 400 mg b.i.d.5.12
KALETRA™ 400/100 mg b.i.d.6.06

[back to top]

Mean Percent Change From Baseline in Non-high-density Lipoprotein Cholesterol (Non-HDL-C) at Week 12

(NCT00443729)
Timeframe: Baseline and Week 12

InterventionPercent Change (Mean)
MK0518 400 mg b.i.d.-14.77
KALETRA™ 400/100 mg b.i.d.2.91

[back to top]

Mean Percent Change From Baseline in Non-high-density Lipoprotein Cholesterol (Non-HDL-C) at Week 24

(NCT00443729)
Timeframe: Baseline and Week 24

InterventionPercent Change (Mean)
MK0518 400 mg b.i.d.-15.83
KALETRA™ 400/100 mg b.i.d.5.26

[back to top]

Median Percent Change From Baseline in Serum Triglyceride at Week 12

Standard Deviation (Robust): calculated as interquartile range (IQR)/1.075, where IQR=3rd quartile-1st quartile. (NCT00443729)
Timeframe: Baseline and Week 12

InterventionPercent Change (Median)
MK0518 400 mg b.i.d.-42.82
KALETRA™ 400/100 mg b.i.d.8.20

[back to top]

Median Percent Change From Baseline in Serum Triglyceride at Week 24

Standard Deviation (Robust): calculated as interquartile range (IQR)/1.075, where IQR=3rd quartile-1st quartile. (NCT00443729)
Timeframe: Baseline and Week 24

InterventionPercent Change (Median)
MK0518 400 mg b.i.d.-44.50
KALETRA™ 400/100 mg b.i.d.7.06

[back to top]

Number of Patients With Plasma Human Immunodeficiency Virus (HIV) RiboNucleic Acid (RNA) <50 Copies/mL at Week 24

(NCT00443729)
Timeframe: 24 Weeks

InterventionParticipants (Number)
MK0518 400 mg b.i.d.154
KALETRA™ 400/100 mg b.i.d.167

[back to top]

Number of Patients That Died by 24 Week Last Patient Last Visit

(NCT00443729)
Timeframe: 24 Week last patient last visit

,
InterventionParticipants (Number)
DiedDid Not Die
KALETRA™ 400/100 mg b.i.d.0178
MK0518 400 mg b.i.d.0176

[back to top]

Number of Patients That Discontinued Due to CAEs Through 24 Weeks

(NCT00443729)
Timeframe: 24 Week last patient last visit

,
InterventionParticipants (Number)
Discontinued with CAEsDid Not Discontinue with CAEs
KALETRA™ 400/100 mg b.i.d.0178
MK0518 400 mg b.i.d.0176

[back to top]

Number of Patients That Discontinued Due to LAEs Through 24 Weeks

(NCT00443729)
Timeframe: 24 Week last patient last visit

,
InterventionParticipants (Number)
Discontinued with LAEsDid Not Discontinue with LAEs
KALETRA™ 400/100 mg b.i.d.0178
MK0518 400 mg b.i.d.0176

[back to top]

Number of Patients With Clinical Adverse Experiences (CAEs) Through 24 Weeks

(NCT00443729)
Timeframe: 24 Week last patient last visit

,
InterventionParticipants (Number)
With CAEsWithout CAEs
KALETRA™ 400/100 mg b.i.d.11266
MK0518 400 mg b.i.d.12353

[back to top] [back to top] [back to top]

Number of Patients With Laboratory Adverse Experiences (LAEs) Through 24 Weeks

(NCT00443729)
Timeframe: 24 Week last patient last visit

,
InterventionParticipants (Number)
With LAEsWithout LAEs
KALETRA™ 400/100 mg b.i.d.6172
MK0518 400 mg b.i.d.8168

[back to top]

Number of Patients With Serious CAEs Through 24 Weeks

Serious CAEs are any AEs occurring at any dose that; results in death; or is life threatening; or results in a persistent or significant disability/incapacity; or results in or prolongs an existing inpatient hospitalization; or is a congenital anomaly/birth defect; or is a cancer; or is an overdose (NCT00443729)
Timeframe: 24 Week last patient last visit

,
InterventionParticipants (Number)
With Serious CAEsWithout Serious CAEs
KALETRA™ 400/100 mg b.i.d.8170
MK0518 400 mg b.i.d.4172

[back to top] [back to top]

Number of Patients With Serious LAEs Through 24 Weeks

Serious LAEs are any LAEs occurring at any dose that; results in death; or is life threatening; or results in a persistent or significant disability/incapacity; or results in or prolongs an existing inpatient hospitalization; or is a congenital anomaly/birth defect; or is a cancer; or is an overdose (NCT00443729)
Timeframe: 24 Week last patient last visit

,
InterventionParticipants (Number)
With LAEsWithout LAEs
KALETRA™ 400/100 mg b.i.d.0178
MK0518 400 mg b.i.d.0176

[back to top]

Mean Percent Change From Baseline in Fasting Serum Cholesterol at Week 12

(NCT00443729)
Timeframe: Baseline and Week 12

InterventionPercent Change (Mean)
MK0518 400 mg b.i.d.-12.41
KALETRA™ 400/100 mg b.i.d.1.29

[back to top]

Mean Percent Change From Baseline in Fasting Serum Cholesterol at Week 24

(NCT00443729)
Timeframe: Baseline and Week 24

InterventionPercent Change (Mean)
MK0518 400 mg b.i.d.-13.64
KALETRA™ 400/100 mg b.i.d.3.55

[back to top]

Mean Percent Change From Baseline in Fasting Serum High-density Lipoprotein Cholesterol (HDL-C) at Week 12

(NCT00443729)
Timeframe: Baseline and Week 12

InterventionPercent Change (Mean)
MK0518 400 mg b.i.d.-0.64
KALETRA™ 400/100 mg b.i.d.-2.50

[back to top]

Frequency of Patients (%) With Possible Clinically Significant Abnormalities of Laboratory Measurements

Frequency of patients (%) with possible clinically significant abnormalities of laboratory measurements (haematology, differentials (automatic and absolute), coagulation, electrolytes, enzymes, substrates, urinalysis, serology and T-cells) (NCT00447902)
Timeframe: Baseline through 48 weeks

,
Interventionpercentage of participants (Number)
Hematocrit - decreaseRed Blood Cell count - decreaseProthrombin time - increaseAST/GOT, SGOT - increaseALT/GPT, SGPT - increaseLipase - increaseBilirubin, total - increaseBilirubin, direct - increaseTriglyceride - increase
Standard of Care25.025.0050.025.00000
Therapeutic Drug Monitoring16.7020.033.333.316.716.716.725.0

[back to top]

Steady-state Levels of Amprenavir (APV) and Ritonavir (RTV) Ctau at Weeks 4, 12, and 24

Blood samples were drawn at Weeks 4, 12, and 24 to determine plasma concentrations (Ctau) of APV and RTV (NCT00450580)
Timeframe: Weeks 4, 12, and 24

,
Interventionmicrograms/mL (Geometric Mean)
Week 4 APV CtauWeek 12 APV CtauWeek 24 APV CtauWeek 4 RTV CtauWeek 12 RTV CtauWeek 24 RTV Ctau
FPV/RTV 1400/100 mg Once Daily (QD) + ABC/3TC FDC 600/300 mg Q1.110.9131.080.03690.02850.0363
FPV/RTV 700/100 mg BID + ABC/3TC FDC 600/300 mg QD1.991.872.000.1660.1750.170

[back to top]

Number of Participants With HIV-1 RNA <400 Copies/mL (Primary Endpoint) at Week 48 Categorised by Baseline Viral Load, TLOVR Analysis

The number of participants with HIV-1 RNA <400 copies/mL at Week 48 was determined (by analysis of blood draw) and categorised by baseline viral load (BVL). (NCT00450580)
Timeframe: Week 48

,
InterventionParticipants (Number)
<50000 cp/mL (n=35, 40)>=50000 to <100000 cp/mL (n=21, 19)>=100000 to <200000 cp/mL (n=25, 17)>=200000 cp/mL (n=25, 30)
FPV/RTV 1400/100 mg Once Daily (QD) + ABC/3TC FDC 600/300 mg Q26182121
FPV/RTV 700/100 mg BID + ABC/3TC FDC 600/300 mg QD37151124

[back to top]

Number of Protocol-defined Virological Failures With Genotypic and Phenotypic Resistance Changes

A blood sample was drawn at the time of confirmation of virological failure, and mutations present in the virus were identified and compared to those found in the blood sample at baseline. New mutations were tabulated by drug class. RT, reverse transcriptase. Virological failure could occur anytime from Week 4 to Week 48. (NCT00450580)
Timeframe: Time to virologic failure; Week 4 up to Week 48

,
InterventionParticipants (Number)
Treatment-Emergent Major HIV RT Mutations (M184V)Treatment-Emergent Major HIV Protease Mutations
FPV/RTV 1400/100 mg Once Daily (QD) + ABC/3TC FDC 600/300 mg Q10
FPV/RTV 700/100 mg BID + ABC/3TC FDC 600/300 mg QD00

[back to top]

Percentage of Participants With HIV-1 RNA <400 and >=400 Copies/mL Over 48 Weeks

A blood sample was drawn to determine the amount of HIV-1 RNA virus in copies/mL at week 48. The percentage of participants with HIV-1 RNA <400 copies/mL at Week 48 was determined by the Time to Loss Of Virologic Response (TLOVR) algorithm. (NCT00450580)
Timeframe: Week 48

,
InterventionPercentage of participants (Number)
HIV-1 RNA <400 copies/mLHIV-1 RNA >=400 copies/mL
FPV/RTV 1400/100 mg Once Daily (QD) + ABC/3TC FDC 600/300 mg Q8119
FPV/RTV 700/100 mg BID + ABC/3TC FDC 600/300 mg QD8218

[back to top]

Number of Participants With HIV-1 RNA <400 Copies/mL (Primary Endpoint) at Week 48 Categorised by Baseline CD4+ Count, TLOVR Analysis

The number of participants with HIV-1 RNA <400 copies/mL at week 48 was determined (by analysis of blood draw) and categorised by baseline CD4+ count. (NCT00450580)
Timeframe: Week 48

,
InterventionParticipants (Number)
<150 cells/mm3 (n=24, 23)>=150 to <250 cells/mm3 (n=29, 31)>=250 to <350 cell/mm3 (n=29, 31)>=350 cell/mm3 (n=24, 21)
FPV/RTV 1400/100 mg Once Daily (QD) + ABC/3TC FDC 600/300 mg Q18242321
FPV/RTV 700/100 mg BID + ABC/3TC FDC 600/300 mg QD16262619

[back to top]

Percentage of Participants With HIV-1 RNA <50 and >=50 Copies/mL by Visit Over 48 Weeks

A blood sample was drawn to determine the amount of HIV-1 RNA virus in copies/mL at week 48. The percentage of participants with HIV-1 RNA <50 copies/mL at Week 48 was determined by the TLOVR algorithm (NCT00450580)
Timeframe: Week 48

,
InterventionPercentage of participants (Number)
HIV-1 RNA <50 copies/mLHIV-1 RNA >=50 copies/mL
FPV/RTV 1400/100 mg Once Daily (QD) + ABC/3TC FDC 600/300 mg Q7624
FPV/RTV 700/100 mg BID + ABC/3TC FDC 600/300 mg QD7723

[back to top]

Change From Baseline in Non-HDL Cholesterol at Week 48

Blood samples were drawn to determine the non-HDL cholesterol levels at Week 48. The mean absolute change in non-HDL cholesterol was defined as the Week 48 levels minus levels at baseline. (NCT00450580)
Timeframe: Week 48

Interventionmmol/L (millimoles/Liter) (Mean)
FPV/RTV 1400/100 mg Once Daily (QD) + ABC/3TC FDC 600/300 mg Q1.10
FPV/RTV 700/100 mg BID + ABC/3TC FDC 600/300 mg QD1.26

[back to top]

Median Change From Baseline in LDL Cholesterol at Week 48.

(NCT00460746)
Timeframe: Week 48

Interventionmg/dL (Median)
Baseline (Day 1)Week 48 Change from Baseline
Darunavir(TMC114)/Eravirine(TMC125)100.0-9.0

[back to top]

CD4+ Cell Count (x 10^6 Cell/L): Baseline and Mean Changes From Baseline at 4, 8, 12, 16, 24,36 and 48 Weeks.

(NCT00460746)
Timeframe: Week 48

Interventioncells/mm^3 (Mean)
Baseline (Day 1)Week 4 Change from BaselineWeek 8 Change from BaselineWeek 12 Change from BaselineWeek 16 Change from BaselineWeek 24 Change from BaselineWeek 36 Change from BaselineWeek 48 Change from Baseline
Darunavir(TMC114)/Eravirine(TMC125)338.32.3-25.0-18.90.524.522.747.3

[back to top]

CD4+ Cell Count (x 10^6 Cell/L): Baseline and Median Changes From Baseline at 4, 8, 12, 16, 24, 36 and 48 Weeks.

(NCT00460746)
Timeframe: Week 48

Interventioncells/mm^3 (Median)
Baseline (Day 1)Week 4 Change from BaselineWeek 8 Change from BaselineWeek 12 Change from BaselineWeek 16 Change from BaselineWeek 24 Change from BaselineWeek 36 Change from BaselineWeek 48 Change from Baseline
Darunavir(TMC114)/Eravirine(TMC125)301.0-12.0-32.0-17.0-7.019.038.063.5

[back to top]

Median Change From Baseline in HDL Cholesterol.

(NCT00460746)
Timeframe: Week 48

Interventionmg/dL (Median)
Baseline (Day 1)Week 48 Change from Baseline
Darunavir(TMC114)/Eravirine(TMC125)47.5-2.5

[back to top]

Median Change From Baseline in Total Cholesterol (TC) / High Denisty Lipoprotein (HDL) Ratio at Week 48.

(NCT00460746)
Timeframe: Week 48

Interventionratio of TC and HDL (Median)
Baseline (Day 1)Week 48 Change from Baseline
Darunavir(TMC114)/Eravirine(TMC125)4.7-0.3

[back to top]

Median Change From Baseline in Total Cholesterol at Week 48.

(NCT00460746)
Timeframe: Week 48

Interventionmg/dL (Median)
Baseline (Day 1)Week 48 Change from Baseline
Darunavir(TMC114)/Eravirine(TMC125)189.0-27.5

[back to top]

Median Change From Baseline in Triglycerides at Week 48.

(NCT00460746)
Timeframe: Week 48

Interventionmg/dL (Median)
Baseline (Day 1)Week 48 Change from Baseline
Darunavir(TMC114)/Eravirine(TMC125)229.0-24.5

[back to top]

Proportion of Patients Who Have Viral Load Measurements <50 Copies/ml at 2, 4, 8, 12, 16, 24, 36 and 48 Weeks After Switching to DRV/r and ETR, Missing Equals Failure.

(NCT00460746)
Timeframe: 48 weeks

Interventionpercentage of participants (Number)
Baseline (Day 1)Week 2Week 4Week 8Week 12Week 16Week 24Week 36Week 48Post-Treatment Follow Up
Darunavir(TMC114)/Eravirine(TMC125)100909090809090908090

[back to top]

Proportion of Patients Who Maintain Plasma HIV Viral Load Measurements < 400 Copies/ml at 2, 4, 8, 12, 16, 24, 36 and 48 Weeks After Switching to DRV/r and ETR, Missing Equals Failure.

(NCT00460746)
Timeframe: 48 weeks

Interventionpercentage of participants (Number)
Baseline (Day 1)Week 2Week 4Week 8Week 12Week 16Week 24Week 36Week 48Post-Treatment Follow Up
Darunavir(TMC114)/Eravirine(TMC125)10010010090809090908090

[back to top]

Median Change From Baseline in Glucose at Week 48.

(NCT00460746)
Timeframe: Week 48

Interventionmg/dL (Median)
Baseline (Day 1)Week 48 Change from Baseline
Darunavir(TMC114)/Eravirine(TMC125)92.51.0

[back to top]

Gemfibrozil Area Under the Concentration vs. Time Curve (AUC)

AUC (ng*hr/mL) of gemfibrozil when given as a 600 mg dose by itself compared to gemfibrozil AUC after 14.5 days of lopinavir-ritonavir (400mg/100mg) twice daily. (NCT00474201)
Timeframe: 22 days per subject (approximately 1 year for entire study completion)

Interventionng*hr/mL (Geometric Mean)
Gemfibrozil Alone (Control Group)104
Gemfibrozil + Lopinavir-ritonavir62

[back to top]

Area Under the Curve of BUP/NLX With TPV/r (h*ng/mL)

Non-compartmental methods were used for pharmacokinetic analysis. The area under the plasma drug concentration-time curve was estimated by linear-log trapezoidal rule at 24-hrs. (NCT00486330)
Timeframe: 10 days

Interventionh*ng/mL (Geometric Mean)
Tipranavir/Ritonavir (500mg/200mg)43.7

[back to top]

Time-averaged Difference (DAVG) of log10 Plasma Viral Load Over 48 Weeks

(NCT00524368)
Timeframe: 48 weeks

Interventionlog10 copies/mL (Least Squares Mean)
DRV/Rtv 800/100 mg Once Daily-1.77
DRV/Rtv 600/100 mg Twice Daily-1.74

[back to top]

Virologic Response at Week 48 (Viral Load Less Than 400 Copies/mL)

Number of participants with confirmed plasma viral load less than 400 copies/mL at Week 48. (NCT00524368)
Timeframe: 48 weeks

InterventionParticipants (Number)
DRV/Rtv 800/100 mg Once Daily226
DRV/Rtv 600/100 mg Twice Daily227

[back to top]

Virological Response at Week 48 (Number of Participants With Plasma Viral Load Less Than 50 Copies/mL) - as Defined by the Time to Loss of Virologic Response (TLOVR) Algorithm

The TLOVR algorithm was used to derive response, ie, response and loss of response needed to be confirmed at 2 consecutive visits and participants who permanently discontinued were considered nonresponders after discontinuation. Participants with intermittent missing viral load values were considered responders if the preceeding and succeeding visits indicated response. In all other cases, intermittent values were imputed with nonresponse. Resuppression after confirmed virologic failure was considered as failure in this algorithm. (NCT00524368)
Timeframe: 48 Weeks

InterventionParticipants (Number)
DRV/Rtv 800/100 mg Once Daily212
DRV/Rtv 600/100 mg Twice Daily210

[back to top]

Predose Plasma Concentration (C0h) of DRV and Rtv.

Pharmacokinetic parameter C0h was assessed. Population Pharmacokinetic Estimates of DRV and rtv were evaluated. (NCT00524368)
Timeframe: 0 hour predose and 1 hour post dose measured at Weeks 4 and 24. Any time point measured at Weeks 8 and 48

,
Interventionng/mL (Median)
Population Pharmacokinetic Estimates of DRVPopulation Pharmacokinetic Estimates of rtv
DRV/Rtv 600/100 mg Twice Daily3197307
DRV/Rtv 800/100 mg Once Daily189659

[back to top]

Percentage of Participants Adherent/Non-adherent to ARV as Determined by Modified Medication Adherence Self Report Inventory (M-MASRI) Questionnaire at Week 48

Self-reported adherence to the ARV medications was measured. The M-MASRI asks participants to report the number of doses taken, as well as the number of doses taken during the last 30 days prior to the study visit by means of a horizontal visual analogue scale (VAS) that generates a self-rated percentage of doses of all the ARV medications taken during the past 30 days. (NCT00524368)
Timeframe: 48 weeks

,
InterventionPercentage of participants (Number)
AdherentNon-adherent
DRV/Rtv 600/100 mg Twice Daily60.339.7
DRV/Rtv 800/100 mg Once Daily67.432.6

[back to top]

Number of Participants Developing Mutations at Endpoint

Development of Mutations in Virologic Failures (Plasma Viral Load less than 50 Copies/mL) at endpoint. (NCT00524368)
Timeframe: 48 weeks

,
InterventionParticipants (Number)
DRV resistance-associated mutation (RAM)Primary (major) protease inhibitor (PI) mutationsProtease inhibitor (PI) RAMsNucleoside reverse transcriptase inhibitor RAMs
DRV/Rtv 600/100 mg Twice Daily0043
DRV/Rtv 800/100 mg Once Daily1174

[back to top]

Change in log10 Viral Load From Baseline at Week 48

(NCT00524368)
Timeframe: 48 weeks

,
Interventionlog10 copies/mL (Median)
Log10 Viral Load at baselineLog10 Viral Load change from baseline at Week 48
DRV/Rtv 600/100 mg Twice Daily4.134-2.13
DRV/Rtv 800/100 mg Once Daily4.23-2.11

[back to top]

Time to Loss of Virologic Response

Time taken to lose the virologic response ie, plasma viral load less than 50 copies/mL by participants. (NCT00524368)
Timeframe: 48 weeks

InterventionDays (Mean)
DRV/Rtv 800/100 mg Once Daily250.235
DRV/Rtv 600/100 mg Twice Daily281.743

[back to top]

Area Under the Curve From the Time of Study Medication Administration Upto 24 Hour Postdose (AUC24h) of DRV and Rtv

Pharmacokinetic parameter AUC24h was assessed from the time of study medication administration upto 24 hour postdose. Population Pharmacokinetic Estimates of DRV and rtv were evaluated. (NCT00524368)
Timeframe: 0 hour predose and 1 hour post dose measured at Weeks 4 and 24. Any time point measured at Weeks 8 and 48.

,
Interventionng*h/mL (Median)
Population Pharmacokinetic Estimates of DRVPopulation Pharmacokinetic Estimates of rtv
DRV/Rtv 600/100 mg Twice Daily10940112588
DRV/Rtv 800/100 mg Once Daily877885776

[back to top]

Change in CD4+ Cell Count From Baseline

CD4+ cell count was calculated using the Last Observation Carried Forward (LOCF) algorithm. (NCT00524368)
Timeframe: 48 Weeks

,
Intervention10e6/l (Median)
Value at BaselineCD4+ cell count change from baseline at Week 48
DRV/Rtv 600/100 mg Twice Daily23694
DRV/Rtv 800/100 mg Once Daily219100

[back to top]

Time to Reach First Virologic Response

Time (in weeks) to achieve viral load less than 50 copies/mL by the participants. (NCT00524368)
Timeframe: 48 weeks

InterventionDays (Median)
DRV/Rtv 800/100 mg Once Daily85
DRV/Rtv 600/100 mg Twice Daily85

[back to top]

Change From Baseline in Total Functional Assessment of HIV Infection (FAHI) Score

The FAHI is a 44-item questionnaire and incorporates 5 functional scales (physical well-being, emotional well-being/living with HIV, functional and global well-being, social well-being, and cognitive functioning). Each scale included several questions (all 5 scales include total 44 questions). For each question, participants gave a score of either 0 (not at all), 1 (a little bit), 2 (somewhat), 3 (quite a bit) and 4 (very much). Total FAHI imputed score is calculated by adding scores for each question. The range of total FAHI score is 0 to 176. Higher scores indicate worsening. (NCT00524368)
Timeframe: 48 weeks

,
InterventionScores on a scale (Median)
FAHI score at baselineChange from baseline in FAHI score at Week 48
DRV/Rtv 600/100 mg Twice Daily123.50.0
DRV/Rtv 800/100 mg Once Daily1292.5

[back to top]

The Primary Outcome of This Study is the Proportion of Patients Having Detectable HIV-1 RNA Using the Single Copy Assay After 48 Weeks of Treatment and the Study Hypothesis is That New Treatment is Better Than the Control Group.

(NCT00525733)
Timeframe: 48 weeks

Intervention# subjects without detectable viremia (Number)
3-drug Standard Therapy3
5-drug Experimental Therapy9

[back to top]

Terminal Half-Life (t1/2) of Tipranavir

Tipranavir pharmacokinetics (NCT00530920)
Timeframe: Final (Day 14)

Interventionh (Geometric Mean)
Tipranavir With Ritonavir (TPV/r) 500/200 mg Once Daily4.79
Tipranavir With Ritonavir (TPV/r) 250/100 mg Twice Daily4.51
Tipranavir With Ritonavir (TPV/r) 500/100 mg Twice Daily4.14

[back to top]

Volume of Distribution (V/F) of Ritonavir

Ritonavir pharmacokinetics (NCT00530920)
Timeframe: Final (Day 14)

InterventionL (Geometric Mean)
Tipranavir With Ritonavir (TPV/r) 500/200 mg Once Daily97.5
Tipranavir With Ritonavir (TPV/r) 250/100 mg Twice Daily90.8
Tipranavir With Ritonavir (TPV/r) 500/100 mg Twice Daily88.9

[back to top]

Volume of Distribution (V/F) of Tipranavir

Tipranavir pharmacokinetics (NCT00530920)
Timeframe: Final (Day 14)

InterventionL (Geometric Mean)
Tipranavir With Ritonavir (TPV/r) 500/200 mg Once Daily10.02
Tipranavir With Ritonavir (TPV/r) 250/100 mg Twice Daily9.33
Tipranavir With Ritonavir (TPV/r) 500/100 mg Twice Daily9.39

[back to top]

Clinical Abnormal Findings in Laboratory and Physical Examination

(NCT00530920)
Timeframe: Screening through the end of the study (14 days)

,,
Interventionparticipants (Number)
Alanine aminotransferase (ALT) increasedAspartate aminotransferase (AST) increased
Tipranavir With Ritonavir (TPV/r) 250/100 mg Twice Daily00
Tipranavir With Ritonavir (TPV/r) 500/100 mg Twice Daily11
Tipranavir With Ritonavir (TPV/r) 500/200 mg Once Daily00

[back to top]

Tmax of Ritonavir

Ritonavir pharmacokinetics (NCT00530920)
Timeframe: Final (Day 14)

Interventionh (Geometric Mean)
Tipranavir With Ritonavir (TPV/r) 500/200 mg Once Daily2.84
Tipranavir With Ritonavir (TPV/r) 250/100 mg Twice Daily2.33
Tipranavir With Ritonavir (TPV/r) 500/100 mg Twice Daily2.05

[back to top]

Trough Concentration (Cmin) of Tipranavir

TPV pharmacokinetics (NCT00530920)
Timeframe: Final (Day 13 for QD, Day 14 for BID)

InterventionuM (Geometric Mean)
Tipranavir With Ritonavir (TPV/r) 500/200 mg Once Daily2.8
Tipranavir With Ritonavir (TPV/r) 250/100 mg Twice Daily12.73
Tipranavir With Ritonavir (TPV/r) 500/100 mg Twice Daily21.26

[back to top]

Viral Load (log10 Copies/mL) Change From Baseline (Last Observation Carried Forward (LOCF))

(NCT00530920)
Timeframe: Baseline (Day 0) to Final (Day 14)

InterventionLog10 copies/mL (Median)
Tipranavir With Ritonavir (TPV/r) 500/200 mg Once Daily-1.43
Tipranavir With Ritonavir (TPV/r) 250/100 mg Twice Daily-1.55
Tipranavir With Ritonavir (TPV/r) 500/100 mg Twice Daily-1.47

[back to top]

Apparent Oral Clearance I(Cl/F) of Ritonavir

Ritonavir pharmacokinetics (NCT00530920)
Timeframe: Final (Day 13 for QD, Day 14 for BID)

InterventionL/h (Geometric Mean)
Tipranavir With Ritonavir (TPV/r) 500/200 mg Once Daily39.6
Tipranavir With Ritonavir (TPV/r) 250/100 mg Twice Daily44.4
Tipranavir With Ritonavir (TPV/r) 500/100 mg Twice Daily57.4

[back to top]

Time to Cmax (Tmax) of Tipranavir

Tipranavir pharmacokinetics (NCT00530920)
Timeframe: Final (Day 14)

Interventionh (Geometric Mean)
Tipranavir With Ritonavir (TPV/r) 500/200 mg Once Daily3.07
Tipranavir With Ritonavir (TPV/r) 250/100 mg Twice Daily2.34
Tipranavir With Ritonavir (TPV/r) 500/100 mg Twice Daily2.38

[back to top]

Apparent Oral Clearance I(Cl/F) of Tipranavir

"Tipranavir pharmacokinetics - Clearance (CL) is defined as the dose of a drug divided by the area-under-the-concentration-time curve (AUC), ie. CL = Dose / AUC. For extravascu-lar models the fraction of dose absorbed cannot be estimated, therefore clear-ance for these models is actually Cl/F where F is the fraction of the drug dose which is absorbed." (NCT00530920)
Timeframe: Final (Day 14)

InterventionL/h (Geometric Mean)
Tipranavir With Ritonavir (TPV/r) 500/200 mg Once Daily1.45
Tipranavir With Ritonavir (TPV/r) 250/100 mg Twice Daily1.43
Tipranavir With Ritonavir (TPV/r) 500/100 mg Twice Daily1.57

[back to top]

Area Under the Curve(AUC) of Tipranavir 24 h for Once Daily (QD) and AUC 12 h for Twice Daily (BID)

Tipranavir (TPV) pharmacokinetics (NCT00530920)
Timeframe: Final (Day 13 for QD, Day 14 for BID)

Interventionh*uM (Geometric Mean)
Tipranavir With Ritonavir (TPV/r) 500/200 mg Once Daily571.3
Tipranavir With Ritonavir (TPV/r) 250/100 mg Twice Daily289.3
Tipranavir With Ritonavir (TPV/r) 500/100 mg Twice Daily538.2

[back to top]

AUC 24 of Ritonavir for QD and AUC 12 of Ritonavir for BID

Ritonavir pharmacokinetics (NCT00530920)
Timeframe: Final (Day 13 for QD, Day 14 for BID)

Interventionh*uM (Geometric Mean)
Tipranavir With Ritonavir (TPV/r) 500/200 mg Once Daily5.05
Tipranavir With Ritonavir (TPV/r) 250/100 mg Twice Daily2.25
Tipranavir With Ritonavir (TPV/r) 500/100 mg Twice Daily1.74

[back to top]

Cmax of Ritonavir

Ritonavir pharmacokinetics (NCT00530920)
Timeframe: Visits baseline, 5, 7, 9 and 13 or 14

InterventionuM (Geometric Mean)
Tipranavir With Ritonavir (TPV/r) 500/200 mg Once Daily0.632
Tipranavir With Ritonavir (TPV/r) 250/100 mg Twice Daily0.346
Tipranavir With Ritonavir (TPV/r) 500/100 mg Twice Daily0.291

[back to top]

Concentration-24 Hour (hr) Post Dose of Tipranavir - (Cp 24 h for QD and 12 hr Post Dose (CP 12h) for BID

TPV pharmacokinetics (NCT00530920)
Timeframe: Final (Day 13 for QD, Day 14 for BID)

InterventionuM (Geometric Mean)
Tipranavir With Ritonavir (TPV/r) 500/200 mg Once Daily3.26
Tipranavir With Ritonavir (TPV/r) 250/100 mg Twice Daily10.27
Tipranavir With Ritonavir (TPV/r) 500/100 mg Twice Daily17.75

[back to top]

Cp 24 h of Ritonavir for QD and CP 12 h of Ritonavir for BID

Ritonavir pharmacokinetics (NCT00530920)
Timeframe: Final (Day 13 for QD, Day 14 for BID)

InterventionuM (Geometric Mean)
Tipranavir With Ritonavir (TPV/r) 500/200 mg Once Daily0.005
Tipranavir With Ritonavir (TPV/r) 250/100 mg Twice Daily0.039
Tipranavir With Ritonavir (TPV/r) 500/100 mg Twice Daily0.025

[back to top]

Maximum Concentration (Cmax) of Tipranavir

TPV pharmacokinetics (NCT00530920)
Timeframe: Final (Day 13 for QD, Day 14 for BID)

InterventionuM (Geometric Mean)
Tipranavir With Ritonavir (TPV/r) 500/200 mg Once Daily54.64
Tipranavir With Ritonavir (TPV/r) 250/100 mg Twice Daily36.98
Tipranavir With Ritonavir (TPV/r) 500/100 mg Twice Daily69.66

[back to top]

Terminal Half-Life (t1/2) of Ritonavir

Ritonavir pharmacokinetics (NCT00530920)
Timeframe: Final (Day 14)

InterventionHours (Geometric Mean)
Tipranavir With Ritonavir (TPV/r) 500/200 mg Once Daily1.71
Tipranavir With Ritonavir (TPV/r) 250/100 mg Twice Daily1.42
Tipranavir With Ritonavir (TPV/r) 500/100 mg Twice Daily1.07

[back to top]

Total Cholesterol Over Time

(NCT00531206)
Timeframe: 52 weeks

Interventionmg/dl (Median)
Month 1 (n=42)Month 3 (n=41)Month 6 (n=48)Month 9 (n=37)Month 12 (n=35)
Aptivus® in Combination With Low-dose Norvir®213.0214.0213.0217.0216.0

[back to top]

Triglycerides Over Time

(NCT00531206)
Timeframe: 52 weeks

Interventionmg/dl (Median)
Month 1 (n=38)Month 3 (n=39)Month 6 (n=46)Month 9 (n=33)Month 12 (n=32)
Aptivus® in Combination With Low-dose Norvir®237.5256.9246.0283.4265.5

[back to top]

Use of Lipid Lowering Agents During the Study

(NCT00531206)
Timeframe: 52 weeks

Interventionparticipants (Number)
NoYes
Aptivus® in Combination With Low-dose Norvir®596

[back to top]

Creatinine Over Time

(NCT00531206)
Timeframe: 52 weeks

Interventionmg/dl (Median)
Month 1 (n=43)Month 3 (n=46)Month 6 (n=49)Month 9 (n=35)Month 12 (n=33)
Aptivus® in Combination With Low-dose Norvir®0.900.900.900.900.90

[back to top]

Number of Anti-retroviral Medications Taken in Combination With Tipranavir/Ritonavir

(NCT00531206)
Timeframe: 52 weeks

Interventionparticipants (Number)
< 66 - 1011 - 15> 15missing
Aptivus® in Combination With Low-dose Norvir®30151631

[back to top]

Low Density Lipoprotein (HDL) Cholesterol Over Time

(NCT00531206)
Timeframe: 52 weeks

Interventionmg/dl (Median)
Month 1 (n=23)Month 3 (n=25)Month 6 (n=29)Month 9 (n=26)Month 12 (n=23)
Aptivus® in Combination With Low-dose Norvir®120.0123.0125.0117.5122.0

[back to top]

High Density Lipoprotein (HDL) Cholesterol Over Time

(NCT00531206)
Timeframe: 52 weeks

Interventionmg/dl (Median)
Month 1 (n=28)Month 3 (n=29)Month 6 (n=32)Month 9 (n=28)Month 12 (n=27)
Aptivus® in Combination With Low-dose Norvir®38.039.039.539.037.0

[back to top]

Total Bilirubin Over Time

(NCT00531206)
Timeframe: 52 weeks

Interventionmg/dl (Median)
Month 1 (n=39)Month 3 (n=38)Month 6 (n=43)Month 9 (n=31)Month 12 (n=32)
Aptivus® in Combination With Low-dose Norvir®0.420.400.500.500.40

[back to top]

Adverse Events

The safety and tolerability of the observed antiretroviral therapy were based on the Adverse Events (AEs) and Serious Adverse Events (SAEs) reported in the case report forms. (NCT00531206)
Timeframe: 52 weeks

InterventionNumber of patients with adverse events (Number)
Aptivus® in Combination With Low-dose Norvir®23

[back to top] [back to top]

Deaths

The safety and tolerability of the observed antiretroviral therapy were based on the Adverse Events (AEs) and Serious Adverse Events (SAEs) reported in the case report forms. (NCT00531206)
Timeframe: 52 weeks

InterventionNumber of patients (Number)
Aptivus® in Combination With Low-dose Norvir®1

[back to top]

Discontinuations Due to an Adverse Event

The safety and tolerability of the observed antiretroviral therapy were based on the Adverse Events (AEs) and Serious Adverse Events (SAEs) reported in the case report forms. (NCT00531206)
Timeframe: 52 weeks

InterventionNumber of patients (Number)
Aptivus® in Combination With Low-dose Norvir®6

[back to top]

Gamma-glutamyl Transpeptidase (GGT) Over Time

(NCT00531206)
Timeframe: 52 weeks

Interventioninternational units/liter (Median)
Month 1 (n=54)Month 3 (n=49)Month 6 (n=57)Month 9 (n=44)Month 12 (n=43)
Aptivus® in Combination With Low-dose Norvir®72.072.072.076.068.0

[back to top]

Serious Adverse Events

The safety and tolerability of the observed antiretroviral therapy were based on the Adverse Events (AEs) and Serious Adverse Events (SAEs) reported in the case report forms. (NCT00531206)
Timeframe: 52 weeks

InterventionNumber of patients (Number)
Aptivus® in Combination With Low-dose Norvir®3

[back to top]

Alanine Aminotransferase (ALT) Over Time

(NCT00531206)
Timeframe: 52 weeks

Interventioninternational units/liter (Median)
Month 1 (n=52)Month 3 (n=49)Month 6 (n=55)Month 9 (n=41)Month 12 (n=42)
Aptivus® in Combination With Low-dose Norvir®33.031.037.034.037.5

[back to top]

Alkaline Phosphatase Over Time

(NCT00531206)
Timeframe: 52 weeks

Interventioninternational units/liter (Median)
Month 1 (n=33)Month 3 (n=38)Month 6 (n=26)Month 9 (n=24)Month 12 (n=43)
Aptivus® in Combination With Low-dose Norvir®90.092.587.586.592.5

[back to top]

Aspartate Aminotransferase (ALT) Over Time

(NCT00531206)
Timeframe: 52 weeks

Interventioninternational units/liter (Median)
Month 1 (n=54)Month 3 (n=50)Month 6 (n=57)Month 9 (n=44)Month 12 (n=42)
Aptivus® in Combination With Low-dose Norvir®34.533.540.044.037.5

[back to top]

Body Mass Index Class (Kilograms/Square Meter)

(NCT00531206)
Timeframe: 52 weeks

Interventionparticipants (Number)
Low (< 18)Normal (18 - 30)High (> 30)missing
Aptivus® in Combination With Low-dose Norvir®26201

[back to top]

Subjective Well-being

Investigator's opinion of patient's general condition (quality of life) (NCT00531206)
Timeframe: 52 weeks

Interventionparticipants (Number)
good general conditionmoderate general conditionbad general conditionmissing
Aptivus® in Combination With Low-dose Norvir®233570

[back to top]

CD4+ Cell Count

Change from baseline in CD4+ count over time (NCT00531206)
Timeframe: Baseline and 52 weeks

Interventioncells/mm3 (Median)
change from baseline to month 1 (4 weeks)change from baseline to month 3 (12 weeks)change from baseline to month 6 (24 weeks)change from baseline to month 9 (36 weeks)change from baseline to month 12 (52 weeks)
Aptivus® in Combination With Low-dose Norvir®30.5043.5039.5063.0036.00

[back to top]

Change in Viral Load

Log10 change from baseline in viral load over time (NCT00531206)
Timeframe: Baseline and 52 weeks

Interventionlog10 copies/ml (Median)
change from baseline to month 1 (4 weeks)change from baseline to month 3 (12 weeks)change from baseline to month 6 (24 weeks)change from baseline to month 9 (36 weeks)change from baseline to month 12 (52 weeks)
Aptivus® in Combination With Low-dose Norvir®-1.56-1.70-1.42-1.42-1.45

[back to top]

CD4+ Cell Count (Percent): Baseline and Median Changes From Baseline (ITT-Observed Case)

(NCT00534352)
Timeframe: Baseline, Day 8, 14, 22, 28 & 42 and Week 48

InterventionPercent Change from Baseline (Median)
Baseline (n=23)Day 8 (n=20)Day 14 (n=20)Day 22 (n=20)Day 28 (n=21)Day 42 (n=19)Week 48 (n=14)
TDF/FTC +/- TMC125 +/- DRV/Rtv26.20.14.21.83.04.23.8

[back to top]

Number of Participants Contributing to the Pharmacokinetic (PK) Evaluations: Cmin, Cmax, AUC24 & Css,av

At visit Days 14 & 28, samples were collected pre-dose and at 1, 2, 3, 4, 6, 9, and 12 hours post-dose. An additional sample was taken at 24 hours (Day 15 or 29 as applicable) post-dose. (NCT00534352)
Timeframe: 6 weeks

Interventionparticipants (Number)
Treatment A: TMC125 + TDF/FTC23
Treatment B: TMC125 + TDF/FTC + DRV/Rtv21

[back to top]

Virologic Response < 50 HIV-1 RNA Copies/mL (ITT-Observed Case)

Virologic Response < 50 HIV-1 RNA Copies/mL (ITT-Observed Case). (NCT00534352)
Timeframe: Day 8, 14, 22, 28, 42 and Week 48

Interventionparticipants (Number)
Day 8 (n=19)Day 14 (n=21)Day 22 (n=19)Day 28 (n=20)Day 42 (n=20)Week 48 (n=13)
TDF/FTC +/- TMC125 +/- DRV/Rtv0346710

[back to top]

Number of Participants With Treatment-Emergent Non-Graded Laboratory Abnormalities(Worst Abnormality): Glucose- Insulin

"Number of participants with Treatment-Emergent Non-Graded Laboratory Abnormalities(Worst Abnormality): Glucose- Insulin.~Normal Range: 3.0 - 27.0 ulU/mL" (NCT00534352)
Timeframe: Day 1 through 42 and Week 48

,,,
Interventionparticipant (Number)
Below 3.0 ulU/mLAbove 27.0 ulU/mL
Optional Extension13
Treatment A11
Treatment B23
Treatment C12

[back to top]

Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities(Worst Grade): Lipids- Triglycerides

"Number of participants with Treatment-Emergent Graded Laboratory Abnormalities(Worst Grade): Lipids- Triglycerides.~Worst Grade is based on the DAIDS toxicity grading scale, 0-5 : No Toxicity-Death." (NCT00534352)
Timeframe: Day 1 through 48 and Week 48

,,,
Interventionparticipants (Number)
Grade 1Grade 2
Optional Extension00
Treatment A00
Treatment B00
Treatment C00

[back to top]

Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities(Worst Grade): Lipids- Low-density Lipoprotein (LDL) Direct

"Number of participants with Treatment-Emergent Graded Laboratory Abnormalities(Worst Grade): Lipids- Low-density lipoprotein (LDL) Direct.~Worst Grade is based on the DAIDS toxicity grading scale, 0-5 : No Toxicity-Death." (NCT00534352)
Timeframe: Day 1 through 42 and Week 48

,,,
Interventionparticipants (Number)
Grade 1Grade 2
Optional Extension11
Treatment A00
Treatment B00
Treatment C11

[back to top]

Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities (Worst Grade): Lipids- Total Cholesteral

"Number of participants with Treatment-Emergent Graded Laboratory Abnormalities (Worst Grade): Lipids- Total Cholesteral.~Worst Grade is based on the DAIDS toxicity grading scale, 0-5 : No Toxicity-Death." (NCT00534352)
Timeframe: Day 1 through 42 and Week 48

,,,
Interventionparticipants (Number)
Grade 1Grade 2
Optional Extension10
Treatment A00
Treatment B00
Treatment C20

[back to top]

Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities (Worst Grade): Glucose-Hyperglycemia

"Number of Participants with Treatment-Emergent Graded Laboratory Abnormalities (Worst Grade): Glucose-Hyperglycemia.~Worst Grade is based on the National Institute of Allergy and Infectious Diseases Division of Acquired Immunodeficiency Syndrome (DAIDS) toxicity grading scale, 0,1,2,3,4 and 5 : None, Mild, Moderate, Severe, Life-threatening and Death." (NCT00534352)
Timeframe: Day 1 through 42 and Week 48

,,,
Interventionparticipants (Number)
Grade 1Grade 2
Optional Extension21
Treatment A11
Treatment B20
Treatment C01

[back to top]

Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities (Worst Grade): Glucose- Hypoglycemia

"Number of participants with Treatment-Emergent Graded Laboratory Abnormalities (Worst Grade): Glucose- Hypoglycemia.~Worst Grade is based on the DAIDS toxicity grading scale 0-5: No Toxicity-Death." (NCT00534352)
Timeframe: Day 1 through 42 and Week 48

,,,
Interventionparticipants (Number)
Grade 1Grade 2
Optional Extension10
Treatment A00
Treatment B21
Treatment C00

[back to top]

Log10 Viral Load (HIV-1 RNA Copies/mL): Mean Changes From Baseline(ITT-Observed Case)

Log10 Viral Load (HIV-1 RNA copies/mL): Mean Changes From Baseline(ITT-Observed Case). (NCT00534352)
Timeframe: Baseline, Day 8, 14, 22, 28 & 42 and Week 48

Interventioncopies/mL (Mean)
Baseline (n=23)Day 8 (n=19)Day 14 (n=21)Day 22 (n=19)Day 28 (n=20)Day 42 (n=20)Week 48 (n=13)
TDF/FTC +/- TMC125 +/- DRV/Rtv4.19-1.41-1.71-1.77-1.86-2.04-2.30

[back to top]

CD4+ Cell Count (x 10^6 Cell/L): Baseline and Median Changes From Baseline (ITT-Observed Case)

CD4+ Cell Count (x 10^6 cell/L): Baseline and Median Changes From Baseline (ITT-Observed Case). (NCT00534352)
Timeframe: Baseline, Day 8, 14, 22, 28 & 42 ans Week 48

Interventionx 10^6 cell/L (Median)
Baseline (n=23)Day 8 (n=20)Day 14 (n=20)Day 22 (n=20)Day 28 (n=21)Day 42 (n=19)Week 48 (n=14)
TDF/FTC +/- TMC125 +/- DRV/Rtv403.045.594.059.062.056.0160.0

[back to top]

Number of Participants With Treatment-Emergent Non-Graded Laboratory Abnormalities(Worst Abnormality): Lipids- High-density Lipoprotein (HDL)

"Number of participants with Treatment-Emergent Non-Graded Laboratory Abnormalities(Worst Abnormality): Lipids- High-density lipoprotein (HDL).~Normal Range:~40 - 59 mG/dL 1.03 - 1.53 mmol/L" (NCT00534352)
Timeframe: Day 1 through 42 and Week 48

,,,
Interventionparticipants (Number)
Below 40 mG/dL (1.03 mmol/L)Above 59 mG/dL (1.53 mmol/L)
Optional Extension21
Treatment A40
Treatment B80
Treatment C60

[back to top]

Change in Fasting Low Density Lipoprotein (LDL)Cholesterol Level

(NCT00552240)
Timeframe: baseline to week 48

Interventionmg/dl (Mean)
Nevirapine (NVP) Plus Truvada8.7
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada6.9

[back to top]

Change in Fasting Plasma Total Cholesterol Level

(NCT00552240)
Timeframe: baseline to week 48

Interventionmg/dl (Mean)
Nevirapine (NVP) Plus Truvada18.2
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada13.8

[back to top]

Change in Fasting Plasma Triglycerides Level

(NCT00552240)
Timeframe: baseline to week 48

Interventionmg/dl (Mean)
Nevirapine (NVP) Plus Truvada-4.7
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada8.4

[back to top]

Change in Fasting Total Cholesterol to High Density Lipoprotein (HDL) Ratio

(NCT00552240)
Timeframe: baseline to week 48

Interventionratio (Mean)
Nevirapine (NVP) Plus Truvada-0.38
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada-0.02

[back to top]

Change in Framingham Score

Framingham prediction of 10-year risk of Coronary Heart Disease (CHD) outcomes (myocardial infarction [MI] or CHD death) based on the patient's gender, age, systolic blood pressure, total cholesterol, HDL-c and smoking status. The scale for the estimated risk ranges from 0 to 30%. (NCT00552240)
Timeframe: baseline to week 48

Interventionpercent 10-year risk (Mean)
Nevirapine (NVP) Plus Truvada-0.09
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada0.14

[back to top]

Change in Glomerular Filtration Rate (GFR) From Baseline to Week 48

using 4-variable Modification of Diet in Renal Disease (MDRD) formula (NCT00552240)
Timeframe: baseline to week 48

Interventionml/min/1.73m^2 (Mean)
Nevirapine (NVP) Plus Truvada-0.06
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada-12.81

[back to top]

Percentage Adherence by Pill Count

Number of pills not returned / number of treatment days in percent (%) (NCT00552240)
Timeframe: baseline to week 48

Interventionpercentage adherence (Mean)
Nevirapine (NVP) Plus Truvada94.3
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada97.0

[back to top]

Proportion of Patients Reporting CNS Side Effects of Any Severity

(NCT00552240)
Timeframe: baseline to week 52

Interventionparticipants (Number)
Nevirapine (NVP) Plus Truvada25
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada23

[back to top]

Proportion of Patients Reporting Hepatic Events of Any Severity

(NCT00552240)
Timeframe: baseline to week 52

Interventionparticipants (Number)
Nevirapine (NVP) Plus Truvada5
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada24

[back to top]

Proportion of Patients Reporting Rash of Any Severity

(NCT00552240)
Timeframe: baseline to week 52

Interventionparticipants (Number)
Nevirapine (NVP) Plus Truvada21
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada19

[back to top]

Time to Virologic Response (First Confirmed Viral Load < 50 Copies/ml), All Participants

Time to response whereby patients withdrawing early were censored after their withdrawal (NCT00552240)
Timeframe: baseline to week 48

Interventiondays (Median)
Nevirapine (NVP) Plus Truvada57
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada84

[back to top]

Time to Virologic Response (First Confirmed Viral Load < 50 Copies/ml), Only Participants With Confirmed Viral Load < 50 Copies/ml

(NCT00552240)
Timeframe: baseline to week 48

Interventiondays (Median)
Nevirapine (NVP) Plus Truvada55
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada84

[back to top]

Incidence of Patients With AIDS Progression at Each Visit

Cumulative incidence of patients with AIDS progression are shown (NCT00552240)
Timeframe: baseline to week 52

,
Interventionparticipants (Number)
week 0week 2week 4week 6week 8week 12week 24week 36week 48week 50End of Study Visit
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada00011233333
Nevirapine (NVP) Plus Truvada00000000112

[back to top]

Number of Participants With HIV Viral Load < 400 Copies/ml at Week 12 of Treatment

Results within time windows, patients on-treatment (NCT00552240)
Timeframe: baseline to week 12

,
InterventionParticipants (Number)
HIV viral load < 400 copies/mlHIV viral load ≥ 400 copies/mlMissing data
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada6377
Nevirapine (NVP) Plus Truvada56613

[back to top]

Number of Participants With HIV Viral Load < 400 Copies/ml at Week 2 of Treatment

Results within time windows, patients on-treatment (NCT00552240)
Timeframe: baseline to week 2

,
InterventionParticipants (Number)
HIV viral load < 400 copies/mlHIV viral load ≥ 400 copies/mlMissing data
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada17519
Nevirapine (NVP) Plus Truvada24447

[back to top]

Number of Participants With HIV Viral Load < 400 Copies/ml at Week 24 of Treatment

Results within time windows, patients on-treatment (NCT00552240)
Timeframe: baseline to week 24

,
InterventionParticipants (Number)
HIV viral load < 400 copies/mlHIV viral load ≥ 400 copies/mlMissing data
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada63311
Nevirapine (NVP) Plus Truvada51618

[back to top]

Number of Participants With HIV Viral Load < 400 Copies/ml at Week 36 of Treatment

Results within time windows, patients on-treatment (NCT00552240)
Timeframe: baseline to week 36

,
InterventionParticipants (Number)
HIV viral load < 400 copies/mlHIV viral load ≥ 400 copies/mlMissing data
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada59117
Nevirapine (NVP) Plus Truvada54318

[back to top]

Number of Participants With HIV Viral Load < 400 Copies/ml at Week 4 of Treatment

Results within time windows, patients on-treatment (NCT00552240)
Timeframe: baseline to week 4

,
InterventionParticipants (Number)
HIV viral load < 400 copies/mlHIV viral load ≥ 400 copies/mlMissing data
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada31415
Nevirapine (NVP) Plus Truvada382710

[back to top]

Number of Participants With HIV Viral Load < 400 Copies/ml at Week 48 of Treatment

Results within time windows, patients on-treatment (NCT00552240)
Timeframe: baseline to week 48

,
InterventionParticipants (Number)
HIV viral load < 400 copies/mlHIV viral load ≥ 400 copies/mlMissing data
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada54221
Nevirapine (NVP) Plus Truvada43131

[back to top]

Number of Participants With HIV Viral Load < 400 Copies/ml at Week 6 of Treatment

Results within time windows, patients on-treatment (NCT00552240)
Timeframe: baseline to week 6

,
InterventionParticipants (Number)
HIV viral load < 400 copies/mlHIV viral load ≥ 400 copies/mlMissing data
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada442310
Nevirapine (NVP) Plus Truvada402114

[back to top]

Number of Participants With HIV Viral Load < 400 Copies/ml at Week 8 of Treatment

Results within time windows, patients on-treatment (NCT00552240)
Timeframe: baseline to week 8

,
InterventionParticipants (Number)
HIV viral load < 400 copies/mlHIV viral load ≥ 400 copies/mlMissing data
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada58154
Nevirapine (NVP) Plus Truvada481116

[back to top]

Number of Participants With HIV Viral Load < 50 Copies/ml at Week 12 of Treatment

Results within time windows, patients on-treatment (NCT00552240)
Timeframe: baseline to week 12

,
InterventionParticipants (Number)
HIV viral load < 50 copies/mlHIV viral load ≥ 50 copies/mlMissing data
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada43277
Nevirapine (NVP) Plus Truvada422013

[back to top]

Number of Participants With HIV Viral Load < 50 Copies/ml at Week 2 of Treatment

Results within time windows, patients on-treatment (NCT00552240)
Timeframe: baseline to week 2

,
InterventionParticipants (Number)
HIV viral load < 50 copies/mlHIV viral load ≥ 50 copies/mlMissing data
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada5639
Nevirapine (NVP) Plus Truvada6627

[back to top]

Number of Participants With HIV Viral Load < 50 Copies/ml at Week 24 of Treatment

Results within time windows, patients on-treatment (NCT00552240)
Timeframe: baseline to week 24

,
InterventionParticipants (Number)
HIV viral load < 50 copies/mlHIV viral load ≥ 50 copies/mlMissing data
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada61511
Nevirapine (NVP) Plus Truvada48918

[back to top]

Number of Participants With HIV Viral Load < 50 Copies/ml at Week 36 of Treatment

Results within time windows, patients on-treatment (NCT00552240)
Timeframe: baseline to week 36

,
InterventionParticipants (Number)
HIV viral load < 50 copies/mlHIV viral load ≥ 50 copies/mlMissing data
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada55517
Nevirapine (NVP) Plus Truvada53418

[back to top]

Number of Participants With HIV Viral Load < 50 Copies/ml at Week 4 of Treatment

Results within time windows, patients on-treatment (NCT00552240)
Timeframe: baseline to week 4

,
InterventionParticipants (Number)
HIV viral load < 50 copies/mlHIV viral load ≥ 50 copies/mlMissing data
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada10625
Nevirapine (NVP) Plus Truvada125310

[back to top]

Number of Participants With HIV Viral Load < 50 Copies/ml at Week 48

HIV viral load <50 copies/ml measured at Week 48 among observed cases on-treatment. (NCT00552240)
Timeframe: baseline to week 48

,
InterventionParticipants (Number)
RespondersNonresponders
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada488
Nevirapine (NVP) Plus Truvada422

[back to top]

Number of Participants With HIV Viral Load < 50 Copies/ml at Week 48 of Treatment

Results within time windows, patients on-treatment (NCT00552240)
Timeframe: baseline to week 48

,
InterventionParticipants (Number)
HIV viral load < 50 copies/mlHIV viral load ≥ 50 copies/mlMissing data
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada48821
Nevirapine (NVP) Plus Truvada42231

[back to top]

Number of Participants With HIV Viral Load < 50 Copies/ml at Week 8 of Treatment

Results within time windows, patients on-treatment (NCT00552240)
Timeframe: baseline to week 8

,
InterventionParticipants (Number)
HIV viral load < 50 copies/mlHIV viral load ≥ 50 copies/mlMissing data
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada23504
Nevirapine (NVP) Plus Truvada342516

[back to top]

Number of Participants With Loss of Virologic Response Following Confirmed Virologic Response

HIV viral load > 50 copies/ml on two consecutive measurements separated by at least 2 weeks, after confirmed virologic response (2 consecutive HIV viral load values < 50 copies/ml) (NCT00552240)
Timeframe: baseline to week 24 and week 48

,
InterventionParticipants (Number)
At week 24At week 48
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada49
Nevirapine (NVP) Plus Truvada12

[back to top]

Number of Participants With Virologic Response (VR)

VR is defined as HIV viral load of <50 copies/ml measured at two consecutive visits PRIOR TO Week 48 and without subsequent rebound or change of ARV therapy prior to Week 48. (NCT00552240)
Timeframe: baseline to week 48

,
Interventionparticipants (Number)
RespondersNonresponders
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada5027
Nevirapine (NVP) Plus Truvada4629

[back to top]

Number of Participants With Virologic Response According to the Time to Loss of Virologic Response (TLOVR) Algorithm

HIV viral load <50 copies/ml measured at two consecutive visits UP TO Week 48 and without subsequent rebound or change of ARV therapy up to Week 48. (NCT00552240)
Timeframe: baseline to week 48

,
InterventionParticipants (Number)
RespondersNonresponders
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada5126
Nevirapine (NVP) Plus Truvada4827

[back to top]

Number of Participants With Virologic Success (FDA Definition)

HIV viral load <50 copies/ml measured in the Week 48 window whereby patients withdrawing early and patients without a Week 48 assessment are considered failures. Includes all participants in full analysis set (FAS). (NCT00552240)
Timeframe: baseline to week 48

,
InterventionParticipants (Number)
RespondersNonresponders
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada4829
Nevirapine (NVP) Plus Truvada4233

[back to top]

Number of Patients With Virologic Rebound to >400 Copies/ml

HIV viral load >400 copies/ml on two consecutive measurements separated by at least 2 weeks, after confirmed virologic response (2 consecutive HIV viral load values < 50 copies/ml) (NCT00552240)
Timeframe: baseline to week 48

,
InterventionParticipants (Number)
Rebound following responseNo rebound following response
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada663
Nevirapine (NVP) Plus Truvada255

[back to top]

Proportion of Patients With DAIDS Grade >= 2 Laboratory Abnormalities

(NCT00552240)
Timeframe: baseline to week 52

,
Interventionparticipants (Number)
Grade 2 moderateGrade 3 severeGrade 4 potential lifethreatening
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada3173
Nevirapine (NVP) Plus Truvada2587

[back to top]

Number of Participants With HIV Viral Load < 50 Copies/ml at Week 6 of Treatment

Results within time windows, patients on-treatment (NCT00552240)
Timeframe: baseline to week 6

,
InterventionParticipants (Number)
HIV viral load < 50 copies/mlHIV viral load ≥ 50 copies/mlMissing data
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada145310
Nevirapine (NVP) Plus Truvada233814

[back to top]

AIDS Progression and Death: Number of Patients With a Treatment-emergent AIDS Defining Illness or an AIDS-defining Illness Leading to Death

"AIDS defining illnesses include: Aspergillosis, Bartonellosis, Candidiasis, Cervical cancer, Chagas disease, Coccidiodomycosis, Cryptococcosis, Cytomegalovirus retinus, encephalopathy, Herpes Simplex Virus, Histoplasmosis, Isosporiasis, Kaposi's sarcoma, Leishmaniasis, Microsporidiosis, Mycobacterium avium complex, mycobacterium (non-tuberculous), Nocardiosis, Pneumocystis carinii pneumonia, Pneumonia, Progressive Multifocal Leukoencephalopathy, Rhodococcus equi, Salmonella, Toxoplasmosis, Wasting.~Number of cases (no time-to analysis was performed due to small numbers)." (NCT00552240)
Timeframe: baseline to week 48

InterventionParticipants (Number)
Nevirapine (NVP) Plus Truvada1
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada3

[back to top]

Change in CD4+ Cell Count From Baseline to Week 12.

Patients on-treatment, data within time windows (NCT00552240)
Timeframe: baseline to week 12

Interventioncells/mm^3 (Mean)
Nevirapine (NVP) Plus Truvada123.1
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada102.2

[back to top]

Change in CD4+ Cell Count From Baseline to Week 2.

Patients on-treatment, data within time windows (NCT00552240)
Timeframe: baseline to week 2

Interventioncells/mm^3 (Mean)
Nevirapine (NVP) Plus Truvada62.6
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada61.0

[back to top]

Change in CD4+ Cell Count From Baseline to Week 24.

Patients on-treatment, data within time windows (NCT00552240)
Timeframe: baseline to week 24

Interventioncells/mm^3 (Mean)
Nevirapine (NVP) Plus Truvada131.8
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada132.5

[back to top]

Change in CD4+ Cell Count From Baseline to Week 36.

Patients on-treatment, data within time windows (NCT00552240)
Timeframe: baseline to week 36

Interventioncells/mm^3 (Mean)
Nevirapine (NVP) Plus Truvada147.6
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada120.5

[back to top]

Change in CD4+ Cell Count From Baseline to Week 4.

Patients on-treatment, data within time windows (NCT00552240)
Timeframe: baseline to week 4

Interventioncells/mm^3 (Mean)
Nevirapine (NVP) Plus Truvada76.4
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada63.0

[back to top]

Change in CD4+ Cell Count From Baseline to Week 48.

Patients on-treatment, data within time windows (NCT00552240)
Timeframe: baseline to week 48

Interventioncells/mm^3 (Mean)
Nevirapine (NVP) Plus Truvada155.1
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada160.4

[back to top]

Change in CD4+ Cell Count From Baseline to Week 6.

Patients on-treatment, data within time windows (NCT00552240)
Timeframe: baseline to week 6

Interventioncells/mm^3 (Mean)
Nevirapine (NVP) Plus Truvada87.2
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada78.4

[back to top]

Change in CD4+ Cell Count From Baseline to Week 8.

Patients on-treatment, data within time windows (NCT00552240)
Timeframe: baseline to week 8

Interventioncells/mm^3 (Mean)
Nevirapine (NVP) Plus Truvada111.9
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada90.5

[back to top]

Change in Fasting High Density Lipoprotein (HDL) Cholesterol Level

(NCT00552240)
Timeframe: baseline to week 48

Interventionmg/dl (Mean)
Nevirapine (NVP) Plus Truvada9.6
Atazanavir Plus Ritonavir (ATV/r) Plus Truvada3.5

[back to top]

Buprenorphine Area Under the Curve With LPV/r (ng/mL*hr)

Pharmacokinetic parameters were determined by use of non compartmental methods. The area under the plasma concentration versus time curve was determined by use of the trapezoidal rule and measured over a 24-hr time period. (NCT00571961)
Timeframe: 15 days

Intervention(ng/mL)*hr (Mean)
Lopinavir Coformulated With Ritonavir (LPV/r), 800mg/200mg46.2

[back to top]

Confirmed Virologic Failure at or Prior to Week 24

Confirmed virologic failure was defined as two successive HIV-1 RNA measurements at least 24 hours apart that were either:1) <1 log10 copies/mL below the baseline level and >400 copies/mL at the week 12 HIV-1 RNA evaluation (obtained at least 11 weeks after the date of the randomization) 2) >400 copies/mL at or after the week 24 HIV-1 RNA evaluation (obtained at least 23 weeks after the date of randomization). 3) subjects who discontinued the study follow-up for any reason other than study completion, including death, and who did so ≤30 weeks after randomization was considered to be a virologic failure. Number of participants experiencing or not experiencing virologic failure at or prior to week 24 was reported. (NCT00608569)
Timeframe: At or prior to Week 24

,
Interventionparticipants (Number)
No FailureExperienced Failure
mDOT Arm10524
Non-mDOT Arm11117

[back to top]

Time to First Grade 3 or 4 Lab Event

5th and 10th percentiles in weeks from randomization to first grade 3 or 4 lab event (NCT00608569)
Timeframe: 52 weeks since randomization

,
Interventionweeks (Number)
5th percentile10th percentile
mDOT Arm24NA
Non-mDOT ArmNANA

[back to top]

Adherence to Second Line HAART Regimen

Number of participants with self-reported 100% adherence over the week prior to study visit (NCT00608569)
Timeframe: At weeks 4, 8, 12, 24, 36, 48 and 52

,
Interventionparticipants (Number)
Week 4Week 8Week 12Week 24Week 48Week 52
mDOT Arm105108114107103104
Non-mDOT Arm117115116116109109

[back to top]

Confirmed Virologic Failure at or Prior to Week 48

Confirmed virologic failure was defined as two successive HIV-1 RNA measurements at least 24 hours apart that were either:1) <1 log10 copies/mL below the baseline level and >400 copies/mL at the week 12 HIV-1 RNA evaluation (obtained at least 11 weeks after the date of the randomization) 2) >400 copies/mL at or after the week 24 HIV-1 RNA evaluation (obtained at least 23 weeks after the date of randomization). 3) subjects who discontinued the study follow-up for any reason other than study completion, including death, and who did so ≤50 weeks after randomization was considered to be a virologic failure. Number of participants experiencing or not experiencing virologic failure at or prior to week 48 was reported. (NCT00608569)
Timeframe: At or prior to Week 48

,
Interventionparticipants (Number)
No FailureExperienced Failure
mDOT Arm9534
Non-mDOT Arm10523

[back to top]

CD4 Count at Follow-up Visits

CD4 cell count (median, inter-quartile range) (NCT00608569)
Timeframe: At Weeks 4, 12, 24, 36, and 48

,
Interventioncells/mm3 (Median)
Week 4Week 12Week 24Week 36Week 48
mDOT Arm212225268281301
Non-mDOT Arm219235266294347

[back to top]

CD8 Count at Follow-up Visits

CD8 cell count (median, inter-quartile range) (NCT00608569)
Timeframe: At week 4, 12, 24, 36, and 48

,
Interventioncells/mm3 (Median)
Week 4Week 12Week 24Week 36Week 48
mDOT Arm776895816787815
Non-mDOT Arm859916818833823

[back to top]

Time to First Grade 3 or 4 Sign or Symptom

5th and 10th percentiles in weeks from randomization to first grade 3 or 4 sign or symptom (NCT00608569)
Timeframe: 52 weeks since randomization

,
Interventionweeks (Number)
5th percentile10th percentile
mDOT Arm13.7NA
Non-mDOT Arm26.748.9

[back to top]

Time to First Grade 3 or 4 Lab or Sign/Symptom Event

5th and 10th percentiles in weeks from randomization to first grade 3 or 4 lab or sign/ symptom event (NCT00608569)
Timeframe: 52 weeks since randomization

,
Interventionweeks (Number)
5th percentile10th percentile
mDOT Arm6.424
Non-mDOT Arm2432.6

[back to top]

AUC: Steady-state Plasma Amprenavir (APV) Pharmacokinetics (PK) Following Administration of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent Raltegravir (RTG) 400mg BID.

APV minimum concentration (Cmin), maximum concentration (Cmax), area under the plasma concentration-time curve (AUC), and oral clearance (CL/F) as determined from APV concentrations observed in blood samples obtained at baseline, and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours during the BID regimens (FPV 1400mg BID, FPV 700mg/RTV 100 mg BID), and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 24 hours during the QD regimen (FPV 1400mg/RTV 100mg QD). As Groups A and B received the same regimens (albeit in different order), PK data for these two groups were collated, then assessed. For the same reason, PK data from Groups C and D regimens were collated before assessment, as were the PK data from Groups E and F (NCT00614991)
Timeframe: Day 7 of the RAL 400mg BID regimen and Day 14 of the RAL 400mg/FPV 1400mg BID, RAL 400mg/FPV 700mg/RTV 100mg BID, and RAL 400mg BID Plus FPV 1400mg/RTV 100mg QD regimens

Interventionng•h/mL (Mean)
Group A & B0.71
Group C & D0.46
Group E & F0.70

[back to top]

CL/F: Steady-state Plasma Amprenavir (APV) Pharmacokinetics (PK) Following Administration of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent Raltegravir (RTG) 400mg BID.

APV minimum concentration (Cmin), maximum concentration (Cmax), area under the plasma concentration-time curve (AUC), and oral clearance (CL/F) as determined from APV concentrations observed in blood samples obtained at baseline, and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours during the BID regimens (FPV 1400mg BID, FPV 700mg/RTV 100 mg BID), and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 24 hours during the QD regimen (FPV 1400mg/RTV 100mg QD). As Groups A and B received the same regimens (albeit in different order), PK data for these two groups were collated, then assessed. For the same reason, PK data from Groups C and D regimens were collated before assessment, as were the PK data from Groups E and F (NCT00614991)
Timeframe: Day 14 of the FPV 1400mg BID, FPV 1400mg/RAL 400mg BID, FPV 700mg/RTV 100mg BID, FPV 700mg/RTV 100mg/RAL 400mg BID, FPV 1400mg/RTV 100mg QD, and FPV 1400mg/RTV 100mg QD plus RAL 400mg BID regimens

InterventionL/H (Mean)
Group A & B1.26
Group C & D1.43
Group E & F1.05

[back to top]

CL/F: Steady-state Plasma Amprenavir (APV) Pharmacokinetics (PK) Following Administration of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent Raltegravir (RTG) 400mg BID.

APV minimum concentration (Cmin), maximum concentration (Cmax), area under the plasma concentration-time curve (AUC), and oral clearance (CL/F) as determined from APV concentrations observed in blood samples obtained at baseline, and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours during the BID regimens (FPV 1400mg BID, FPV 700mg/RTV 100 mg BID), and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 24 hours during the QD regimen (FPV 1400mg/RTV 100mg QD). As Groups A and B received the same regimens (albeit in different order), PK data for these two groups were collated, then assessed. For the same reason, PK data from Groups C and D regimens were collated before assessment, as were the PK data from Groups E and F (NCT00614991)
Timeframe: Day 7 of the RAL 400mg BID regimen and Day 14 of the RAL 400mg/FPV 1400mg BID, RAL 400mg/FPV 700mg/RTV 100mg BID, and RAL 400mg BID Plus FPV 1400mg/RTV 100mg QD regimens

InterventionL/h (Mean)
Group A & B1.02
Group C & D2.30
Group E & F1.46

[back to top]

Cmin/Cmax: Steady-state Plasma Amprenavir (APV) Pharmacokinetics (PK) Following Admin of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent Raltegravir (RTG) 400mg BID.

APV minimum concentration (Cmin), maximum concentration (Cmax), area under the plasma concentration-time curve (AUC), and oral clearance (CL/F) as determined from APV concentrations observed in blood samples obtained at baseline, and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours during the BID regimens (FPV 1400mg BID, FPV 700mg/RTV 100 mg BID), and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 24 hours during the QD regimen (FPV 1400mg/RTV 100mg QD). As Groups A and B received the same regimens (albeit in different order), PK data for these two groups were collated, then assessed. For the same reason, PK data from Groups C and D regimens were collated before assessment, as were the PK data from Groups E and F (NCT00614991)
Timeframe: Day 14 of the FPV 1400mg BID, FPV 1400mg/RAL 400mg BID, FPV 700mg/RTV 100mg BID, FPV 700mg/RTV 100mg/RAL 400mg BID, FPV 1400mg/RTV 100mg QD, and FPV 1400mg/RTV 100mg QD plus RAL 400mg BID regimens

,,
Interventionng/mL (Mean)
Cmin (ng/mL)Cmax (ng/mL)
Group A & B0.670.83
Group C & D0.670.75
Group E & F0.831.27

[back to top]

Number of Participants Who Experienced Adverse Events

"Safety/tolerability data included all adverse events (AEs) reported within the time frame of each regimen evaluated. The intent was to compare adverse events for each sequence and not for each regimen. The regimens for which AE information was culled were:~RAL 400mg BID alone~FPV 1400mg BID alone~FPV 700mg/RTV 100 mg BID alone~FPV 1400mg/RTV 100mg QD alone~FPV 1400mg BID combined with RAL 400mg BID~FPV 700mg/RTV 100 mg BID combined with RAL 400mg BID~FPV 1400mg/RTV 100mg QD combined with RAL 400mg BID The severity of reported AEs was graded according to DAIDS criteria, Version 1.0 (National Institute of Allergy and Infectious Diseases (NIAID). Table for Grading the Severity of Adult and Pediatric Adverse Events, Version 1.0. Division of Acquired Immunodeficiency Syndrome (DAIDS), Washington D.C.; 2004." (NCT00614991)
Timeframe: Day 0 through Day 49

Interventionparticipants (Number)
Group A0
Group B4
Group C6
Group D5
Group E5
Group F4

[back to top]

Cmin/Cmax: Steady-state Plasma RTG PK Following Admin of FPV 1400mg BID, FPV 700mg/RTV 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent RTG 400mg BID.

RAL minimum concentration (Cmin), maximum concentration (Cmax), area under the plasma concentration-time curve (AUC), and oral clearance (CL/F) as determined from RAL concentrations observed in blood samples obtained at baseline, and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours during the period when RAL 400mg BID was administered with the FPV-Containing BID regimens (FPV 1400mg BID, FPV 700mg/RTV 100 mg BID), and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 24 hours during the period when RAL 400mg BID was administered with the FPV QD regimen (FPV 1400mg/RTV 100mg QD). As Groups A and B received the same regimens (albeit in different order), PK data for these two groups were collated, then assessed. For the same reason, PK data from Groups C and D regimens were collated before assessment, as were the PK data from Groups E and F. (NCT00614991)
Timeframe: Day 7 of the RAL 400mg BID regimen and Day 14 of the RAL 400mg/FPV 1400mg BID, RAL 400mg/FPV 700mg/RTV 100mg BID, and RAL 400mg BID Plus FPV 1400mg/RTV 100mg QD regimens

,,
Interventionng/mL (Mean)
Cmin (ng/mL)Cmax (ng/mL)
Group A & B0.320.95
Group C & D0.460.44
Group E & F0.590.85

[back to top]

AUC: Steady-state Plasma Amprenavir (APV) Pharmacokinetics (PK) Following Admin of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent Raltegravir (RTG) 400mg BID.

APV minimum concentration (Cmin), maximum concentration (Cmax), area under the plasma concentration-time curve (AUC), and oral clearance (CL/F) as determined from APV concentrations observed in blood samples obtained at baseline, and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours during the BID regimens (FPV 1400mg BID, FPV 700mg/RTV 100 mg BID), and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 24 hours during the QD regimen (FPV 1400mg/RTV 100mg QD). As Groups A and B received the same regimens (albeit in different order), PK data for these two groups were collated, then assessed. For the same reason, PK data from Groups C and D regimens were collated before assessment, as were the PK data from Groups E and F (NCT00614991)
Timeframe: Day 14 of the FPV 1400mg BID, FPV 1400mg/RAL 400mg BID, FPV 700mg/RTV 100mg BID, FPV 700mg/RTV 100mg/RAL 400mg BID, FPV 1400mg/RTV 100mg QD, and FPV 1400mg/RTV 100mg QD plus RAL 400mg BID regimens

Interventionng•h/mL (Mean)
Group A & B0.81
Group C & D0.75
Group E & F1.13

[back to top]

Change In Hematuria, Glycosuria And Proteinuria From Baseline

Change from Baseline was calculated as the post-Baseline value minus the Baseline value. (NCT00623597)
Timeframe: Baseline (Day 1), Week 24 and Week 48

,,
Intervention[0 to 4+] (Mean)
Hematuria-Change from BL at Wk 24 (n= 1, 2, 3)Hematuria-Change from BL at Wk 48 (n= 1, 2, 3)Glycosuria-Change from BL at Wk 24 (n= 1, 2, 3)Glycosuria-Change from BL at Wk 48 (n= 1, 2, 3)Proteinuria-Change from BL at Wk 24 (n= 1, 2, 3)Proteinuria-Change from BL at Wk 48 (n= 1, 2, 3)
Group A000000
Group B000000
Total000000

[back to top]

Change In Hemoglobin, Total Protein And Total Albumin From Baseline

Change from Baseline was calculated as the post-Baseline value minus the Baseline value. (NCT00623597)
Timeframe: Baseline (Day 1), Week 24 and Week 48

,,
Interventiong/L (Mean)
Hemoglobin, Change from BL at Wk 24 (n= 4, 13, 17)Hemoglobin, Change from BL at Wk 48 (n= 4, 13, 17)Total Protein, Change from BL at Wk 24 (n=4,13,17)Total Protein, Change from BL at Wk 48 (n=4,13,17)Total Albumin, Change from BL at Wk 24 (n=4,13,17)Total Albumin, Change from BL at Wk 48 (n=4,13,17)
Group A-06-4-50.51.3
Group B102-054
Total120-143.4

[back to top]

Change In Red Blood Cell (RBC) Counts From Baseline

Change from Baseline was calculated as the post-Baseline value minus the Baseline value. (NCT00623597)
Timeframe: Baseline (Day 1), Week 24 and Week 48

,,
Intervention10*12/L (Mean)
RBC,Change from Baseline at Week 24 (n= 4, 13, 17)RBC,Change from Baseline at Week 48 (n= 4, 13, 17)
Group A-0.180.02
Group B-0.07-0.16
Total-0.1-0.12

[back to top]

Plasma Trough Concentrations (Ctrough) for Ritonavir

Plasma trough concentration is the average steady state concentration prior to morning and evening dose. Ctrough of Ritonavir was normalized to a dose of 100 mg/kg. (NCT00623597)
Timeframe: Pre-dose at Weeks 8, 12, 24

Interventionng/mL (Mean)
Group A577
Group B995

[back to top]

Area Under the Plasma Concentration-time Curve Over the Time Interval From Zero to Twelve Hours (AUC0-12h) for Ritonavir

The area under the plasma concentration-time curve from time zero to twelve hours (AUC0-12h) is area under the plasma concentration-time curve from time zero through actual tlast. The area under the plasma concentration-time curve from time zero to twelve hours of ritonasvir was normalized to a dose of 100 mg/kg. (NCT00623597)
Timeframe: Pre-dose and 3, 4, 8, 12 hours (post-dose) on Day 14 (± 2 days), or Day 28(+ 2 days) for patients switching from an NNRTI containing regimen).

Interventionh*ug/mL (Mean)
Group A13.6
Group B21.8

[back to top]

Change From Baseline in Cluster Differentiation Antigen 8 (CD8) Lymphocyte Count

Change from baseline in CD8+ lymphocyte count at 24 weeks and 48 weeks were presented by age group. Change from baseline in CD8+ lymphocyte count was derived as follows: Change from baseline = (CD8+ count at week 24/48) - (CD8+ count at baseline). A baseline collection was made if there was not already a value available taken within the previous 4 weeks. Baseline was on Day 1. (NCT00623597)
Timeframe: Baseline (Day 1), Weeks 8, 12, 24, 36, and 48 or upon premature discontinuation

,,
Interventioncount/uL (Mean)
Change from Baseline at Week 24 (n= 3, 12, 15)Change from Baseline at Week 48 (n= 3, 12, 15)
Group A-200.49-92.07
Group B-3.5040.52
Total-42.9014.00

[back to top]

Number of Participants With >1 Log Decrease From Baseline in Human Immunodeficiency Virus (HIV) -Ribonucleic Acid (RNA )

The number of participants experiencing a greater than 1 log drop from baseline (day 1) (log 10 transformed) were reported (NCT00623597)
Timeframe: From Week 8 till Week 48

,,
Interventionparticipants (Number)
Week 8 (n= 4, 12, 16)Week 24 (n= 3, 13, 16)Week 48 (n= 3, 13, 16)
Group A111
Group B787
Total898

[back to top]

Maximum Observed Concentration (Cmax) for Saquinavir and Ritonavir

The Plasma Concentration (Cmax) is defined as maximum observed analyte concentration. Cmax was normalized to a dose of 50 mg/kg for Saquinavir and100 mg/kg for Ritonavir. (NCT00623597)
Timeframe: Pre-dose and 3, 4, 8, 12 hours (post-dose) on Day 14 (± 2 days), or Day 28(+ 2 days) for patients switching from an NNRTI containing regimen and at Week 24

,
Interventionng/mL (Mean)
SaquinavirRitonavir
Group A29102050
Group B55703370

[back to top]

Incidence of Adverse Events (AE) and Serious Adverse Events (SAE)

An AE is defined as any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product. An SAE is defined as any untoward medical occurrence that, at any dose, results in death, is life threatening, requires hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect, or is a significant medical event in the investigator's judgment or requires intervention to prevent one or other of these outcomes (NCT00623597)
Timeframe: From Baseline (Day 1) till Week 48 and Follow-up (Week 52)

,,
Interventionparticipants (Number)
Any serious adverse eventsAny non-serious adverse events
Group A15
Group B29
Total314

[back to top]

Change In Total Bilirubin, Creatinine, Uric Acid From Baseline

Change from Baseline was calculated as the post-Baseline value minus the Baseline value. (NCT00623597)
Timeframe: Baseline (Day 1), Week 24 and Week 48

,,
Interventionumol/L (Mean)
Total Bilirubin-Change from BL at Wk 24(n=4,12,16)Total Bilirubin-Change from BL at Wk 48(n=4,12,16)Cretainine- Change from BL at Wk 24 (n=4, 13,17)Cretainine- Change from BL at Wk 48 (n=4, 13,17)Uric acid- Change from BL at Wk 24 (n=4, 13,17)Uric acid- Change from BL at Wk 48 (n=3,13,16)
Group A21-23-27-42
Group B22356861
Total21254641

[back to top]

Change In White Blood Cell (WBC), Platelet, Basophil, Lymphocyte, Monocyte, Neutrophil And Eosinophil Cell Counts From Baseline

Change from Baseline was calculated as the post-Baseline value minus the Baseline value. (NCT00623597)
Timeframe: Baseline (Day 1), Week 24 and Week 48

,,
Intervention10*9/L (Mean)
WBC, Change from BL at Wk 24 (n= 4,13,17)WBC, Change from BL at Wk 48 (n= 4,13,17)Platelet, Change from BL at Wk 24 (n= 4,13,17)Platelet, Change from BL at Wk 48 (n= 4,13,17)Basophil, Change from BL at Wk 24 (n=4 ,13,17)Basophil, Change from BL at Wk 48 (n=4 ,13,17)Lymphocyte, Change from BL at Wk 24 (n=2, 2, 4)Lymphocyte, Change from BL at Wk 48 (n= 2, 2, 4)Monocyte, Change from BL at Wk 24 (n= ,13,17)Monocyte, Change from BL at Wk 48 (n= ,13,17)Neutrophil, Change from BL at Wk 24 (n=2, 2, 4)Neutrophil, Change from BL at Wk 48 (n= 2, 2, 4)Eosinophil, Change from BL at Wk 24 (n=2, 2, 4)Eosinophil, Change from BL at Wk 48 (n= 2, 2, 4)
Group A-3-2.1-119-50-0.02-0-0.9-1.5-0.41-0.21-1.4-1.7-0-0
Group B-1-0.924-20-0.01-1.1-0.7-0.01-0.05-2.8-1.6-00
Total-1.5-1.2-9-13-0.01-0.01-1-1.1-0.11-0.09-2.1-1.6-00

[back to top]

Number of Participants With Human Immunodeficiency Virus (HIV) -Ribonucleic Acid (RNA) <400 Copies/mL

The number of participants with HIV-1 RNA results <400 copies/mL were reported (NCT00623597)
Timeframe: Baseline (Day 1), Weeks 8, 12, 24, 36, and 48 (or upon premature discontinuation); a baseline collection was made if there was not already a value available taken within the previous 4 weeks.

,,
Interventionparticipants (Number)
Baseline (n= 5, 13, 18)Week 24 (n= 3, 13, 16)Week 48 (n= 3, 13, 16)
Group A122
Group B51311
Total61513

[back to top]

Number of Participants With Human Immunodeficiency Virus (HIV) -Ribonucleic Acid (RNA) <50 Copies/mL

The number of participants with HIV-1 RNA results <50 copies/mL were reported. (NCT00623597)
Timeframe: Baseline (Day 1), Weeks 8, 12, 24, 36, and 48 (or upon premature discontinuation); a baseline collection was made if there was not already a value available taken within the previous 4 weeks.

,,
Interventionparticipants (Number)
Baseline (n= 5, 13, 18)Week 24 (n= 3, 13, 16)Week 48 (n= 3, 13, 16)
Group A022
Group B4119
Total41311

[back to top]

Number of Participants With Virological Failure

Virological failure was defined as: viral load >= 400 copies/mL on two consecutive occasions (missing visits was assumed to be above 400 copies/mL). The number of participants classified as virological failure by Age Group and viral load (≤ 10,000 copies, >10,000 copies) were presented. (NCT00623597)
Timeframe: From Week 12 till Week 48

,,
Interventionparticipants (Number)
VF at Week 12 (n= 5, 13, 18)VF at Week 24 (n= 5, 13, 18)VF at Week 48 (n= 5, 13, 18)HIV-RNA <= 10,000 copies/mL at Wk 12 (n= 3, 6, 9)HIV-RNA <= 10,000 copies/mL at Wk 24 (n= 3, 6, 9)HIV-RNA <= 10,000 copies/mL at Wk 48 (n= 3, 6, 9)HIV-RNA >10,000 copies/mL at Week 12 (n= 2, 7, 9)HIV-RNA >10,000 copies/mL at Week 24 (n= 2, 7, 9)HIV-RNA >10,000 copies/mL at Week 48 (n= 2, 7, 9)
Group A223112111
Group B101000101
Total324112212

[back to top]

Change In Hematocrit From Baseline

Change from Baseline was calculated as the post-Baseline value minus the Baseline value. (NCT00623597)
Timeframe: Baseline (Day 1), Week 24 and Week 48

,,
Interventionfraction (Mean)
Change from Baseline at Week 24 (n= 4, 13, 17)Change from Baseline at Week 48 (n= 4, 13, 17)
Group A0.010.03
Group B-0-0.01
Total-00

[back to top]

Change In Creatine Kinase (CK), Serum Glutamic Oxaloacetic Transaminase (SGOT), Alkaline Phosphatase (ALP), Serum Glutamic-Pyruvic Transaminase (SGPT), Gamma-Glutamyl Transferase (GGT) Counts From Baseline

Change from Baseline was calculated as the post-Baseline value minus the Baseline value. (NCT00623597)
Timeframe: Baseline (Day 1), Week 24 and Week 48

,,
InterventionU/L (Mean)
CK, Change from Baseline at Week 24 (n= 3, 3, 16)CK, Change from Baseline at Week 48 (n= 3, 3, 16)SGOT, Change from Baseline at Week 24 (n= 4,13,17)SGOT, Change from Baseline at Week 48 (n= 4,13,17)ALP, Change from Baseline at Week 24 (n=4, 12, 16)ALP, Change from Baseline at Week 48 (n=4, 12, 16)SGPT, Change from Baseline at Week 24 (n=4,13,17)SGPT, Change from Baseline at Week 48 (n=4,13,17)GGT, Change from Baseline at Week 24 (n=4, 13, 17)GGT, Change from Baseline at Week 48 (n=4, 13, 17)
Group A112257-0-11047-9-255
Group B-12-7-0-02636-2-3-7-5
Total1143-0-02239-4-3-4-2

[back to top]

Change In Blood Urea Nitrogen (BUN), Low Density Lipoprotein (LDL) Cholesterol, High Density Lipoprotein (HDL Cholesterol), Triglycerides, Calcium, Potassium, Sodium, Chloride, Phosphate, Fasting Glucose From Baseline

Change from Baseline was calculated as the post-Baseline value minus the Baseline value. (NCT00623597)
Timeframe: Baseline (Day 1), Week 24 and Week 48

,,
Interventionmmol/L (Mean)
BUN,Change from Baseline at Week 24 (n= 3, 13, 16)BUN,Change from Baseline at Week 48 (n= 3, 13, 16)LDL,Change from Baseline at Week 24 (n= 3, 13, 16)LDL,Change from Baseline at Week 48 (n= 3, 13, 16)HDL,Change from Baseline at Week 24 (n= 3, 13, 16)HDL,Change from Baseline at Week 48 (n= 3, 13, 16)Triglyceride,Change from BL at Wk 24 (n=4,13, 17)Triglyceride,Change from BL at Wk 48 (n=4,13, 17)Calcium,Change from BL at Wk 24 (n=2, 12, 14)Calcium,Change from BL at Wk 48 (n=2, 12, 14)Potassium,Change from BL at Wk 24 (n=4,13, 17)Potassium,Change from BL at Wk 48 (n=4,13, 17)Sodium,Change from BL at Wk 24 (n=4,13, 17)Sodium,Change from BL at Wk 48 (n=4,13, 17)Chloride,Change from BL at Wk 24 (n=4,13, 17)Chloride,Change from BL at Wk 48 (n=4,13, 17)Phosphate,Change from BL at Wk 24 (n=2, 12, 14)Phosphate,Change from BL at Wk 48 (n=2, 13, 15)Fasting Glucose,Change from BL at Wk 24(n=4,13,17)Fasting Glucose,Change from BL at Wk 48(n=4,13,17)
Group A-1.50.40.210.230.210.41-0.28-0.19-0.15-0.16-0.10.221-11-0.02-0.26-0.26-0.07
Group B11.10.21-0.110.130.17-0.19-0.150.150.09-0.3-0.2-1-1-2-20.170.120.020.14
Total0.50.90.21-0.040.150.21-0.21-0.160.110.06-0.2-0.1-0-0-2-10.140.07-0.050.09

[back to top]

Change From Baseline in Mean Human Immunodeficiency Virus Viral Load

Change from baseline in plasma HIV-1 RNA was derived as Change from baseline = Log10 (HIV-1 RNA at week x) - Log10 (HIV-1 RNA at baseline) (NCT00623597)
Timeframe: Baseline (Day 1), Weeks 8, 12, 24, 36, and 48 (or upon premature discontinuation); a baseline collection was made if there was not already a value available taken within the previous 4 weeks.

,,
Interventionlog10 copies/mL (Mean)
Change from Baseline at Week 24 (n= 3, 13, 16)Change from Baseline at Week 48 (n= 3, 13, 16)
Group A-0.75-1.27
Group B-1.81-1.39
Total-1.61-1.36

[back to top]

Area Under the Plasma Concentration-time Curve Over the Time Interval From Zero to Twelve Hours (AUC0-12h) for Saquinavir

The area under the plasma concentration-time curve from time zero to twelve hours (AUC0-12h) is area under the plasma concentration-time curve from time zero through actual tlast. The area under the plasma concentration-time curve from time zero to twelve hours of saquinavir was normalized to a dose of 50 mg/kg. (NCT00623597)
Timeframe: Pre-dose and 3, 4, 8, 12 hours (post-dose) on Day 14 (± 2 days), or Day 28(+ 2 days) for patients switching from an Non-nucleoside reverse transcriptase inhibitor [NNRTI] containing regimen).

Interventionh*ug/mL (Mean)
Group A18.7
Group B38

[back to top]

Change From Baseline in Cluster Differentiation Antigen 4 (CD4) Lymphocyte Count

Change from Baseline in CD4+ lymphocyte count at 24 weeks and 48 weeks were presented by age group. Change from baseline in CD4+ lymphocyte count was derived as follows: Change from baseline = (CD4+ count at week 24/48) - (CD4+ count at baseline). A baseline collection was made if there was not already a value available taken within the previous 4 weeks. Baseline was on Day 1. (NCT00623597)
Timeframe: Baseline (Day 1), Weeks 8, 12, 24, 36, and 48 or upon premature discontinuation

,,
Interventioncount/uL (Mean)
Change from Baseline at Week 24 (n= 3, 12, 15)Change from Baseline at Week 48 (n= 3, 12, 15)
Group A94.90-50.07
Group B-34.53126.11
Total-8.6590.87

[back to top]

Plasma Trough Concentrations (Ctrough) for Saquinavir

Plasma trough concentration is the average steady state concentration prior to morning and evening dose. Ctrough of Saquinavir was normalized to a dose of 50 mg/kg. (NCT00623597)
Timeframe: Pre-dose at Weeks 8, 12, 24.

Interventionng/mL (Mean)
Group A645
Group B1860

[back to top]

Darunavir Area Under the Plasma Concentration-time Curve (AUC) Over the Dosing Interval

AUC of darunavir over a 12-hour dosing interval. (NCT00630734)
Timeframe: 0, 1, 2, 3, 4, 5, 6, 8, 12 hours post-dose

Interventionng*h/ml (Mean)
SLCO1B1 Group 158552
SLCO1B1 Group 263679
SLCO1B1 Group 360156

[back to top]

Darunavir Maximum Plasma Concentration (Cmax)

Cmax of darunavir over a 12-hour dosing interval (NCT00630734)
Timeframe: 0, 1, 2, 3, 4, 5, 6, 8, 12 hours post-dose

Interventionng/ml (Mean)
SLCO1B1 Group 17770
SLCO1B1 Group 28047
SLCO1B1 Group 37789

[back to top]

Pravastatin + Darunavir/Ritonavir: Pravastatin Area Under the Plasma Concentration-time Curve (AUC) Over the Dosing Interval

Dosing interval of 24 hours (NCT00630734)
Timeframe: 0, 0.5, 1, 2, 3, 4, 6, 8, 12, 16, 20, 24 hours post-dose

Interventionng*h/ml (Mean)
SLCO1B1 Group 168.4
SLCO1B1 Group 2106.8
SLCO1B1 Group 3145.7

[back to top]

Ritonavir Maximum Plasma Concentration (Cmax)

Cmax of ritonavir over a 12-hour dosing interval (NCT00630734)
Timeframe: 0,1, 2, 3, 4, 5, 6, 8, 12 hours post-dose

Interventionng/ml (Mean)
SLCO1B1 Group 1844
SLCO1B1 Group 21143
SLCO1B1 Group 31279

[back to top]

Relative Change in Pravastatin Maximum Plasma Concentration (Cmax)

Cmax of pravastatin when administered with darunavir/ritonavir divided by the Cmax of pravastatin when administered alone. (NCT00630734)
Timeframe: 0, 0.5, 1, 2, 3, 4, 6, 8, 12, 16, 20, 24 hours post-dose

Interventionng/ml (Mean)
SLCO1B1 Group 11.11
SLCO1B1 Group 21.69
SLCO1B1 Group 32.32

[back to top]

Pravastatin Alone: Pravastatin Area Under the Plasma Concentration-time Curve (AUC) Over the Dosing Interval

Dosing interval of 24 hours (NCT00630734)
Timeframe: 0, 0.5, 1, 2, 3, 4, 6, 8, 12, 16, 20, 24 hours post-dose

Interventionng*h/ml (Mean)
SLCO1B1 Group 163
SLCO1B1 Group 286.6
SLCO1B1 Group 3123.4

[back to top]

Ritonavir Area Under the Plasma Concentration-time Curve (AUC) Over the Dosing Interval

AUC of ritonavir over a 12-hour dosing interval. (NCT00630734)
Timeframe: 0, 1, 2, 3, 4, 5, 6, 8, 12 hours post-dose

Interventionng*hr/ml (Mean)
SLCO1B1 Group 15239
SLCO1B1 Group 27178
SLCO1B1 Group 37907

[back to top]

Relative Change in Pravastatin Area Under the Plasma Concentration-time Curve (AUC) Over the Dosing Interval

AUC of pravastatin when administered with darunavir/ritonavir divided by AUC of pravastatin when administered alone. The AUC was measured over a 24-hour dosing interval. (NCT00630734)
Timeframe: 0, 0.5, 1, 2, 3, 4, 6, 8, 12, 16, 20, 24 hours post-dose

Interventionng*hr/ml (Mean)
SLCO1B1 Group 11.09
SLCO1B1 Group 21.59
SLCO1B1 Group 31.75

[back to top]

Pravastatin + Darunavir/Ritonavir: Pravastatin Maximum Plasma Concentration (Cmax)

(NCT00630734)
Timeframe: 0, 0.5, 1, 2, 3, 4, 6, 8, 12, 16, 20, 24 hours post-dose

Interventionng/ml (Mean)
SLCO1B1 Group 130.1
SLCO1B1 Group 242.4
SLCO1B1 Group 352.8

[back to top]

Pravastatin Alone: Pravastatin Maximum Plasma Concentration (Cmax)

(NCT00630734)
Timeframe: 0, 0.5, 1, 2, 3, 4, 6, 8, 12, 16, 20, 24 hours post-dose

Interventionng/ml (Mean)
SLCO1B1 Group 127.7
SLCO1B1 Group 233.3
SLCO1B1 Group 346.2

[back to top]

Absolute Change in CD4 Cell Counts

(NCT00632970)
Timeframe: 24 and 48 weeks

Interventioncells/mm^3 (Mean)
Raltegravir50
Lopinavir/Ritonavir50

[back to top]

Cmin of RTV

Cmin was derived from the plasma concentration versus time for RTV. (NCT00646776)
Timeframe: Pre-dose (0h) on Days 4, 8, 11 to 18 and post-dose (1h,2h,3h,4h,6h,8h,12h) on Days 11 and 15 for ATV/RTV + RIB.

Interventionng/mL (Geometric Mean)
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly40.54

[back to top]

Maximum Plasma Concentration (Cmax) of RIB

Cmax was derived from plasma concentration versus time for RIB and was recorded directly from experimental observations for each treatment period. (NCT00646776)
Timeframe: Pre-dose (0h) on Days 6, 8, 10, and 11, and post-dose (1h,2h,3h,4h,6h,8h,12h) on Day 10 for RIB 150 mg QD; and pre-dose (0h) on Days 4, 8,11 to 18 and post-dose (1h,2h,3h,4h,6h,8h,12h) on Days 11 and 15 for ATV/RTV + RIB.

Interventionng/mL (Geometric Mean)
RIB 150 mg Once Daily (QD)159.0
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly395.6

[back to top]

Minimum Plasma Concentration (Cmin) of RIB

Cmin was derived from plasma concentration versus time for RIB. (NCT00646776)
Timeframe: Pre-dose (0h) on Days 6, 8, 10, and 11, and post-dose (1h,2h,3h,4h,6h,8h,12h) on Day 10 for RIB 150 mg QD; and pre-dose (0h) on Days 4, 8,11 to 18 and post-dose (1h,2h,3h,4h,6h,8h,12h) on Days 11 and 15 for ATV/RTV + RIB.

Interventionng/mL (Geometric Mean)
RIB 150 mg Once Daily (QD)28.89
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly40.49

[back to top]

Number of Participants With Clinically Significant Vital Signs or Physical Examination Findings

Vital signs assessments and physical examination were conducted throughout the study. Vital signs assessments included body temperature, respiratory rate, blood pressure (systolic and diastolic), and heart rate. Physical examination included a neurological examination (if ocular signs or symptoms occurred, a reflex to slit lamp exam was performed by an ophthalmologist). The investigator used his/her clinical judgment to decide whether or not abnormalities in vital signs or physical examination were clinically meaningful. (NCT00646776)
Timeframe: Vital signs:screening, prior to dosing on Day 1, Day 7, study discharge. Physical examination:screening, Day -1, Day 7, study discharge

InterventionParticipants (Number)
RIB 150 mg Once Daily (QD)0
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly0

[back to top]

Number of Participants With Identified Electrocardiogram (ECG) Abnormalities

ECG abnormalities were defined as findings that are clinically meaningful as judged by the investigator. A 12-lead ECG was recorded at least 5 minutes after the participant had been lying down and all ECG recordings were evaluated by the investigator. Abnormalities, if present at any study time point, were listed. (NCT00646776)
Timeframe: Pre-dose on Day -1 and study discharge.

InterventionParticipants (Number)
RIB 150 mg Once Daily (QD)0
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly3

[back to top]

T-half of RTV

T-half was obtained directly from the concentration-time data. (NCT00646776)
Timeframe: Pre-dose (0h) on Days 4, 8, 11 to 18 and post-dose (1h,2h,3h,4h,6h,8h,12h) on Days 11 and 15 for ATV/RTV + RIB.

InterventionHour (Mean)
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly4.45

[back to top]

Terminal Elimination Half-life (T-half) of ATV

T-half was obtained directly from the concentration-time data. T-half following doses administered for treatment ATV/RTV 300/100 mg QD+RIB 150 mg twice weekly. (NCT00646776)
Timeframe: Pre-dose (0h) on Days 4, 8, 11 to 18 and post-dose (1h,2h,3h,4h,6h,8h,12h) on Days 11 and 15 for ATV/RTV + RIB.

InterventionHour (Mean)
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly11.89

[back to top]

Number of Participants Who Died, Experienced Other Serious Adverse Events (SAEs), Experienced Adverse Events (AEs) and Experienced Events Leading to Discontinuation.

AEs were defined as new, untoward medical occurrences/worsening of pre-existing medical condition, whether drug-related or not. SAEs were defined as any AE that: resulted in death; was life threatening; resulted in a persistent or significant disability/incapacity; resulted in/prolonged an existing in-patient hospitalization; was a congenital anomaly/birth defect; was a cancer; or was an overdose. Discontinuation from the study was due either to an AE or was conducted at the investigator's discretion. (NCT00646776)
Timeframe: From Day 1 to 30 days after the last dose of study drug.

,
InterventionParticipants (Number)
DeathsOther SAEsAEsAE leading to discontinuationDiscontinuation due to investigator discretion
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly021394
RIB 150 mg Once Daily (QD)00510

[back to top]

Number of Participants With Marked Abnormalities (MAs) in Hematology: Hemoglobin, Hematocrit, Platelet Count and Leukocytes

MAs are laboratory measurements marked as abnormal, per pre-defined study criteria, at any study time point. The following hematology MA definitions specify the criteria for the data presented. Hemoglobin/hematocrit: <0.85 x pre-treatment (pre-Rx) value. Platelet count: <0.85 x lower limit of normal (LLN) (or, if pre-Rx value 1.5 x upper limit of normal (ULN). Leukocytes: <0.9 x LLN or >1.2 x ULN (or, if pre-Rx value ULN. If pre-Rx value >ULN, then >1.15 x pre-Rx or NCT00646776)
Timeframe: Pre-dose on Day -1 and post-dose on Days 3, 7, 11, 14, 20 and 26 for RIB 150 mg QD; and pre-dose on Day -1 and post-dose on Days 3, 7, 11, 14 and 18 for ATV/RTV 300/100 mg QD + RIB 150 mg twice weekly.

,
InterventionParticipants (Number)
HemoglobinHematocritPlatelet countLeukocytes
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly1107
RIB 150 mg Once Daily (QD)0001

[back to top]

Number of Participants With MAs in Hematology: Neutrophils + Bands (Absolute), Lymphocytes (Absolute), Monocytes (Absolute), Basophils (Absolute) and Eosinophils (Absolute)

MAs are laboratory measurements marked as abnormal, per pre-defined study criteria, at any study time point. The following hematology MA definitions specify the criteria for the data presented. Neutrophils+bands (absolute): <=1.50 x 10^3 cells/microliter (uL). Lymphocytes (absolute): <0.75 x 10^3 cells/uL or >7.50 x 10^3 cells/uL. Monocytes (absolute): >2.00 x 10^3 cells/uL. Basophils (absolute): >0.40 x 10^3 cells/uL. Eosinophils (absolute): >0.75 x 10^3 cells/uL. (NCT00646776)
Timeframe: Pre-dose on Day -1 and post-dose on Days 3, 7, 11, 14, 20 and 26 for RIB 150 mg QD; and pre-dose on Day -1 and post-dose on Days 3, 7, 11, 14 and 18 for ATV/RTV 300/100 mg QD + RIB 150 mg twice weekly.

,
InterventionParticipants (Number)
Neutrophils+bands (absolute)Lymphocytes (absolute)Monocytes (absolute)Basophils (absolute)Eosinophils (absolute)
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly105000
RIB 150 mg Once Daily (QD)11000

[back to top]

Time to Reach Maximum Observed Plasma Concentration (Tmax) of ATV

Tmax was derived from the plasma concentration versus time for ATV and was recorded directly from experimental observations for each treatment period. (NCT00646776)
Timeframe: Pre-dose (0h) on Days 4, 8, 11 to 18 and post-dose (1h,2h,3h,4h,6h,8h,12h) on Days 11 and 15 for ATV/RTV + RIB.

InterventionHour (Median)
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly2.00

[back to top]

Tmax of RTV

Tmax was derived from the plasma concentration versus time for RTV and was recorded directly from experimental observations for each treatment period. (NCT00646776)
Timeframe: Pre-dose (0h) on Days 4, 8, 11 to 18 and post-dose (1h,2h,3h,4h,6h,8h,12h) on Days 11 and 15 for ATV/RTV + RIB.

InterventionHour (Median)
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly4.00

[back to top]

Number of Participants With MAs in Serum Chemistry: Alkaline Phosphatase (ALP),Aspartate Aminotransferase (AST),Alanine Aminotransferase (ALT),Bilirubin (Total),Bilirubin (Direct),Blood Urea Nitrogen (BUN),Creatinine,Sodium (Serum),Potassium (Serum)

MAs=laboratory measurements marked as abnormal, per pre-defined study criteria, at any study time point. The following serum chemistry MA definitions specify MA criteria. ALP, AST, ALT:>1.25xULN (if pre-Rx >ULN, then >1.25xpre-Rx). Bilirubin (total), bilirubin (direct), BUN:>1.1xULN (if pre-Rx >ULN, then >1.25xpre-Rx). Creatinine:>1.33xpre-Rx. Sodium (serum):<0.95xLLN or >1.05xULN (if pre-Rx ULN. If pre-Rx >ULN, then >1.05xpre-Rx or 1.1xULN (if pre-Rx ULN. If pre-Rx >ULN, then >1.1xpre-Rx or NCT00646776)
Timeframe: Pre-dose on Day -1 and post-dose on Days 3, 7, 11, 14, 20 and 26 for RIB 150 mg QD; and pre-dose on Day -1 and post-dose on Days 3, 7, 11, 14 and 18 for ATV/RTV 300/100 mg QD + RIB 150 mg twice weekly.

,
InterventionParticipants (Number)
ALPASTALTBilirubin (total)Bilirubin (direct)BUNCreatinineSodium (serum)Potassium (serum)
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly00017160100
RIB 150 mg Once Daily (QD)000000000

[back to top]

Number of Participants With MAs in Serum Chemistry: Chloride (Serum), Calcium (Total), Protein (Total), Bicarbonate, Phosphorous (Inorganic)

MAs=laboratory measurements marked as abnormal, per pre-defined study criteria, at any study time point. The following serum chemistry MA definitions specify MA criteria. Chloride (serum), calcium (total), protein (total):<0.9xLLN or >1.1xULN (if pre-Rx ULN. If pre-Rx >ULN, then >1.1xpre-Rx or 1.2xULN (if pre-Rx value ULN. If pre-Rx >ULN, then >1.2xpre-Rx value or 1.25xULN (if pre-Rx ULN, then >1.25x re-Rx or NCT00646776)
Timeframe: Pre-dose on Day -1 and post-dose on Days 3, 7, 11, 14, 20 and 26 for RIB 150 mg QD; and pre-dose on Day -1 and post-dose on Days 3, 7, 11, 14 and 18 for ATV/RTV 300/100 mg QD + RIB 150 mg twice weekly.

,
InterventionParticipants (Number)
Chloride (serum)Calcium (total)Protein (total)BicarbonatePhosphorous (inorganic)
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly00000
RIB 150 mg Once Daily (QD)00000

[back to top]

Number of Participants With MAs in Serum Chemistry: Glucose (Fasting Serum), Albumin, Creatine Kinase, Uric Acid, Lactate Dehydrogenase (LDH)

MAs=laboratory measurements marked as abnormal, per pre-defined study criteria, at any study time point. The following serum chemistry MA definitions specify MA criteria. Glucose (fasting serum): <0.8 x LLN or >1.5 ULN (if pre-Rx ULN. If pre-Rx >ULN, then >2.0 x pre-Rx or 1.5 x ULN (if pre-Rx >ULN, then >1.5 x pre-Rx). Uric acid: >1.2 x ULN (if pre-Rx >ULN, then >1.25 x pre-Rx). LDH: >1.25 x ULN (if pre-Rx >ULN, then >1.5 x pre-Rx). (NCT00646776)
Timeframe: Pre-dose on Day -1 and post-dose on Days 3, 7, 11, 14, 20 and 26 for RIB 150 mg QD; and pre-dose on Day -1 and post-dose on Days 3, 7, 11, 14 and 18 for ATV/RTV 300/100 mg QD + RIB 150 mg twice weekly.

,
InterventionParticipants (Number)
Glucose (Fasting Serum)AlbuminCreatine KinaseUric AcidLDH
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly00000
RIB 150 mg Once Daily (QD)00000

[back to top]

Number of Participants With MAs in Urinalysis

MAs are laboratory measurements marked as abnormal, per pre-defined study criteria, at any study time point. The following definitions specify the criteria for MAs. Protein, glucose and blood: >=2+ (or, if pre-treatment value >=1+, then >= 2 x pre-treatment value). (NCT00646776)
Timeframe: Pre-dose on Day -1, Day 7 and discharge.

,
InterventionParticipants (Number)
ProteinGlucoseBlood
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly101
RIB 150 mg Once Daily (QD)000

[back to top]

AUC(TAU) for ATV

AUC(TAU) was derived from the plasma concentration versus time for ATV, and was calculated by linear and log-linear trapezoidal summations using a mixed log-linear algorithm. (NCT00646776)
Timeframe: Pre-dose (0h) on Days 4, 8, 11 to 18 and post-dose (1h,2h,3h,4h,6h,8h,12h) on Days 11 and 15 for ATV/RTV + RIB.

Interventionng*h/mL (Geometric Mean)
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly51795

[back to top]

AUC(TAU) for RTV

AUC(TAU) was derived from the plasma concentration versus time for RTV, and was calculated by linear and log-linear trapezoidal summations using a mixed log-linear algorithm. (NCT00646776)
Timeframe: Pre-dose (0h) on Days 4, 8, 11 to 18 and post-dose (1h,2h,3h,4h,6h,8h,12h) on Days 11 and 15 for ATV/RTV + RIB.

Interventionng*h/mL (Geometric Mean)
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly8699

[back to top]

AUC24avg for 25-O-Desacetyl-RIB

AUC24avg is AUC(0-24 hour) following dosing on Day 10 for RIB 150 mg QD; AUC24avg is the area under the plasma concentration-time curve in 1 dosing interval (AUC[TAU]) divided by the number of days over the sampling duration for ATV/RTV 300/100 mg QD+RIB 150 mg twice weekly, i.e. AUC(TAU)/7 (NCT00646776)
Timeframe: Pre-dose (0h) on Days 6, 8, 10, and 11, and post-dose (1h,2h,3h,4h,6h,8h,12h) on Day 10 for RIB 150 mg QD; and pre-dose (0h) on Days 4, 8,11 to 18 and post-dose (1h,2h,3h,4h,6h,8h,12h) on Days 11 and 15 for ATV/RTV + RIB.

Interventionng*h/mL (Geometric Mean)
RIB 150 mg Once Daily (QD)117.7
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly1283

[back to top]

Average Area Under the Plasma Concentration-time Curve for 24 Hours (AUC24avg) for Rifabutin (RIB)

AUC24avg is AUC(0-24 hour) following dosing on Day 10 for RIB 150mg once daily (QD); AUC24avg is the area under the plasma concentration-time curve in 1 dosing interval (AUC[TAU]) divided by the number of days over the sampling duration for ATV/RTV 300/100 mg QD+RIB 150 mg twice weekly, i.e. AUC(TAU)/7. (NCT00646776)
Timeframe: Pre-dose (0 hours [h]) on Days 6, 8, 10, and 11, and post-dose (1h,2h,3h,4h,6h,8h,12h) on Day 10 for RIB 150 mg QD; and pre-dose (0h) on Days 4, 8,11 to 18 and post-dose (1h,2h,3h,4h,6h,8h,12h) on Days 11 and 15 for ATV/RTV + RIB.

Interventionnanograms*hour /milliliters (ng*h/mL) (Geometric Mean)
RIB 150 mg Once Daily (QD)1565
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly2311

[back to top]

Cmax of 25-O-Desacetylrifabutin (25-O-Desacetyl-RIB)

Cmax was derived from the plasma concentration versus time for 25-O-Desacetyl-RIB (a metabolite of RIB) and was recorded directly from experimental observations for each treatment period. (NCT00646776)
Timeframe: Pre-dose (0h) on Days 6, 8, 10, and 11, and post-dose (1h,2h,3h,4h,6h,8h,12h) on Day 10 for RIB 150 mg QD; and pre-dose (0h) on Days 4, 8,11 to 18 and post-dose (1h,2h,3h,4h,6h,8h,12h) on Days 11 and 15 for ATV/RTV + RIB.

Interventionng/mL (Geometric Mean)
RIB 150 mg Once Daily (QD)13.44
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly104.36

[back to top]

Cmax of ATV

Cmax was derived from the plasma concentration versus time for ATV and was recorded directly from experimental observations for each treatment period. (NCT00646776)
Timeframe: Pre-dose (0h) on Days 4, 8, 11 to 18 and post-dose (1h,2h,3h,4h,6h,8h,12h) on Days 11 and 15 for ATV/RTV + RIB.

Interventionng/mL (Geometric Mean)
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly5633

[back to top]

Total Area Under the Plasma Concentration-time Curve (AUCtot)

AUCtot represents the total free RIB plus 25-O-Desacetyl-RIB output. It is calculated as: AUCtot (micromolar[µM]*h) = AUC24avg(RIB)(ng*h/mL)/847.016 (g/mole) + AUC24avg(25-O-Desacetyl-RIB)(ng*h/mL)/804.979(g/mole). The 300 mg RIB arm represents an extrapolation from the 150 mg RIB group. (NCT00646776)
Timeframe: Pre-dose (0h) on Days 6, 8, 10, and 11, and post-dose (1h,2h,3h,4h,6h,8h,12h) on Day 10 for RIB 150 mg QD; and pre-dose (0h) on Days 4, 8,11 to 18 and post-dose (1h,2h,3h,4h,6h,8h,12h) on Days 11 and 15 for ATV/RTV + RIB.

InterventionµM*h (Geometric Mean)
RIB 150 mg Once Daily (QD)2.00
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly4.38

[back to top]

Cmax of RTV

Cmax was derived from the plasma concentration versus time for RTV and was recorded directly from experimental observations for each treatment period. (NCT00646776)
Timeframe: Pre-dose (0h) on Days 4, 8, 11 to 18 and post-dose (1h,2h,3h,4h,6h,8h,12h) on Days 11 and 15 for ATV/RTV + RIB.

Interventionng/mL (Geometric Mean)
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly1466

[back to top]

Cmin of 25-O-Desacetyl-RIB

Cmin was derived from plasma concentration versus time for 25-O-Desacetyl-RIB. (NCT00646776)
Timeframe: Pre-dose (0h) on Days 6, 8, 10, and 11, and post-dose (1h,2h,3h,4h,6h,8h,12h) on Day 10 for RIB 150 mg QD; and pre-dose (0h) on Days 4, 8,11 to 18 and post-dose (1h,2h,3h,4h,6h,8h,12h) on Days 11 and 15 for ATV/RTV + RIB.

Interventionng/mL (Geometric Mean)
RIB 150 mg Once Daily (QD)2.79
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly31.97

[back to top]

Cmin of ATV

Cmin was derived from the plasma concentration versus time for ATV. (NCT00646776)
Timeframe: Pre-dose (0h) on Days 4, 8, 11 to 18 and post-dose (1h,2h,3h,4h,6h,8h,12h) on Days 11 and 15 for ATV/RTV + RIB.

Interventionng/mL (Geometric Mean)
ATV/RTV 300/100 mg QD+RIB 150 mg Twice Weekly920.69

[back to top]

Weeks to HIV-1 RNA <200 Copies/ml

time to viral suppression noted as week on study treatment to attain HIV-1 RNA < 200 copies/ml (NCT00654147)
Timeframe: from date of treatment start to first week documented viral suppression

,
Interventionweek to viral supresssion (Median)
week to <200 Copies/mlweek to <50 Copies/ml
Raltegravir & Emtricitabine/Tenofovir2856
Raltegravir & Lopinavir/Ritonavir2856

[back to top]

Change From Baseline CD4+ and CD8+ Cell Counts

mean change in CD4+ and CD8+ T-lymphocytes counts from baseline (defined as the average of pre-entry and entry values) at weeks 16 and 24 in the two treatment arms (NCT00654147)
Timeframe: Baseline, Weeks 16 and 24

,
Interventioncells/mm3 (Mean)
week 16 CD4 cellsweek 24 CD4 cells
Raltegravir & Emtricitabine/Tenofovir452.11482.36
Raltegravir & Lopinavir/Ritonavir516.34521.31

[back to top]

Time to Virologic Failure

time to virologic failure at week 24 (up to 48 weeks) (NCT00654147)
Timeframe: week 24 (up to 48 weeks)

Interventionweeks (Median)
Raltegravir & Lopinavir/Ritonavir3.2296
Raltegravir & Emtricitabine/Tenofovir2.9952

[back to top]

Time to Confirmed Virologic Failure

time to confirmed viologic failure at 24 weeks (up to 48 weeks) (NCT00654147)
Timeframe: weeks

Interventionweeks (Median)
Raltegravir & Lopinavir/Ritonavir28
Raltegravir & Emtricitabine/Tenofovir29

[back to top] [back to top]

Study Medication Tolerability

study treatment tolerability as measured by number of subjects receiving study treatment who either discontinued or changed any component of study treatment (NCT00654147)
Timeframe: date started study treatment to first week documented change study treatment up to week 48

Interventionparticipants (Number)
Raltegravir & Lopinavir/Ritonavir1
Raltegravir & Emtricitabine/Tenofovir0

[back to top]

Plasma Viral Loads (HIV-1 RNA PCR)

Percentage subjects with undetectable Plasma viral loads (NCT00700115)
Timeframe: baseline to week 48

Interventionpercentage of subjects (Number)
Kaletra + Isentress92.7
Standard HAART88

[back to top]

To Compare Plasma Triglyceride Levels at 48 Weeks Between LPV/r + RAL and Standard HAART Treated Subjects

(NCT00700115)
Timeframe: 48 weeks

Interventionmg/dL (Mean)
Kaletra + Isentress238.1
Standard HAART133.3

[back to top]

Mean Change From Baseline in Triglycerides (Micromoles/Liter)

Included in measures of metabolic toxicity (NCT00711009)
Timeframe: Baseline to Week 96

Interventionmicromoles/liter (Mean)
LPV/r + FTC/TDF0.846
LPV/r + RAL1.103

[back to top]

Mean Change From Baseline in Total Protein (Grams/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventiongrams/liter (Mean)
LPV/r + FTC/TDF-6.3
LPV/r + RAL-7.2

[back to top]

Mean Change From Baseline in Total Bilirubin (Micromoles/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionmicromoles/liter (Mean)
LPV/r + FTC/TDF0.9
LPV/r + RAL1.9

[back to top]

Mean Change From Baseline in Temperature (°F)

(NCT00711009)
Timeframe: Baseline to Week 96

Intervention°F (Mean)
LPV/r + FTC/TDF-0.152
LPV/r + RAL-0.183

[back to top]

Mean Change From Baseline in Soluble Tumor Necrosis Factor Receptor-2 (Picograms/Milliliter)

Included in measures of metabolic toxicity (NCT00711009)
Timeframe: Baseline to Week 96

Interventionpicograms/milliliter (Mean)
LPV/r + FTC/TDF-1257.9
LPV/r + RAL-1594.7

[back to top]

Mean Change From Baseline in Soluble Tumor Necrosis Factor Receptor-1 (Picograms/Milliliter)

Included in measures of metabolic toxicity (NCT00711009)
Timeframe: Baseline to Week 96

Interventionpicograms/milliliter (Mean)
LPV/r + FTC/TDF-138.602
LPV/r + RAL-166.403

[back to top]

Mean Change From Baseline in Sodium (Micromoles/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionmicromoles/liter (Mean)
LPV/r + FTC/TDF0.1
LPV/r + RAL0.7

[back to top]

Mean Change From Baseline in Sitting Systolic Blood Pressure (mm Hg)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionmm Hg (Mean)
LPV/r + FTC/TDF-0.7
LPV/r + RAL-2.4

[back to top]

Mean Change From Baseline in Sitting Diastolic Blood Pressure (mm Hg)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionmm Hg (Mean)
LPV/r + FTC/TDF-2.4
LPV/r + RAL-1.8

[back to top]

Mean Change From Baseline in Red Blood Cell Count (x 10^12/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionnumber of cells x 10^12/liter (Mean)
LPV/r + FTC/TDF0.12
LPV/r + RAL0.16

[back to top]

Mean Change From Baseline in Potassium (Micromoles/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionmicromoles/liter (Mean)
LPV/r + FTC/TDF0.13
LPV/r + RAL0.03

[back to top]

Mean Change From Baseline in Platelet Count (x 10^9/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionnumber of cells x 10^9/liter (Mean)
LPV/r + FTC/TDF46.8
LPV/r + RAL34.2

[back to top]

Mean Change From Baseline in Neutrophils (x 10^9/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionnumber of cells x 10^9/liter (Mean)
LPV/r + FTC/TDF0.509
LPV/r + RAL0.705

[back to top]

Mean Change From Baseline in Monocytes (x 10^9/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionnumber of cells x 10^9/liter (Mean)
LPV/r + FTC/TDF0.065
LPV/r + RAL0.112

[back to top]

Mean Change From Baseline in Mid-Thigh Measurement (cm)

Mid-thigh circumference is included in the measures of somatic toxicity, which is characterized by loss of fat in the face, arms, and legs, and increase in fat in the base of the back of the neck and in the abdomen. Particpant's thigh circumference was measured halfway between the inguinal crease and the midpoint of the upper border of the patella using non-stretchable measuring tape with half centimeter marks. (NCT00711009)
Timeframe: Baseline to Week 96

Interventioncm (Mean)
LPV/r + FTC/TDF2.09
LPV/r + RAL5.13

[back to top]

Mean Change From Baseline in Mid-Arm Measurement (cm)

Arm circumference is included in the measures of somatic toxicity, which is characterized by loss of fat in the face, arms, and legs, and increase in fat in the base of the back of the neck and in the abdomen. Particpant's arm circumference was measured halfway between the acromial process on the shoulder and the tip of the elbow (olecranon process) using non-stretchable measuring tape with half centimeter marks. (NCT00711009)
Timeframe: Baseline to Week 96

Interventioncm (Mean)
LPV/r + FTC/TDF1.76
LPV/r + RAL4.71

[back to top]

Mean Change From Baseline in Magnesium (Millimoles/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionmillimoles/liter (Mean)
LPV/r + FTC/TDF0.019
LPV/r + RAL-0.009

[back to top]

Mean Change From Baseline in Lymphocytes (x 10^9/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionnumber of cells x 10^9/liter (Mean)
LPV/r + FTC/TDF0.332
LPV/r + RAL0.368

[back to top]

Mean Change From Baseline in Low Density Lipoprotein (LDL): High Density Lipoprotein (HDL) Ratio (Ratio)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionratio (Mean)
LPV/r + FTC/TDF-0.056
LPV/r + RAL-0.040

[back to top]

Mean Change From Baseline in Low Density Lipoprotein (LDL) (Micromoles/Liter)

Included in measures of metabolic toxicity (NCT00711009)
Timeframe: Baseline to Week 96

Interventionmicromoles/liter (Mean)
LPV/r + FTC/TDF0.535
LPV/r + RAL0.715

[back to top]

Mean Change From Baseline in Lipase (Units/Liter)

Included in measures of metabolic toxicity (NCT00711009)
Timeframe: Baseline to Week 96

Interventionunits/liter (Mean)
LPV/r + FTC/TDF4.674
LPV/r + RAL-1.898

[back to top]

Mean Change From Baseline in Leptin (Nanograms/Milliliter)

Included in measures of metabolic toxicity (NCT00711009)
Timeframe: Baseline to Week 96

Interventionnanograms/milliliter (Mean)
LPV/r + FTC/TDF3.623
LPV/r + RAL2.927

[back to top]

Mean Change From Baseline in Lactate Dehydrogenase (Units/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionunits/liter (Mean)
LPV/r + FTC/TDF-21.157
LPV/r + RAL-28.926

[back to top]

Mean Change From Baseline in Lactate (Millimoles/Liter)

Included in measures of metabolic toxicity (NCT00711009)
Timeframe: Baseline to Week 96

Interventionmillimoles/liter (Mean)
LPV/r + FTC/TDF0.281
LPV/r + RAL0.444

[back to top]

Mean Change From Baseline in Interleukin-6 (Nanograms/Liter)

Included in measures of metabolic toxicity (NCT00711009)
Timeframe: Baseline to Week 96

Interventionnanograms/liter (Mean)
LPV/r + FTC/TDF-1.584
LPV/r + RAL-53.286

[back to top]

Mean Change From Baseline in Insulin (Picomoles/Liter)

Included in measures of metabolic toxicity (NCT00711009)
Timeframe: Baseline to Week 96

Interventionpicomoles/liter (Mean)
LPV/r + FTC/TDF-6.724
LPV/r + RAL4.441

[back to top]

Mean Change From Baseline in Inorganic Phosphate (Micromoles/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionmicromoles/liter (Mean)
LPV/r + FTC/TDF-0.046
LPV/r + RAL-0.028

[back to top]

Mean Change From Baseline in Hips Measurement (cm)

Hip circumference is included in the measures of somatic toxicity, which is characterized by loss of fat in the face, arms, and legs, and increase in fat in the base of the back of the neck and in the abdomen. Participant was measured at widest width of the hip using non-stretchable measuring tape with half centimeter marks. (NCT00711009)
Timeframe: Baseline to Week 96

Interventioncm (Mean)
LPV/r + FTC/TDF2.45
LPV/r + RAL4.70

[back to top]

Mean Change From Baseline in High Density Lipoprotein Cholesterol (HDL) (Micromoles/Liter)

Included in measures of metabolic toxicity (NCT00711009)
Timeframe: Baseline to Week 96

Interventionmicromoles/liter (Mean)
LPV/r + FTC/TDF0.257
LPV/r + RAL0.346

[back to top]

Mean Change From Baseline in Hematocrit (Fraction)

Hematocrit fraction is the percentage (%) by volume of packed red blood cells (RBCs) in the participant's blood. It was measured using standard clinical laboratory analysis of participants' blood samples. (NCT00711009)
Timeframe: Baseline to Week 96

Intervention% by volume of packed RBCs in blood (Mean)
LPV/r + FTC/TDF0.038
LPV/r + RAL0.036

[back to top]

Mean Change From Baseline in Fasting Glucose (Millimoles/Liter)

Included in measures of metabolic toxicity (NCT00711009)
Timeframe: Baseline to Week 96

Interventionmillimoles/liter (Mean)
LPV/r + FTC/TDF-0.011
LPV/r + RAL0.109

[back to top]

Mean Change From Baseline in Eosinophils (x 10^9/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionnumber of cells x 10^9/liter (Mean)
LPV/r + FTC/TDF-0.012
LPV/r + RAL0.015

[back to top]

Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Upper Extremity Total Mass (Grams)

The dual energy X-ray absorptiometry (DEXA) scan is included in the measures of somatic toxicity, which is characterized by loss of fat in the face, arms, and legs, and increase in fat in the base of the back of the neck and in the abdomen. (NCT00711009)
Timeframe: Baseline to Week 96

Interventiongrams (Mean)
LPV/r + FTC/TDF-0.33
LPV/r + RAL1.52

[back to top]

Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Upper Extremity Lean Mass (Grams)

The dual energy X-ray absorptiometry (DEXA) scan is included in the measures of somatic toxicity, which is characterized by loss of fat in the face, arms, and legs, and increase in fat in the base of the back of the neck and in the abdomen. (NCT00711009)
Timeframe: Baseline to Week 96

Interventiongrams (Mean)
LPV/r + FTC/TDF-1.49
LPV/r + RAL-1.25

[back to top]

Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Upper Extremity Fat (Grams)

The dual energy X-ray absorptiometry (DEXA) scan is included in the measures of somatic toxicity, which is characterized by loss of fat in the face, arms, and legs, and increase in fat in the base of the back of the neck and in the abdomen. (NCT00711009)
Timeframe: Baseline to Week 96

Interventiongrams (Mean)
LPV/r + FTC/TDF7.28
LPV/r + RAL21.53

[back to top]

Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Trunk Mass (Grams)

The dual energy X-ray absorptiometry (DEXA) scan is included in the measures of somatic toxicity, which is characterized by loss of fat in the face, arms, and legs, and increase in fat in the base of the back of the neck and in the abdomen. (NCT00711009)
Timeframe: Baseline to Week 96

Interventiongrams (Mean)
LPV/r + FTC/TDF3.48
LPV/r + RAL6.34

[back to top]

Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Trunk Lean Mass (Grams)

The dual energy X-ray absorptiometry (DEXA) scan is included in the measures of somatic toxicity, which is characterized by loss of fat in the face, arms, and legs, and increase in fat in the base of the back of the neck and in the abdomen. (NCT00711009)
Timeframe: Baseline to Week 96

Interventiongrams (Mean)
LPV/r + FTC/TDF1.67
LPV/r + RAL2.56

[back to top]

Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Trunk Fat (Grams)

The dual energy X-ray absorptiometry (DEXA) scan is included in the measures of somatic toxicity, which is characterized by loss of fat in the face, arms, and legs, and increase in fat in the base of the back of the neck and in the abdomen. (NCT00711009)
Timeframe: Baseline to Week 96

Interventiongrams (Mean)
LPV/r + FTC/TDF13.75
LPV/r + RAL27.01

[back to top]

Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Total Body Mass (Grams)

The dual energy X-ray absorptiometry (DEXA) scan is included in the measures of somatic toxicity, which is characterized by loss of fat in the face, arms, and legs, and increase in fat in the base of the back of the neck and in the abdomen. (NCT00711009)
Timeframe: Baseline to Week 96

Interventiongrams (Mean)
LPV/r + FTC/TDF2.9
LPV/r + RAL5.4

[back to top]

Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Total Body Lean Mass (Grams)

The dual energy X-ray absorptiometry (DEXA) scan is included in the measures of somatic toxicity, which is characterized by loss of fat in the face, arms, and legs, and increase in fat in the base of the back of the neck and in the abdomen. (NCT00711009)
Timeframe: Baseline to Week 96

Interventiongrams (Mean)
LPV/r + FTC/TDF1.08
LPV/r + RAL1.56

[back to top]

Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Lower Extremity Total Mass (Grams)

The dual energy X-ray absorptiometry (DEXA) scan is included in the measures of somatic toxicity, which is characterized by loss of fat in the face, arms, and legs, and increase in fat in the base of the back of the neck and in the abdomen. (NCT00711009)
Timeframe: Baseline to Week 96

Interventiongrams (Mean)
LPV/r + FTC/TDF4.32
LPV/r + RAL6.96

[back to top]

Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Lower Extremity Fat (Grams)

The dual energy X-ray absorptiometry (DEXA) is included in the measures of somatic toxicity, which is characterized by loss of fat in the face, arms, and legs, and increase in fat in the base of the back of the neck and in the abdomen. (NCT00711009)
Timeframe: Baseline to Week 96

Interventiongrams (Mean)
LPV/r + FTC/TDF15.32
LPV/r + RAL28.82

[back to top]

Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Bone Mineral Density (Grams/cm^2)

The dual energy X-ray absorptiometry (DEXA) scan of bone mineral content was used to evaluate potential bone effects of treatment. (NCT00711009)
Timeframe: Baseline to Week 96

Interventiongrams/cm^2 (Mean)
LPV/r + FTC/TDF-2.48
LPV/r + RAL0.68

[back to top]

Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Bone Mineral Content (Grams)

The dual energy X-ray absorptiometry (DEXA) scan of bone mineral content was used to evaluate potential bone effects of treatment. (NCT00711009)
Timeframe: Baseline to Week 96

Interventiongrams (Mean)
LPV/r + FTC/TDF-3.69
LPV/r + RAL0.52

[back to top]

Mean Change From Baseline in DEXA Scan of Lower Extremity Lean Mass (Grams)

The dual energy X-ray absorptiometry (DEXA) scan is included in the measures of somatic toxicity, which is characterized by loss of fat in the face, arms, and legs, and increase in fat in the base of the back of the neck and in the abdomen. (NCT00711009)
Timeframe: Baseline to Week 96

Interventiongrams (Mean)
LPV/r + FTC/TDF1.97
LPV/r + RAL2.27

[back to top]

Mean Change From Baseline in Creatinine (Micromoles/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionmicromoles/liter (Mean)
LPV/r + FTC/TDF5.7
LPV/r + RAL1.6

[back to top]

Mean Change From Baseline in Creatine Phosphokinase (Units/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionunits/liter (Mean)
LPV/r + FTC/TDF398.9
LPV/r + RAL157.2

[back to top]

Mean Change From Baseline in Cholesterol (Micromoles/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionmicromoles/liter (Mean)
LPV/r + FTC/TDF0.808
LPV/r + RAL1.113

[back to top]

Mean Change From Baseline in Chloride (Micromoles/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionmicromoles/liter (Mean)
LPV/r + FTC/TDF-0.4
LPV/r + RAL0.2

[back to top]

Mean Change From Baseline in Chest Measurement (cm)

Chest circumference is included in the measures of somatic toxicity, which is characterized by loss of fat in the face, arms, and legs, and increase in fat in the base of the back of the neck and in the abdomen. Participant's chest circumference was measured at 5 cm above the xiphoid process using non-stretchable measuring tape with half centimeter marks. (NCT00711009)
Timeframe: Baseline to Week 96

Interventioncm (Mean)
LPV/r + FTC/TDF1.13
LPV/r + RAL4.06

[back to top]

Mean Change From Baseline in Calculated Creatinine Clearance (Milliliters/Second)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionmilliliters/second (Mean)
LPV/r + FTC/TDF-0.122
LPV/r + RAL-0.024

[back to top]

Mean Change From Baseline in Calcium (Micromoles/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionmicromoles/liter (Mean)
LPV/r + FTC/TDF-0.040
LPV/r + RAL-0.016

[back to top]

Mean Change From Baseline in Blood Urea Nitrogen (Micromoles/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionmicromoles/liter (Mean)
LPV/r + FTC/TDF0.00
LPV/r + RAL0.37

[back to top]

Mean Change From Baseline in Bicarbonate (Micromoles/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionmicromoles/liter (Mean)
LPV/r + FTC/TDF-0.5
LPV/r + RAL-0.8

[back to top]

Mean Change From Baseline in Basophils (x 10^9/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionnumber of cells x 10^9/liter (Mean)
LPV/r + FTC/TDF0.005
LPV/r + RAL0.003

[back to top]

Mean Change From Baseline in Aspartate Aminotransferase (Units/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionunits/liter (Mean)
LPV/r + FTC/TDF-0.8
LPV/r + RAL-9.6

[back to top]

Mean Change From Baseline in Alkaline Phosphatase (Units/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionunits/liter (Mean)
LPV/r + FTC/TDF14.5
LPV/r + RAL1.7

[back to top]

Mean Change From Baseline in Albumin (Grams/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventiongrams/liter (Mean)
LPV/r + FTC/TDF1.4
LPV/r + RAL1.3

[back to top]

Mean Change From Baseline in Alanine Aminotransferase (Units/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionunits/liter (Mean)
LPV/r + FTC/TDF-6.1
LPV/r + RAL-13.4

[back to top]

Mean Change From Baseline in Adiponectin (Micrograms/Milliliter)

Included in measures of metabolic toxicity (NCT00711009)
Timeframe: Baseline to Week 96

Interventionmicrograms/milliliter (Mean)
LPV/r + FTC/TDF2.112
LPV/r + RAL2.064

[back to top]

Change From Baseline on Physical Component Score of the Medical Outcomes Study HIV Health Survey

The Survey is a brief, comprehensive health status measure used in studies of people with HIV/AIDS. Participants rate their health and mental/emotional condition, how much their health limits physical activities (eating, dressing, bathing, climbing stairs, walking one block, etc.) and social activities (for example, visiting with friends or relatives), and other questions that measure quality of life. The physical component summarizes answers to questions about physical status. Possible scores range from 0 to 100. A higher score indicates better health, and increases indicate improvement. (NCT00711009)
Timeframe: Baseline to Week 96

InterventionScores on a scale (Mean)
LPV/r + FTC/TDF-1.0
LPV/r + RAL-1.1

[back to top]

Change From Baseline on Mental Component of Medical Outcomes Study HIV Health Survey

The Survey is a brief, comprehensive health status measure used in studies of people with HIV/AIDS. Participants rate their health and mental/emotional condition, how much their health limits physical activities (eating, dressing, bathing, climbing stairs, walking one block, etc.) and social activities (visiting with friends or relatives, etc.), and other questions that measure quality of life. The mental component summarizes answers to questions about emotional and mental wellbeing. Possible scores range from 0 to 100. Higher scores indicates better health, and increases indicate improvement. (NCT00711009)
Timeframe: Baseline to Week 96

InterventionScores on a scale (Mean)
LPV/r + FTC/TDF1.3
LPV/r + RAL1.3

[back to top]

Mean Change From Baseline in Uric Acid (Micromoles/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionmicromoles/liter (Mean)
LPV/r + FTC/TDF-29.0
LPV/r + RAL-6.1

[back to top]

Mean Change From Baseline in Hemoglobin (Grams/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventiongrams/liter (Mean)
LPV/r + FTC/TDF5.4
LPV/r + RAL5.1

[back to top]

Mean Change From Baseline in Sitting Heart Rate (Beats Per Minute)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionbeats per minute (Mean)
LPV/r + FTC/TDF-4.6
LPV/r + RAL-6.3

[back to top]

Mean Change From Baseline in Dual Energy X-ray Absorptiometry (DEXA) Scan of Total Body Fat (Grams)

The dual energy X-ray absorptiometry (DEXA) scan is included in the measures of somatic toxicity, which is characterized by loss of fat in the face, arms, and legs, and increase in fat in the base of the back of the neck and in the abdomen. (NCT00711009)
Timeframe: Baseline to Week 96

Interventiongrams (Mean)
LPV/r + FTC/TDF12.71
LPV/r + RAL25.31

[back to top]

Primary Outcome: Percentage of Participants With Potentially Clinically Significant Laboratory Values

Potentially clinically significant laboratory values that occurred in at least 2% of participants in either treatment arm are presented. (NCT00711009)
Timeframe: Baseline to Week 96

,
InterventionPercentage of participants (Number)
Alananine aminotransferase >5x upper limit normalAspartate aminotransferase >5x upper limit normalCreatinine phosphokinase >4x upper limit of normalCalcium <1.75 millimoles/literCholesterol >7.77 millimoles/literTriglycerides >8.475 millimoles/literCalc. creatinine clearance <50 milliliters/minuteLipase >2x upper limit of normalNeutrophils < 0.75 x 10^9/literMagnesium < 0.5 millimoles/liter
LPV/r + FTC/TDF2.92.98.7013.54.83.87.73.80
LPV/r + RAL5.05.019.82.016.89.91.04.002.0

[back to top] [back to top]

Percentage of Participants Responding (Plasma HIV-1 RNA Levels Below 40 Copies/Milliliter [mL]) at Each Visit Based on the FDA Time to Loss of Virologic Response (TLOVR) Algorithm

A participant was classified as a responder at the first of 2 consecutive visits with plasma HIV-1 RNA levels below 40 copies/mL. The participant continued to be a responder until one of the following: 1) the participant had 2 consecutive values greater than or equal to 40 copies/mL; the final measurement, if the final measurement was the first one documenting an increase in plasma HIV-1 RNA level to greater than or equal to 40 copies/mL; the participant discontinued participation in the study or died. (NCT00711009)
Timeframe: Baseline to Week 96

,
InterventionPercentage of Participants (Number)
Week 2Week 4Week 8Week 16Week 24Week 32Week 40Week 48Week 60Week 72Week 84Week 96
LPV/r + FTC/TDF7.617.136.267.680.085.784.884.882.978.174.368.6
LPV/r + RAL33.763.475.281.283.285.187.183.275.271.370.366.3

[back to top]

Number of Participants Who Developed Resistance to Each Drug in the Study Regimen, as Defined by the International AIDS Society-USA (IAS-USA) Panel.

Resistance to study drugs was defined as described by the International AIDS Society-USA (IAS-USA) Panel. All participants had an HIV-1 drug resistance genotype (lopinavir/ritonavir, tenofovir, or emtricitabine) obtained at the Screening Visit. Beginning at Week 8, if participant's plasma HIV-1 RNA was greater than or equal to 40 copies/milliliter (mL) and was below 40 copies/mL at the previous visit, additional procedures were undertaken to determine if resistance to study drug occurred. (NCT00711009)
Timeframe: Baseline to Week 96

,
InterventionParticipants (Number)
Lopinavir resistanceEmtricitabine resistanceTenofovir resistanceRaltegravir resistance
LPV/r + FTC/TDF010NA
LPV/r + RAL0003

[back to top]

Mean Change in CD4+ T-Cell Counts From Baseline to Each Visit

(NCT00711009)
Timeframe: Baseline to Week 96

,
Interventioncells/microliter (Mean)
Week 4Week 8Week 16Week 24Week 32Week 40Week 48Week 60Week 72Week 84Week 96
LPV/r + FTC/TDF97.2107.9158.7154.9180.0204.6245.0243.4277.4309.6296.4
LPV/r + RAL113.4124.5141.6174.5188.2223.0241.9250.6269.9280.2281.0

[back to top]

Time to Loss of Virologic Response - Percentage of Participants Still Categorized as Responders at Day 672

Time of loss of virologic response was defined as the first of the following: first of 2 consecutive visits with plasma HIV-1 RNA greater than or equal to 40 copies/milliliter (mL), if the participant previously demonstrated 2 consecutive plasma HIV-1 RNA levels below 40 copies/mL; Study Day 1, if the subject never achieved 2 consecutive plasma HIV-1 RNA levels below 40 copies/mL; the day of the final measurement, if the final measurement was the first one documenting an increase in plasma HIV-1 RNA level to greater than or equal to 40 copies/mL. (NCT00711009)
Timeframe: Baseline to Week 96

InterventionPercentage of Participants (Number)
LPV/r + FTC/TDF79.1
LPV/r + RAL77.8

[back to top]

Score on Side Effects Scale of Treatment Satisfaction Questionnaire for Medication

The Side Effects scale of the TSQM asks if the participant experiences side effects (yes/no), and if so, how bothersome the side effects are, to what extent they interfere with physical health and ability to function (for example, strength and energy levels), to what extent they interfere with mental function (for example, ability to think clearly, stay awake, etc.), and to what extent the side effects affect the participants overall satisfaction with the medication. Scores are converted to a range of 0 to 100. Higher scores indicate less interference and/or less dissatisfaction. (NCT00711009)
Timeframe: Week 96

InterventionScores on a scale (Mean)
LPV/r + FTC/TDF84.6
LPV/r + RAL86.2

[back to top]

Score on Global Satisfaction Scale of Treatment Satisfaction Questionnaire for Medication

The Global Satisfaction scale of the TSQM evaluates the participants rating of whether the good things about the medication outweigh the bad things (1=not at all certain to 5=extremely certain) and how satisfied or dissatisfied the participant is with the medication (1=extremely dissatisfied to 7=extremely satisfied). Scores are converted to a range of 0 to 100. Higher scores indicate greater satisfaction. (NCT00711009)
Timeframe: Week 96

InterventionScores on a scale (Mean)
LPV/r + FTC/TDF82.5
LPV/r + RAL85.5

[back to top]

Score on Effectiveness Scale of Treatment Satisfaction Questionnaire for Medication (TSQM)

The Effectiveness Scale of the TSQM evaluates the participant's satisfaction or dissatisfaction (1=extremely dissatisfied to 7=extremely satisfied) with the ability of the medication to prevent or treat the condition, the way the medication relieves symptoms, the amount of time it takes for the medication to start working, and other questions. Scores are converted to a range of 0 to 100. A higher score indicates greater satisfaction. (NCT00711009)
Timeframe: Week 96

InterventionScores on a scale (Mean)
LPV/r + FTC/TDF75.5
LPV/r + RAL76.0

[back to top]

Percentage of Participants Responding (Plasma HIV-1 Ribonucleic Acid [RNA] Levels Less Than 40 Copies/Milliliter [mL]) at Week 48 Based on the Food and Drug Administration (FDA) Time to Loss of Virologic Response (TLOVR) Algorithm

A participant was classified as a responder at the first of 2 consecutive visits with plasma HIV-1 RNA levels below 40 copies/mL. The participant continued to be a responder until one of the following: the participant had 2 consecutive values greater than or equal to 40 copies/mL; the final measurement, if the final measurement was the first one documenting an increase in plasma HIV-1 RNA level to greater than or equal to 40 copies/mL; the participant discontinued participation in the study or died. (NCT00711009)
Timeframe: Baseline to Week 48

InterventionPercentage of Participants (Number)
LPV/r + FTC/TDF84.8
LPV/r + RAL83.2

[back to top]

Number of Participants Who Developed Resistance, Defined Conservatively, to Lopinavir

Beginning at Week 8, if participant's plasma HIV-1 RNA was greater than/equal to 40 copies/milliliter (mL) and was below 40 copies/mL at the previous visit, additional procedures were undertaken to determine if resistance occurred. Evidence of lopinavir resistance was more conservatively defined as the presence of 1 or more of these mutations: protease I47V or A, G48V, I50V, V82A or F or T or S, I84V, L90M; or presence of at least 3 or more of these mutations: protease L10F or I or R or V, K20M or R, L24I, V32I, L33F, M36I, M46I or L, F53L, any change to I54, A71V or T, and G73S. (NCT00711009)
Timeframe: Baseline to Week 96

InterventionParticipants (Number)
LPV/r + FTC/TDF0
LPV/r + RAL1

[back to top]

Mean Change From Baseline in White Blood Cell Count (x 10^9/Liter)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionnumber of cells x 10^9/liter (Mean)
LPV/r + FTC/TDF0.90
LPV/r + RAL1.20

[back to top]

Mean Change From Baseline in Weight (kg)

(NCT00711009)
Timeframe: Baseline to Week 96

Interventionkg (Mean)
LPV/r + FTC/TDF1.83
LPV/r + RAL3.77

[back to top]

Mean Change From Baseline in Waist Measurement (cm)

Waist circumference is included in the measures of somatic toxicity, which is characterized by loss of fat in the face, arms, and legs, and increase in fat in the base of the back of the neck and in the abdomen. Circumference of participant's waist was measured at the level of the navel using non-stretchable measuring tape with half centimeter marks. (NCT00711009)
Timeframe: Baseline to Week 96

Interventioncm (Mean)
LPV/r + FTC/TDF1.88
LPV/r + RAL4.93

[back to top]

Mean Change From Baseline in Urine Specific Gravity

Urine specific gravity is a laboratory test that measures the concentration of all chemical particles in the urine. The measurement produces a ratio of the urine density to water density. (NCT00711009)
Timeframe: Baseline to Week 96

Interventionratio of urine density to water density (Mean)
LPV/r + FTC/TDF0.0042
LPV/r + RAL0.0052

[back to top]

Mean Change From Baseline in Urine pH

(NCT00711009)
Timeframe: Baseline to Week 96

InterventionpH (Mean)
LPV/r + FTC/TDF0.00
LPV/r + RAL0.03

[back to top]

Half-life (t1/2)

Elimination half-life (t1/2) measured in hours: time required for half the quantity of maraviroc to be metabolized or eliminated by normal biological processes. (NCT00717067)
Timeframe: Pre-dose, post-dose hours 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48, and 72.

Interventionhour (Mean)
Healthy Subjects: Multiple Dose14.22
Mild Renal Impairment: Multiple Dose16.84
Moderate Renal Impairment: Multiple Dose16.99
Healthy Subjects: Single Dose14.36
Severe Renal Impairment: Single Dose17.29
ESRD: Single Dose; After Dialysis15.03
ESRD: Single Dose; Before Dialysis13.86

[back to top]

Hemodialysis Clearance of Maraviroc (MVC) in Subjects With End Stage Renal Disease (ESRD) Undergoing Hemodialysis: CLdD

CLdD: dialysate clearance before dialysis; measured in milliliters per minute. (NCT00717067)
Timeframe: Before dialysis

InterventionmL/min (Geometric Mean)
ESRD: Single Dose; Before Dialysis36.42

[back to top]

Maximum Observed Plasma Concentration (Cmax)

Maximum observed plasma concentration (Cmax) within the dosing interval; measured in nanograms per milliliter (ng/mL). (NCT00717067)
Timeframe: Pre-dose, post-dose hours 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48, and 72.

Interventionng/mL (Geometric Mean)
Healthy Subjects: Multiple Dose950.91
Mild Renal Impairment: Multiple Dose1150.74
Moderate Renal Impairment: Multiple Dose674.20
Healthy Subjects: Single Dose335.60
Severe Renal Impairment: Single Dose801.16
ESRD: Single Dose; After Dialysis576.7
ESRD: Single Dose; Before Dialysis478.5

[back to top]

Renal Clearance (CLR) in Subjects With Normal, Mild, Moderate and Severe Renal Function

Renal clearance (CLR) measured in milliliters per minute (mL/min). (NCT00717067)
Timeframe: Hour 0 (prior to MVC dosing [single dose] or prior to last MVC dose [multiple dose]) to 72 hours post-dose ; hours 0, 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48, and 72.

InterventionmL/min (Geometric Mean)
Healthy Subjects: Multiple Dose105.7
Mild Renal Impairment: Multiple Dose77.2
Moderate Renal Impairment: Multiple Dose62.5
Healthy Subjects: Single Dose110.0
Severe Renal Impairment: Single Dose26.6

[back to top]

Time of First Occurrence (Tmax)

Time (hours) of first occurrence (Tmax); time after dosing when Cmax (maximum plasma concentration) occured. (NCT00717067)
Timeframe: Pre-dose, post-dose hours 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48, and 72.

Interventionhours (Median)
Healthy Subjects: Multiple Dose1.000
Mild Renal Impairment: Multiple Dose1.500
Moderate Renal Impairment: Multiple Dose2.000
Healthy Subjects: Single Dose2.500
Severe Renal Impairment: Single Dose2.500
ESRD: Single Dose; After Dialysis3.000
ESRD: Single Dose; Before Dialysis2.000

[back to top]

Safety and Tolerability of Maraviroc in the Absence and Presence of a Potent CYP3A4 Inhibitor in Subjects With Various Degrees of Renal Impairment or Undergoing Hemodialysis: Number of Subjects With Maximum EGC QTC, QTCB and QTCF Intervals

Single 12-lead ECG: number of subjects with maximum QTC interval, maximum QTCB interval (Bazett's correction), and maximum QTCF interval (Friderica's correction) measured in milliseconds (msec); range: 450 to <480 msec, 480 to <500 msec, and >500 msec. Maximum QTC interval increase from Baseline; citeria: change = ≥ 30 msec to < 60 msec, and change = ≥ 60 msec. (NCT00717067)
Timeframe: Normal renal function: screening, Day -3 and Day -1; normal renal function, mild and moderate RI: Day 7 to Day 9 and follow-up; severe RI: screening, Day 1, Day 3, Day 4, and follow-up; ESRD: screening, Day 1, Day 3, Day 4, and follow-up

,,,,,,
Interventionsubjects (Number)
Maximum QTC Interval: 450 to < 480 msecMaximum QTC Interval: 480 to < 500 msecMaximum QTC Interval: > 500 msecMaximum QTCB Interval: 450 to < 480 msecMaximum QTCB Interval: 480 to < 500 msecMaximum QTCB Interval: > 500 msecMaximum QTCF Interval: 450 to 480 msecMaximum QTCF Interval: 480 to < 500 msecMaximum QTCF Interval: > 500 msecMax. QTC Interval Increase from BL: change ≥30 <60QTC Interval Increase from BL: change ≥ 60
ESRD: Single Dose; After Dialysis10000000000
ESRD: Single Dose; Before Dialysis00000000010
Healthy Subjects: Multiple Dose10010010000
Healthy Subjects: Single Dose10010000000
Mild Renal Impairment: Multiple Dose00000000000
Moderate Renal Impairment: Multiple Dose00000000000
Severe Renal Impairment: Single Dose20010010000

[back to top]

Safety and Tolerability of Maraviroc in the Absence and Presence of a Potent CYP3A4 Inhibitor in Subjects With Various Degrees of Renal Impairment or Undergoing Hemodialysis: Number of Subjects With Maximum Increase and Decrease in Supine Blood Pressure

Number of subjects with absolute values of supine systolic blood pressure (BP) measured in millimeters of mercury (mm/Hg), range: <90 mmHg; and supine diastolic blood pressure, range: <50 mmHg. Number of subjects with a maximum increase and decrease from Baseline in supine systolic BP ≥ 30 mmHg. Number of subjects with a maximum increase and decrease from Baseline in supine diastolic BP ≥ 20 mmHg. (NCT00717067)
Timeframe: Normal renal function: screening, Day -3 to Day -1; normal, mild and moderate RI: Day 7 to Day 10 and follow-up; severe RI: Day 1 to Day 4 and follow-up; ESRD: Day 1, Day 4, and follow-up

,,,,,,
Interventionsubjects (Number)
BL Supine Systolic Blood Pressure (BP) <90 mmHgMaximum Increase from BL: Supine Systolic BP≥ 30Maximum Decrease from BL: Supine Systolic BP ≥ 30BL Supine Diastolic Blood Pressure (BP) <50 mmHgMaximum Increase from BL: Supine Diastolic BP≥ 20Maximun Decrease from BL: Supine Diastolic BP ≥20
ESRD: Single Dose; After Dialysis001000
ESRD: Single Dose; Before Dialysis000000
Healthy Subjects: Multiple Dose000000
Healthy Subjects: Single Dose000000
Mild Renal Impairment: Multiple Dose000000
Moderate Renal Impairment: Multiple Dose000010
Severe Renal Impairment: Single Dose000000

[back to top]

Area Under the Plasma Concentration Time-curve From Zero to the Last Measured Concentration (AUClast)

Area under the plasma concentration time-curve from zero to the last measured concentration (AUClast) measured in nanograms * hour divided by milliliters (ng*hr/mL). (NCT00717067)
Timeframe: Pre-dose, post-dose hours 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48, and 72.

Interventionng*hr/mL (Geometric Mean)
Healthy Subjects: Multiple Dose7356.3
Mild Renal Impairment: Multiple Dose9502.1
Moderate Renal Impairment: Multiple Dose6496.0
Healthy Subjects: Single Dose1320.7
Severe Renal Impairment: Single Dose4255.5
ESRD: Single Dose; After Dialysis2636.5
ESRD: Single Dose; Before Dialysis2770.1

[back to top]

Safety and Tolerability of Maraviroc in the Absence and Presence of a Potent CYP3A4 Inhibitor in Subjects With Various Degrees of Renal Impairment or Undergoing Hemodialysis: Number of Subjects With Pulse Rate < 40 and > 120 Beats Per Minute

Number of subjects with pulse rate < 40 beats per minute (BPM), number of subjects with pulse rate > 120 BPM. (NCT00717067)
Timeframe: Normal renal function: screening, Day -3 to Day -1; normal, mild and moderate RI: Day 7 to Day 10 and follow-up; severe RI: Day 1 to Day 4 and follow-up; ESRD: Day 1, Day 4, and follow-up

,,,,,,
Interventionbpm (Number)
Supine Pulse Rate <40 BPMSupine Pulse Rate >120 BPM
ESRD: Single Dose; After Dialysis00
ESRD: Single Dose; Before Dialysis00
Healthy Subjects: Multiple Dose00
Healthy Subjects: Single Dose00
Mild Renal Impairment: Multiple Dose00
Moderate Renal Impairment: Multiple Dose00
Severe Renal Impairment: Single Dose00

[back to top]

Derivation of Renal Clearance in Subjects With Normal, Mild, Moderate and Severe Renal Function: Ae

Ae: amount of drug excreted unchanged in the urine; measured in milligrams (mg). (NCT00717067)
Timeframe: Hour 0 (prior to MVC dosing [single dose] or prior to last MVC dose [multiple dose]) to 72 hours post-dose ; hours 0, 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48, and 72.

Interventionmg (Mean)
Healthy Subjects: Multiple Dose41.6
Mild Renal Impairment: Multiple Dose39.1
Moderate Renal Impairment: Multiple Dose24.9
Healthy Subjects: Single Dose10.8
Severe Renal Impairment: Single Dose8.4

[back to top]

Area Under the Time Curve From 0 to Infinity (AUCinf)

Area under the plasma concentration-time profile from time zero to the time infinate in subjects who received single dose treatment; measured in nanograms * hour divided by millilters (ng*hr/mL). (NCT00717067)
Timeframe: Pre-dose, post-dose hours 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48, and 72

Interventionng*hr/mL (Geometric Mean)
Healthy Subjects: Single Dose1348.4
Severe Renal Impairment: Single Dose4367.7
ESRD: Single Dose; After Dialysis2677.4
ESRD: Single Dose; Before Dialysis2805.5

[back to top]

AUCtau

AUCtau: area under the plasma concentration-time profile from time zero to the end of the dosing interval (tau); measured in nanograms * hours divided by milliliters (ng.hr/mL). (NCT00717067)
Timeframe: Pre-dose, post-dose hours 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48, and 72.

Interventionng*hr/mL (Geometric Mean)
Healthy Subjects: Multiple Dose5341.4
Mild Renal Impairment: Multiple Dose8118.7
Moderate Renal Impairment: Multiple Dose6193.3

[back to top]

Plasma Protein Binding

Percent protein binding (protein unbound maraviroc (MVC) fraction [percent free]) was determined by rapid equilibrium dialysis. Percent free = 100 - percent bound. (NCT00717067)
Timeframe: 2 hours post-dose; normal Day -3 and Day 7; mild moderate: Day 7; severe and ESRD: Day 1

,,,,,
Interventionpercent free (Number)
minimum protein unbound MVC fractionmaximum protein unbound MVC fraction
ESRD18.227.8
Healthy Subjects: Multiple Dose19.726.6
Healthy Subjects: Single Dose14.628.2
Mild Renal Impairment: Multiple Dose15.329.1
Moderate Renal Impairment: Multiple Dose18.131.6
Severe Renal Impairment: Single Dose19.228.1

[back to top]

Compare Late Activated CD4+ T-cell Recovery Rates Between Treatment Regimens From Baseline to Week 48

To compare late (baseline to Week 48) activated CD4+ T-cell recovery rates between treatment regimens. (NCT00752856)
Timeframe: 48 weeks

Interventioncells/mm^3 (Mean)
1 - Kaletra + Isentress Taken Twice Daily-2.24
2 - Atripla Taken Once Daily-5.65

[back to top]

To Compare the Phase 1 Viral Decay Rates Between LPV/r + RAL vs. EFV/TDF/FTC Treatment Combinations.

Repeated HIV RNA measured at different time points (baseline, days 2, 7, 10, 14) will be treated as the outcome variable in a linear mixed-effects model. The primary fixed effects will include time, treatment group, treatment group-by-time interaction; random effects will include both intercept and slope allowing each subject to have individual baseline viral load and viral decay (rate of decrease in viral load following initiation of antiretroviral therapy). The treatment group-by- time interaction term in the model will indicate the difference in viral decay rates between the two treatment groups. Baseline covariate adjustment will be included if necessary. (NCT00752856)
Timeframe: Baseline, days 2, 7, 10, 14

Interventionlog(10)/day (Median)
1 - Kaletra + Isentress Taken Twice Daily0.47
2 - Atripla Taken Once Daily0.55

[back to top]

Viral Suppression Efficacy at 48 Weeks

To determine the antiviral efficacy of LPV/r + RAL compared to EFV/TDF/FTC after 48 weeks of treatment by achieving undetectable viral load (NCT00752856)
Timeframe: 48 weeks

Interventionpercentage of participants (Number)
1 - Kaletra + Isentress Taken Twice Daily86
2 - Atripla Taken Once Daily87.5

[back to top]

Compare Early Activated CD4+ T-cell Recovery Rates From Baseline to Week 4.

To compare early (baseline to Week 4) activated CD4+ T-cell recovery rates between treatment regimens. (NCT00752856)
Timeframe: Baseline to Week 4

Interventioncells/mm^3 (Mean)
1 - Kaletra + Isentress Taken Twice Daily-3.81
2 - Atripla Taken Once Daily-1.18

[back to top]

Change From Baseline in Apolipoprotein B in the LE Set at Week 12 and 48.

Observed Values (NCT00757783)
Timeframe: Baseline, Week 12 and 48

,
Interventiong/L (Mean)
Baseline (n=28,27)Change at Week 12 (n=27,27)Change at Week 48 (n=26,22)
Atazanavir0.8-0.050.0
Darunavir0.7-0.0040.0

[back to top]

Change From Baseline in CD4 Cell Count at Week 12 and 48, Observed Values.

Participants' Cluster of Differentiation (CD) 4 Cell Count were at baseline and the change values at Week 12 and 48 were observed. (NCT00757783)
Timeframe: Baseline, Week 12 and 48

,
Interventioncells/micro L (Mean)
Baseline (n=34,31)Change at Week 12 (n=32,29)Change at Week 48 (n=29,25)
Atazanavir326.768.3205.3
Darunavir268.3111.1217.4

[back to top]

Change From Baseline in Cluster of Differentiation (CD) 4 Cell Count at Week 12 and 48, Last Observation Carried Forward (LOCF).

Participants' Cluster of Differentiation (CD) 4 Cell Count were observed at baseline and the change values at Week 12 and 48 was calculated using LOCF. (NCT00757783)
Timeframe: Baseline, Week 12 and 48

,
Interventioncells/uL (Mean)
Baseline (n=34,31)Change at Week 12 (n=34,31)Change at Week 48 (n=34,31)
Atazanavir326.774.6187.7
Darunavir268.3103.4194.9

[back to top]

Change From Baseline in Cluster of Differentiation (CD) 4 Percent at Week 12 and 48, Last Observation Carried Forward (LOCF).

Participants' Cluster of Differentiation (CD) 4 percent were observed at baseline and the change values at Week 12 and 48 was calculated using LOCF. (NCT00757783)
Timeframe: Baseline, Week 12 and 48

,
Interventionpercentage of CD4 cells (Mean)
Baseline (n=34,31)Change at Week 12 (n=32,30)Change at Week 48 (n=29,25)
Atazanavir21.44.58.5
Darunavir18.65.99.6

[back to top]

Change From Baseline in Fasting Triglyceride (TG) Levels in the Lipid Evaluable (LE) Set at Week12

Observed values. (NCT00757783)
Timeframe: Baseline, Week 12

,
Interventionmilligram per deciliters (mg/dL) (Mean)
Baseline (n=28,27)Change at Week 12 (n=27,27)
Atazanavir114.28.1
Darunavir113.722.0

[back to top]

Change From Baseline in Glucose at Week 12 and 48.

Participants glucose level was analyzed at Baseline and Week 12 and 48. Change from Baseline at Week 12 and 48 was reported. (NCT00757783)
Timeframe: Baseline, Week 12 and 48

,
Interventionmg/dL (Mean)
Baseline (n=33,30)Change at Week 12 (n=30,29)Change at Week 48 (n=28,24)
Atazanavir89.75.86.4
Darunavir88.51.52.8

[back to top]

Change From Baseline in High Density Lipoprotein (HDL) in the LE Set at Week 12 and 48.

Observed Values (NCT00757783)
Timeframe: Baseline, Week 12 and 48

,
Interventionmg/dL (Mean)
Baseline (n=28,27)Change at Week 12 (n=27,27)Change at Week 48 (n=26,22)
Atazanavir45.02.23.7
Darunavir37.96.66.0

[back to top]

Change From Baseline in HIV-1 RNA Viral Load at Week 12 and 48.

the HIV-1 RNA viral load was calculated using Log Base 10 transformed HIV-1 RNA observed values. (NCT00757783)
Timeframe: Baseline, Week 12 and 48

,
InterventionLog10 HIV RNA (Mean)
Baseline (n=34,31)Change at Week 12 (n=32,30)Change at Week 48 (n=29,24)
Atazanavir4.562-2.605-2.902
Darunavir5.016-2.955-3.269

[back to top]

Change From Baseline in Homeostasis Model Assessment-Insulin Resistance (HOMA-IR) at Week 12 and 48.

Participants homeostasis model assessment-insulin resistance (HOMA-IR) were observed and change from Baseline were reported. HOMA-IR score was calculated as: (fasting plasma glucose*fasting serum insulin)/22.5. Low HOMA IR values indicate high insulin sensitivity and high HOMA IR values indicate low insulin sensitivity (insulin resistance). (NCT00757783)
Timeframe: Baseline, Week 12 and 48

,
InterventionHOMA-IR score (Mean)
Baseline (n=27,22)Change at Week 12 (n=20,21)Change at Week 48 (n=19,14)
Atazanavir2.9430.105-1.236
Darunavir1.624-0.4830.035

[back to top]

Change From Baseline in Insulin at Week 12 and 48.

Participants insulin was analyzed at Baseline and Week 12 and 48 and change from Baseline at Week 12 and 48 were reported. (NCT00757783)
Timeframe: Baseline, Week 12 and 48

,
InterventionIU/mL (Mean)
Baseline (n=33,30)Change at Week 12 (n=30,29)Change at Week 48 (n=28,24)
Atazanavir8.590.70-2.88
Darunavir5.96-1.070.95

[back to top]

Change From Baseline in TC/HDL Ratio in the LE Set at Week 12 and 48.

Participants TC and HDL was analyzed at Baseline and Week 12 and 48. Change from Baseline at Week 12 and 48 was calculated as ratio using observed values. (NCT00757783)
Timeframe: Baseline, Week 12 and 48

,
Interventionratio (Mean)
Baseline (n=28,27)Change at Week 12 (n=27,27)Change at Week 48 (n=26,22)
Atazanavir3.9-0.1-0.1
Darunavir4.1-0.10.1

[back to top]

Change From Baseline in Total Cholesterol (TC) Levels in the LE Set at Week 12 and 48

Observed Values (NCT00757783)
Timeframe: Baseline, Week 12 and 48

,
Interventionmg/dL (Mean)
Baseline (n= 28, 27)Change at Week 12 (n= 27, 27)Change at Week 48 (n= 26, 22)
Atazanavir165.14.611.8
Darunavir141.820.322.3

[back to top]

Number of Participants With Antiviral Activity, HIV-1 RNA, Missing Values as Treatment Failure (M=F)

Number of participants with antiviral activity, HIV-1 RNA, missing values as treatment failure (Missing = Failure) were observed. (NCT00757783)
Timeframe: Week 12 and 48

,
Interventionnumber of participants (Number)
Week 12: HIV-1 RNA Less Than (<) 50 copies/mLWeek 12: HIV-1 RNA < 400 copies/mLWeek 48: HIV-1 RNA < 50 copies/mLWeek 48: HIV-1 RNA < 400 copies/mL
Atazanavir19282224
Darunavir13282528

[back to top]

Change From Baseline in Low Density Lipoprotein (LDL) Direct in the LE Set at Week 12 and 48.

Observed Values (NCT00757783)
Timeframe: Baseline, Week 12 and 48

,
Interventionmg/dL (Mean)
Baseline (n=28, 27)Change at Week 12 (n=27, 27)Change at Week 48 (n=26, 22)
Atazanavir100.29.613.9
Darunavir84.613.614.7

[back to top]

Antiviral Activity, Human Immunodeficiency Virus Type 1 (HIV-1) RNA.

Number of Participants with antiviral activity, human immunodeficiency virus Type 1 (HIV-1) RNA less than (<) 50 copies per milliliters (copies/mL) or < 400 copies/mL. (NCT00757783)
Timeframe: Week 12 and 48

,
Interventionnumber of participants (Number)
Week12: HIV-1 RNA Less Than (<) 50 copies/mLWeek 12: HIV-1 RNA < 400 copies/mLWeek 48: HIV-1 RNA < 50 copies/mLWeek 48: HIV-1 RNA < 400 copies/mL
Atazanavir19292224
Darunavir13282528

[back to top]

Change From Baseline in Apolipoprotein A1 in the LE Set at Week 12 and 48.

Observed Values (NCT00757783)
Timeframe: Baseline, Week 12 and 48

,
Interventiongrams per liters (g/L) (Mean)
Baseline (n=28,27)Change at Week 12 (n=27,27)Change at Week 48 (n=26,22)
Atazanavir1.3-0.0070.0
Darunavir1.10.10.1

[back to top]

Lopinavir (Lpv) and Ritonavir (Rtv) Cmax at 4 Weeks

Lpv and rtv Cmax at 4 weeks when participants are receiving study intervention, low dose Kaletra. Time points for data collection: 0, 2hrs, 4hrs, 6hrs, 8hrs (NCT00762320)
Timeframe: 4 weeks

Interventionng/ml (Median)
Lpv Cmax at 4 weeksRtv Cmax at 4 weeks
Low Dose Kaletra11143912.1

[back to top]

Symptoms Across All Patients

Cumulative tally of symptoms for each patients across all visits. Targetted symptoms were asked for at each visit and patients and parents were encouraged to report additional symptoms that were experienced. Each patient got a score for the total number of symptoms at each visit. Scores were totalled, but it the same symptoms occurred continuously it was counted as 1 symptom. (NCT00762320)
Timeframe: Baseline, 1 month, 3 months, 6 months

Interventionnumer of symptoms (Mean)
Symptoms for all subjects at BaselineSymptoms for all subjects at 4 WeeksSymptoms for all subjects at 12 WeeksSymptoms for all subjects at 24 weeks
Low Dose Kaletra20.2021.8026.4026.60

[back to top]

Lopinavir and Ritonavir AUC on Low Dose Tablet

Lopinavir and Ritonavir AUC at 4 weeks when participants are receiving the study intervention, low dose tablet formulation of Kaletra. Data collection points for AUC were 0, 2, 4, 6, and 8 hours post dose. (NCT00762320)
Timeframe: 4 weeks

Interventionhr*ng/ml (Median)
Lpv AUC at 4 weeksRtv AUC at 4 weeks
Low Dose Kaletra856704876.1

[back to top]

Lopinavir (Lpv) and Ritonavir (Rtv) Maximumu Plasma Concentration (CMax) Liquid

Cmax values at baseline (participants are taking liquid Kaletra as part of baseline treatment). Time points for data collection: 0, 2hrs post dose, 4 hrs post dose, 8 hrs post dose. (NCT00762320)
Timeframe: Baseline

Interventionng/ml (Median)
Cmax LpvCmax Rtv
Low Dose Kaletra9742637.0

[back to top]

Lopinavir and Ritonavir Area Under the Curve (AUC) Liquid Kaletra

Area under the curve values for lopinavir at baseline when participants are taking liquid Kaletra as part of their baseline treatment. Time points for data collection: 0, 2 hrs post, 4 hrs post, 6 hrs post, 8 hrs post. (NCT00762320)
Timeframe: Baseline

Interventionhr*ng/ml (Median)
Lpv AUC at baselineRtv AUC at baseline
Low Dose Kaletra906513701.2

[back to top]

Viral Load (VL)

Number of participants who maintained their Viral load undetectable (< 20 copies/ml) for the duration of the study (NCT00762320)
Timeframe: Baseline, Week 4, Week 12 and Week 24

Interventionparticipants (Number)
Low Dose Kaletra8

[back to top]

Patient Satisfaction

Patient Satisfaction Survey. Eight item Likert scale of patient satisfaction with their HIV treatment regimen for patients 7 years of age and older. Items scores are summed to compute a total score. Total scores are reported with a minimum of 0 and a maximum of 32, with higher scores indicating higher satisfaction. (NCT00762320)
Timeframe: Baseline, 1 month

Interventionunits on a scale (Mean)
Low Dose Kaletra Baseline Visit20.2
Low Dose Kaletra Week 4 Visit21.8

[back to top]

Parent Satisfaction

Parent Satisfaction Survey. Eight item Likert scale of parent/guardian satisfaction with the child's HIV treatment regimen. Item scores are summed to compute a total score. Total scores are reported with a minimum of 0 and a maximum of 32, with higher scores indicating higher satisfaction. (NCT00762320)
Timeframe: Baseline, 4 week, 12 weeks and 24 weeks

InterventionScore on a survey (Mean)
Low Dose Kaletra Baseline26.75
Low Dose Kaletra Week 428.38

[back to top]

Lopinavir AUC Ratio of Baseline:Week 4

Ratio of AUC at baseline (liquid)to week 4 (reduced dose tablet). AUC data were collected at 0, 2, 4, 6, and 8 hours post dose. (NCT00762320)
Timeframe: Baseline, week 4

Interventionratio (Mean)
Low Dose Kaletra1.01

[back to top]

Absolute CD4 and CD4 %

Number of participants who had no clinically significant deterioration in absolute CD4 and % CD4 count for the duration of the study. Absolute CD4 and Percent CD4 counts were determined by single or dual platform analysis performed on blood samples by Phoenix Children's Hospital Laboratory, Sonora Quest Laboratory or Labcorp Laboratory. Clinically significant change was determine to be a deterioration in both Absolute CD4 to less than 500 and %CD4 to less than 25%. (NCT00762320)
Timeframe: Baseline, 4 weeks, 12 weeks, 26 weeks

Interventionparticipants (Number)
Low Dose Kaletra8

[back to top]

Number of Participants Who Experienced an Adverse Event

"Safety/tolerability data collected included all adverse events (AEs) reported within the time frame of each regimen evaluated. The intent was to compare adverse events for each sequence and not for each regimen. The regimens for which AE information was culled were:~MVC 300mg BID alone~FPV 1400mg BID alone~FPV 700mg/RTV 100 mg BID alone~FPV 1400mg/RTV 100mg QD alone~FPV 1400mg BID combined with MVC 300mg BID~FPV 700mg/RTV 100 mg BID combined with MVC 300mg BID~FPV 1400mg/RTV 100mg QD combined with MVC 300mg BID The severity of reported AEs was graded according to DAIDS criteria, Version 1.0 (National Institute of Allergy and Infectious Diseases (NIAID). Table for Grading the Severity of Adult and Pediatric Adverse Events, Version 1.0. Division of Acquired Immunodeficiency Syndrome (DAIDS), Washington D.C.; 2004" (NCT00764465)
Timeframe: Day 0 through Day 49

Interventionparticipants (Number)
Group A4
Group B1
Group C3
Group D6
Group E4
Group F7

[back to top]

AUC: Steady-state Plasma MVC PK Following Administration of RTV

Amprenavir (APV) is the active ingredient/ metabolite of Fosamprenavir (FPV). MVC minimum concentration (Cmin), maximum concentration (Cmax), and area under the plasma concentration-time curve (AUC), as determined from MVC concentrations observed in blood samples obtained at baseline, and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours during the period when MVC 300mg BID was administered with the FPV-Containing BID regimens (FPV 1400mg BID, FPV 700mg/RTV 100 mg BID), and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 24 hours during the period when MVC 300mg BID was administered with the FPV QD regimen (FPV 1400mg/RTV 100mg QD). As Groups A and B received the same regimens (albeit in different order), PK data for these two groups were collated, then assessed. For the same reason, PK data from Groups C and D regimens were collated before assessment, as were the PK data from Groups E and F. (NCT00764465)
Timeframe: Day 7 of the MVC 300mg BID regimen and Day 14 of the MVC 300mg/FPV 1400mg BID, MVC 300mg/FPV 700mg/RTV 100mg BID, and MVC 300mg BID Plus FPV 1400mg/RTV 100mg QD regimens

Interventionng•h/mL (Mean)
Group A & B0.87
Group C & D0.34
Group E & F0.98

[back to top]

Cmin/Cmax: Steady-state Plasma Amprenavir (APV) Pharmacokinetics ( PK) Following Admin of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent (Maraviroc) MVC 300mg BID.

Amprenavir (APV) is the active ingredient/metabolite of Fosamprenavir (FPV). (NCT00764465)
Timeframe: Day 14 of the FPV 1400mg BID, FPV 1400mg/MVC 300mg BID, FPV 700mg/RTV 100mg BID, FPV 700mg/RTV 100mg/MVC 300mg BID, FPV 1400mg/RTV 100mg QD, and FPV 1400mg/RTV 100mg QD plus MVC 300mg BID regimens

,,
Interventionng/mL (Mean)
CminCmax
Group A & B0.990.49
Group C & D0.760.69
Group E & F0.680.68

[back to top]

Cmin/Cmax: Steady-state Plasma MVC PK Following Administration of RTV

Amprenavir (APV) is the active ingredient/ metabolite of Fosamprenavir (FPV). MVC minimum concentration (Cmin), maximum concentration (Cmax), and area under the plasma concentration-time curve (AUC), as determined from MVC concentrations observed in blood samples obtained at baseline, and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours during the period when MVC 300mg BID was administered with the FPV-Containing BID regimens (FPV 1400mg BID, FPV 700mg/RTV 100 mg BID), and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 24 hours during the period when MVC 300mg BID was administered with the FPV QD regimen (FPV 1400mg/RTV 100mg QD). As Groups A and B received the same regimens (albeit in different order), PK data for these two groups were collated, then assessed. For the same reason, PK data from Groups C and D regimens were collated before assessment, as were the PK data from Groups E and F. (NCT00764465)
Timeframe: Day 7 of the MVC 300mg BID regimen and Day 14 of the MVC 300mg/FPV 1400mg BID, MVC 300mg/FPV 700mg/RTV 100mg BID, and MVC 300mg BID Plus FPV 1400mg/RTV 100mg QD regimens

,,
Interventionng/mL (Mean)
Cmin (ng/mL)Cmax (ng/mL)
Group A & B0.720.89
Group C & D0.460.30
Group E & F0.770.93

[back to top]

AUC: Steady-state Plasma Amprenavir (APV) Pharmacokinetics ( PK) Following Admin of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent (Maraviroc) MVC 300mg BID.

Amprenavir (APV) is the active ingredient/metabolite of Fosamprenavir (FPV). (NCT00764465)
Timeframe: Day 14 of the FPV 1400mg BID, FPV 1400mg/MVC 300mg BID, FPV 700mg/RTV 100mg BID, FPV 700mg/RTV 100mg/MVC 300mg BID, FPV 1400mg/RTV 100mg QD, and FPV 1400mg/RTV 100mg QD plus MVC 300mg BID regimens

Interventionng•h/mL (Mean)
Group A & B0.56
Group C & D0.74
Group E & F0.79

[back to top]

Atazanavir Trough Plasma Concentration (Cmin) 12 Hours Postdose

(NCT00768989)
Timeframe: At Week 2 from Baseline

Interventionng•h/mL (Geometric Mean)
Atazanavir + Raltegravir687.1

[back to top]

Atazanavir Cmin Prior to the Morning Dose

(NCT00768989)
Timeframe: At Week 2 from Baseline

Interventionng*h / mL (Geometric Mean)
Atazanavir + Raltegravir879.25

[back to top]

Atazanavir Area Under the Concentration Curve From Time 0 to 24 Hours (AUC [0-24h]) in 1 Dosing Interval

AUC (0-24h) was estimated by multiplying AUC (0-12h) by 2. (NCT00768989)
Timeframe: At Week 2 from Baseline

Interventionng•h/mL (Geometric Mean)
Atazanavir + Raltegravir39806.7

[back to top]

Atazanavir Area Under the Concentration Curve From Time 0 to 12 Hours (AUC [0-12h]) in 1 Dosing Interval

(NCT00768989)
Timeframe: At Week 2 from Baseline

Interventionng•h/mL (Geometric Mean)
Atazanavir + Raltegravir19903.4

[back to top]

Raltegravir Cmin Prior to the Morning Dose

(NCT00768989)
Timeframe: At Week 2 from Baseline

Interventionng•h/mL (Geometric Mean)
Atazanavir + Raltegravir445.42

[back to top]

Raltegravir Terminal Elimination Half Life

(NCT00768989)
Timeframe: At Week 2 from Baseline

InterventionHours (Mean)
Atazanavir + Raltegravir2.9

[back to top]

Raltegravir Tmax

Serial blood samples were collected over a 12-hour period after the morning dose at Week 2. (NCT00768989)
Timeframe: At Week 2 from Baseline

InterventionHours (Geometric Mean)
Atazanavir + Raltegravir2.08

[back to top]

Baseline and Mean Change From Baseline in Total Cholesterol Levels

The mean change from baseline in participant fasting lipids was determined using fasting serum samples. (NCT00768989)
Timeframe: From Baseline to Week 24 and Week 48

,
Interventionmg/dL (Mean)
Baseline (n=56, 26)Mean change from Baseline at Week 24 (n=51, 20)Mean change from Baseline at Week 48 (n=38, 20)
Atazanavir + Raltegravir164.614.718.0
Atazanavir + Ritonavir + Tenofovir/Emtricitabine169.615.117.1

[back to top]

Mean Change From Baseline in Absolute Cluster of Differentiation 4 Cell Count

(NCT00768989)
Timeframe: From Baseline to Weeks 2, 4, 8, 12, 16, 20, and 24

,
Interventioncells/mm^3 (Mean)
Mean change from Baseline at Week 2 (n=59, 26)Mean change from Baseline at Week 4 (n=62, 27)Mean change from Baseline at Week 8 (n=60, 29)Mean change from Baseline at Week 12 (n=62, 28)Mean change from Baseline at Week 16 (n=58, 27)Mean change from Baseline at Week 20 (n=58, 24)Mean change from Baseline at Week 24 (n=55, 24)
Atazanavir + Raltegravir81.182.7111.5128.6143.6166.5166.0
Atazanavir + Ritonavir + Tenofovir/Emtricitabine63.1100.1111.9129.3127.6140.7127.0

[back to top]

Mean Change From Baseline in Electrocardiogram Findings

The incidence of QRS wave widening and QT and PR prolongation on participant electrocardiogram findings were evaluated at study Week 24. (NCT00768989)
Timeframe: From Baseline to Week 24

,
Interventionmsec (Mean)
QRS IntervalQTc Friderica IntervalPR Interval
Atazanavir + Raltegravir8.9-2.717.6
Atazanavir + Ritonavir + Tenofovir/Emtricitabine3.66.04.9

[back to top]

Atazanavir Time of Maximum Observed Plasma Concentration (Tmax)

Serial blood samples were collected over a 12-hour period after the morning dose at Week 2. (NCT00768989)
Timeframe: At Week 2 from Baseline

InterventionHours (Geometric Mean)
Atazanavir + Raltegravir3.0

[back to top]

Number of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs), Death as Outcome, AEs Leading to Discontinuation, SAEs Leading to Discontinuation

AE=any new untoward medical occurrence or worsening of a preexisting medical condition that does not necessarily have a causal relationship with this treatment. SAE=any untoward medical occurrence that at any dose results in death, is life-threatening, requires inpatient hospitalization or causes prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect, results in drug dependency or drug abuse, or is an important medical event. (NCT00768989)
Timeframe: Week 1 to Week 96, continuously

,
InterventionParticipants (Number)
AEsSAEsDeathsAEs leading to discontinuationSAEs leading to discontinuation
Atazanavir + Raltegravir607041
Atazanavir + Ritonavir + Tenofovir/Emtricitabine292010

[back to top]

Number of Participants With Blood Chemistry Laboratory Test Results With Worst Toxicity of Grades 1 to 4

Blood urea nitrogen Gr 1:1.25-2.5*ULN;Gr 2:2.6-5.0*ULN; Gr 3:5.1-10*ULN; Gr 4:>10*ULN. Creatinine (mg/dL) Gr 1: 1.1-1.5 *ULN; Gr 2: 1.6-3*ULN: Gr 3: 3.1-6*ULN; Gr 4: >6*ULN. Hypercarbia (meq/L)Gr 1: 33-36; Gr 2:37-40; Gr 3: 41-45; Gr 4:>45. Hypocarbia (meq/L)Gr 1:19-21; Gr 2: 15-18; Gr 3: 10-14; Gr 4:<10. Hypercalcemia (mg/dL)Gr 1:10.6-11.5;Gr 2:11.6-12.5; Gr 3:12.6-13.5;Gr 4: >13.5. Hypocalcemia (mg/dL)Gr 1: 8.4-7.8;Gr 2:7.7-7; Gr 3:6.9-6.1; Gr 4: <6.1.Hyperchloremia(meq/L)Gr 1:113-116; Gr 2:117-120; Gr 3:121-125; Gr 4: >125.Hypochloremia(meq/L)Gr 1: 90-93; Gr 2: 85-89; Gr 3:80-84; Gr 4:<80. (NCT00768989)
Timeframe: While on treatment from Baseline through Week 96

,
InterventionParticipants (Number)
Blood urea nitrogenCreatinineHypercarbiaHypocarbiaHypercalcemiaHypocalcemiaHyperchloremiaHypochloremiaHyperkalemiaHypokalemiaHypernatremiaHyponatremiaHyperclycemiaHypoglycemia
Atazanavir + Raltegravir031152101260386
Atazanavir + Ritonavir + Tenofovir/Emtricitabine12171101050154

[back to top]

Number of Participants With Blood Chemistry Laboratory Test Results With Worst Toxicity of Grades 1 to 4 (Continued)

Hyperkalemia(meq/L) Gr 1: 5.6-6; Gr 2: 6.1-6.5; Gr 3: 6.6-7; Gr4: >7. Hypokalemia(meq/L) Gr 1: 3-3.4; Gr 2: 2.5-2.9; Gr 3: 2-2.4; Gr 4:<2. Hypernatremia (meq/L) Gr 1: 148-150; Gr 2: 151-157; Gr 3: 148-165; Gr 4: >165. Hyponatremia (meq/L) Gr 1: 130-132; Gr 2: 123-129; Gr 3: 116-122; Gr 4: >115.Hyperglycemia(mg/dL)Gr 1: 116-160; Gr 2: 161-250; Gr 3: 251-500; Gr 4: >500. Hypoglycemia(mg/dL)Gr 1: 55-64; Gr 2: 40-54; Gr 3:30-39;Gr 4:<30.Creatine kinase (IU/L) Gr 1: >ULN-1.5*ULN; Gr 2: 1.5-3*ULN; Gr 3: >3-6*ULN; Gr 4: >6.0*ULN. Albumin (g/dL) Gr 1: NCT00768989)
Timeframe: While on treatment from Baseline through Week 96

,
InterventionParticipants (Number)
HyperkalemiaHypokalemiaHypernatremiaHyponatremiaHyperclycemiaHypoglycemiaCreatine kinaseAlbumin
Atazanavir + Raltegravir210386213
Atazanavir + Ritonavir + Tenofovir/Emtricitabine11015472

[back to top]

Number of Participants With Enzyme and Urine Laboratory Test Results With Worst Toxicity of Grades 1 to 4

AST/SGOT=Aspartate aminotransferase/serum glutamate oxaloacetate transaminase; ALT/SGPT=Alanine transaminase/serum glutamic pyruvic transaminase. Bilirubin (mg/dL)Gr 1: 1.1-1.5*ULN;Gr 2:1.6-2.5*ULN;Gr3:2.6-5*ULN;Gr4:>5*ULN.AST/SGOT(U/L)Gr 1:1.25-2.5*ULN;Gr 2: 2.6-5*ULN;Gr 3:5.1-10*ULN;Gr4:>10*ULN.ALT/SGPT (U/L)Gr 1:1.25-2.5*ULN;Gr 2:1.4-2.09*ULN;Gr 3:5.1-10*ULN;Gr4:>10*ULN. Lipase(U/L)Gr 1:1.1-1.39*ULN;Gr 2:>1.5-2*ULN;Gr 3:2.5-5;Gr 4:5*ULN.Proteinuria(g/24 hr loss)Gr 1:1+or <1;Gr 2:2-3+or>1-2; Gr 3:4+or>2-3.5;Gr4:>3.5.Creatine kinase(IU/L)Gr1:2-3*ULN;Gr 2:3.1-5*ULN;Gr 3:5.1-10*ULN;Gr4:>10*ULN. (NCT00768989)
Timeframe: While on treatment from Baseline through Week 96

,
InterventionParticipants (Number)
Total BilirubinAST/SGOTALT/ SGPTLipaseProteinureaCreatine kinase
Atazanavir + Raltegravir621110111421
Atazanavir + Ritonavir + Tenofovir/Emtricitabine288813117

[back to top]

Number of Participants With Hematology Laboratory Test Results With Worst Toxicity of Grades 1 to 4 Among All Treated Participants

ULN=upper limit of normal. Hematocrit(%) Grade (Gr) 1: ≥28.5-<31; Gr 2: ≥24-<28.5; Gr 3: ≥19.5-<24; Gr 4: <19.5. Hemoglobin (g/dL) Gr 1: 9.5-11; Gr 2: 8-9.4; Gr 3: 6.5-7.9; Gr 4: <6.5. Platelets (/mm^3) Gr 1: 75,000-99,000; Gr 2: 50,000-74,999; Gr 3: 20,000-49,999; Gr 4: <20,000. White Blood Cells (/mm^3) Gr 1: >2500-4000; Gr 2: >1000-<2500; Gr 3: >800-<1000; Gr 4: <800. . Prothrombin time (seconds) Gr 1: 1.01-1.25*ULN; Gr 2: 1.26-1.5*ULN; Gr 3: 1.51-3*ULN; Gr 4: >3*ULN. (NCT00768989)
Timeframe: While on treatment from Baseline through Week 96

,
InterventionParticipants (Number)
HematocritHemoglobinPlateletsProthrombin TimeWhite Blood Cells
Atazanavir + Raltegravir1211222
Atazanavir + Ritonavir + Tenofovir/Emtricitabine001714

[back to top]

Atazanavir Terminal Elimination Half Life

(NCT00768989)
Timeframe: At Week 2 from Baseline

InterventionHours (Mean)
Atazanavir + Raltegravir5.0

[back to top]

Number of Participants With HIV RNA Levels <400 Copies/mL at Week 24

NC=F: noncompleter=failure; NC=M: noncompleter=missing; VR-OC: virologic response-observed (NCT00768989)
Timeframe: At Week 24 from Baseline

,
InterventionParticipants (Number)
NC=F (n= 63, 30)NC=M (n=58, 27)VR-OC (n=52, 25)
Atazanavir + Raltegravir525246
Atazanavir + Ritonavir + Tenofovir/Emtricitabine262624

[back to top]

Atazanavir Maximum Observed Plasma Concentration (Cmax) in 1 Dosing Interval

Serial blood samples were collected over a 12-hour period after the morning dose at Week 2. (NCT00768989)
Timeframe: At Week 2 from Baseline

Interventionng/mL (Geometric Mean)
Atazanavir + Raltegravir3506.5

[back to top]

Atazanavir Individual Inhibitory Quotient (IQ)

Individual IQ was defined at Cmin at Week 2 divided by the protein binding adjusted EC90 (ie, the drug concentration observed to inhibit virion production by 90% in a cell-based assay) values for Atazanavir that were derived from individual participant clinical isolates. (NCT00768989)
Timeframe: At Week 2 from Baseline

InterventionUnits on a Scale (Geometric Mean)
Atazanavir + Raltegravir23.47

[back to top]

Number of Nonresponders at Week 8

Participants were classified as nonresponders if they had an HIV RNA level ≥400 copies/mL and a decrease from baseline <2 log10 copies/mL. (NCT00768989)
Timeframe: At Week 8 from Baseline

InterventionParticipants (Number)
Atazanavir + Raltegravir0
Atazanavir + Ritonavir + Tenofovir/Emtricitabine1

[back to top]

Number of Participants With HIV RNA Levels <400 Copies/mL at Week 48

(NCT00768989)
Timeframe: At Week 48 from Baseline

InterventionParticipants (Number)
Atazanavir + Raltegravir45
Atazanavir + Ritonavir + Tenofovir/Emtricitabine25

[back to top]

Number of Participants With HIV RNA Levels <50 Copies/mL at Weeks 48 and 96

Participant HIV RNA level was determined at Weeks 48 and 96 using the Roche Amplicor® Ultrasensitive Assay Version 1. VR-OC=Virologic response-observed cases. (NCT00768989)
Timeframe: At Weeks 48 and 96 from Baseline

InterventionParticipants (Number)
Atazanavir + Raltegravir37
Atazanavir + Ritonavir + Tenofovir/Emtricitabine19

[back to top]

Raltegravir AUC (0-12h) in 1 Dosing Interval

(NCT00768989)
Timeframe: At Week 2 from Baseline

Interventionng•h/mL (Geometric Mean)
Atazanavir + Raltegravir6446.4

[back to top]

Raltegravir Cmax in 1 Dosing Interval

Serial blood samples were collected over a 12-hour period after the morning dose at Week 2. (NCT00768989)
Timeframe: At Week 2 from Baseline

Interventionng/mL (Geometric Mean)
Atazanavir + Raltegravir1577.0

[back to top]

Raltegravir Cmin 12 Hours Postdose

(NCT00768989)
Timeframe: At Week 2 from Baseline

Interventionng•h/mL (Geometric Mean)
Atazanavir + Raltegravir76.2

[back to top]

Mean Change From Baseline in Total Bilirubin Level

(NCT00768989)
Timeframe: From Baseline to Week 24 and Week 48

,
Interventionmg/dL (Mean)
Mean change from Baseline at Week 24Mean change from Baseline at Week 48
Atazanavir + Raltegravir2.152.08
Atazanavir + Ritonavir + Tenofovir/Emtricitabine1.711.52

[back to top]

Number of Participants With Human Immunodeficiency Virus (HIV) Ribonucleic Acid (RNA) Level <50 Copies/mL at Week 24

The number of HIV 1-infected treatment-naive participants with an HIV RNA level <50 copies/mL after 24 weeks of treatment. Confirmed virologic response noncompleter=failure (NC=F); noncompleter=missing (NC=M); virologic response-observed cases (VR-OC). (NCT00768989)
Timeframe: At Week 24 from Baseline

,
InterventionParticipants (Number)
NC=F (n=63, 30)NC=M (n=58, 27)VR-OC (n=52, 25)
Atazanavir + Raltegravir474741
Atazanavir + Ritonavir + Tenofovir/Emtricitabine191919

[back to top]

CD4+ (Cluster of Differentiation 4) T-cell Apoptosis

Change in the percentage of naive CD4 T-cells undergoing apoptosis as measured by propidium iodide staining. This is a lab test that measures the percentage of naive CD4 T-cells that are undergoing cell death. The change in this measure is obtained by determining the difference between the percentage of naive CD4 T-cells undergoing apoptosis at week 24 of treatment and the percentage undergoing apoptosis at baseline. (NCT00775606)
Timeframe: 24 weeks from treatment initiation (baseline and week 24)

Interventionper cent (Mean)
ARM A/Lopinavir-ritonavir-12.33
ARM B/Efavirenz-8.01

[back to top]

CD4+ T-cell Change

This measures the change in CD4+ T-cells from baseline to week 24 of treatment. (NCT00775606)
Timeframe: 24 weeks after treatment initiation (baseline and week 24)

Interventioncells/mm3 (Mean)
ARM A/Lopinavir-ritonavir176.83
ARM B/Efavirenz102.6

[back to top]

Activated and Regulatory CD4+ and CD8+ T-cell Frequencies

Activation of CD4+ and CD8+ T cells were measured at week 24 (NCT00775606)
Timeframe: week 24 measurements

,
Interventionpercentage of cells (Mean)
Activation of CD4+ T cells at week 24Activation of CD8+ T cells at week 24Proliferation of CD4+ T cells at week 24Proliferation of CD8+ T cells at week 24
ARM A Lopinavir-ritonavir8.7020.920.470.81
ARM B Efavirenz7.3917.170.520.48

[back to top]

Activation and Proliferation of CD4+ and CD8+ T-cell Frequencies

Activation and proliferation of CD4+ and CD8+ T cells were measured at baseline (NCT00775606)
Timeframe: baseline measurements

,
Interventionpercentage of cells (Mean)
Activation of CD4+ T cells at baselineActivation of CD8+ T cells at baselineProliferation of CD4+ T cells at baselineProliferation of CD8+ T cells at baseline
ARM A Lopinavir-ritonavir12.8534.611.211.39
ARM B Efavirenz12.3533.571.251.25

[back to top]

Naive, Central Memory, Effector Memory, and T Reg CD4+ T-cell Frequency

Naive, central memory, effector memory, and T reg CD4+ T-cell frequency at baseline (NCT00775606)
Timeframe: baseline measurements

,
Interventionpercentage of cells (Mean)
Mean percentage of naive CD4+ T cells at baselineMean percentage of central memory CD4+ T cells at baselineMean percentage of effector memory CD4+ T cells at baselineMean percentage of CD4+ Treg cells at baseline
ARM A Lopinavir/Ritonavir32.6711.5436.447.35
ARM B Efavirenz24.709.0247.326.96

[back to top]

Naive, Central Memory, Effector Memory, and T Reg CD4+ T-cell Frequency

Naive, central memory and effector memory, and T reg CD4+ T-cell frequency at week 24 (NCT00775606)
Timeframe: week 24 measurements

,
Interventionpercentage of cells (Mean)
Mean percentage of naive CD4+ T cells at week 24Mean percentage of central memory CD4+ T cells at week 24Mean percentage of effector memory CD4+ T cells at week 24Mean percentage of CD4+ Treg cells at week 24
ARM A Lopinavir/Ritonavir29.0810.8934.985.58
ARM B Efavirenz25.738.2844.825.51

[back to top]

CL/F: Fasting Steady-state Plasma Amprenavir (APV) Pharmacokinetics (PK) Following Administration of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent Raltegravir (RTG) 400mg BID.

Amprenavir (APV) is the active ingredient/metabolite of Fosamprenavir (FPV). APV minimum concentration (Cmin), maximum concentration (Cmax), area under the plasma concentration-time curve (AUC), and oral clearance (CL/F) as determined from APV concentrations observed in blood samples obtained at baseline, and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours during the BID regimens (FPV 1400mg BID, FPV 700mg/RTV 100 mg BID), and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 24 hours during the QD regimen (FPV 1400mg/RTV 100mg QD). As Groups A and B received the same regimens (albeit in different order), PK data for these two groups were collated, then assessed. For the same reason, PK data from Groups C and D regimens were collated before assessment, as were the PK data from Groups E and F. (NCT00802074)
Timeframe: Day 14 of the FPV 1400mg BID, FPV 1400mg/RAL 400mg BID, FPV 700mg/RTV 100mg BID, FPV 700mg/RTV 100mg/RAL 400mg BID, FPV 1400mg/RTV 100mg QD, and FPV 1400mg/RTV 100mg QD plus RAL 400mg BID regimens

InterventionL/h (Mean)
Group A & B1.57
Group C & D1.05
Group E & F1.63

[back to top]

Cmin/Cmax: Steady-state Plasma RTG PK Following Admin of RTG 400mg BID Alone and Following Co-administration With FPV 1400mg BID, FPV 700mg/RTV 100 mg BID, or FPV 1400mg/RTV 100mg QD

Amprenavir (APV) is the active ingredient/metabolite of Fosamprenavir (FPV). RAL minimum concentration (Cmin), maximum concentration (Cmax), area under the plasma concentration-time curve (AUC), and oral clearance (CL/F) as determined from RAL concentrations observed in blood samples obtained at baseline, and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours during the period when RAL 400mg BID was administered with the FPV-Containing BID regimens (FPV 1400mg BID, FPV 700mg/RTV 100 mg BID), and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 24 hours during the period when RAL 400mg BID was administered with the FPV QD regimen (FPV 1400mg/RTV 100mg QD). As Groups A and B received the same regimens (albeit in different order), PK data for these two groups were collated, then assessed. For the same reason, PK data from Groups C and D regimens were collated before assessment, as were the PK data from Groups E and F. (NCT00802074)
Timeframe: Day 7 of the RAL 400mg BID regimen and Day 14 of the RAL 400mg/FPV 1400mg BID, RAL 400mg/FPV 700mg/RTV 100mg BID, and RAL 400mg BID Plus FPV 1400mg/RTV 100mg QD regimens

,,
Interventionng/mL (Mean)
Cmin (ng/mL)Cmax (ng/mL)
Group A & B0.620.72
Group C & D0.640.49
Group E & F0.751.06

[back to top]

AUC: Steady-state Plasma RTG PK Following Administration of RTG 400mg BID Alone and Following Co-administration With FPV 1400mg BID, FPV 700mg/RTV 100 mg BID, or FPV 1400mg/RTV 100mg QD

Amprenavir (APV) is the active ingredient/metabolite of Fosamprenavir (FPV). RAL minimum concentration (Cmin), maximum concentration (Cmax), area under the plasma concentration-time curve (AUC), and oral clearance (CL/F) as determined from RAL concentrations observed in blood samples obtained at baseline, and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours during the period when RAL 400mg BID was administered with the FPV-Containing BID regimens (FPV 1400mg BID, FPV 700mg/RTV 100 mg BID), and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 24 hours during the period when RAL 400mg BID was administered with the FPV QD regimen (FPV 1400mg/RTV 100mg QD). As Groups A and B received the same regimens (albeit in different order), PK data for these two groups were collated, then assessed. For the same reason, PK data from Groups C and D regimens were collated before assessment, as were the PK data from Groups E and F. (NCT00802074)
Timeframe: Day 7 of the RAL 400mg BID regimen and Day 14 of the RAL 400mg/FPV 1400mg BID, RAL 400mg/FPV 700mg/RTV 100mg BID, and RAL 400mg BID Plus FPV 1400mg/RTV 100mg QD regimens

Interventionng*h/mL (Mean)
Group A & B0.63
Group C & D0.45
Group E & F0.85

[back to top]

AUC: Fasting Steady-state Plasma Amprenavir (APV) Pharmacokinetics (PK) Following Administration of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent Raltegravir (RTG) 400mg BID.

Amprenavir (APV) is the active ingredient/metabolite of Fosamprenavir (FPV). APV minimum concentration (Cmin), maximum concentration (Cmax), area under the plasma concentration-time curve (AUC), and oral clearance (CL/F) as determined from APV concentrations observed in blood samples obtained at baseline, and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours during the BID regimens (FPV 1400mg BID, FPV 700mg/RTV 100 mg BID), and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 24 hours during the QD regimen (FPV 1400mg/RTV 100mg QD). As Groups A and B received the same regimens (albeit in different order), PK data for these two groups were collated, then assessed. For the same reason, PK data from Groups C and D regimens were collated before assessment, as were the PK data from Groups E and F. (NCT00802074)
Timeframe: Day 14 of the FPV 1400mg BID, FPV 1400mg/RAL 400mg BID, FPV 700mg/RTV 100mg BID, FPV 700mg/RTV 100mg/RAL 400mg BID, FPV 1400mg/RTV 100mg QD, and FPV 1400mg/RTV 100mg QD plus RAL 400mg BID regimens

Interventionng•h/mL (Mean)
Group A & B0.64
Group C & D0.84
Group E & F0.76

[back to top]

Cmin/Cmax: Fasting Steady-state Plasma Amprenavir (APV) Pharmacokinetics (PK) Following Admin of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent Raltegravir (RTG) 400mg BID.

Amprenavir (APV) is the active ingredient/metabolite of Fosamprenavir (FPV). APV minimum concentration (Cmin), maximum concentration (Cmax), area under the plasma concentration-time curve (AUC), and oral clearance (CL/F) as determined from APV concentrations observed in blood samples obtained at baseline, and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours during the BID regimens (FPV 1400mg BID, FPV 700mg/RTV 100 mg BID), and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 24 hours during the QD regimen (FPV 1400mg/RTV 100mg QD). As Groups A and B received the same regimens (albeit in different order), PK data for these two groups were collated, then assessed. For the same reason, PK data from Groups C and D regimens were collated before assessment, as were the PK data from Groups E and F. (NCT00802074)
Timeframe: Day 14 of the FPV 1400mg BID, FPV 1400mg/RAL 400mg BID, FPV 700mg/RTV 100mg BID, FPV 700mg/RTV 100mg/RAL 400mg BID, FPV 1400mg/RTV 100mg QD, and FPV 1400mg/RTV 100mg QD plus RAL 400mg BID regimens

,,
Interventionng/mL (Mean)
Cmin (ng/mL)Cmax (ng/mL)
Group A & B0.570.73
Group C & D0.810.86
Group E & F0.500.82

[back to top]

Number of Participants Who Experienced Adverse Events

"Safety/tolerability data included all adverse events (AEs) reported within the time frame of each regimen evaluated. The intent was to compare AE's for each sequence and not for each regimen. 3The regimens for which AE information was culled were:~RAL 400mg BID alone~FPV 1400mg BID alone~FPV 700mg/RTV 100 mg BID alone~FPV 1400mg/RTV 100mg QD alone~FPV 1400mg BID combined with RAL 400mg BID~FPV 700mg/RTV 100 mg BID combined with RAL 400mg BID~FPV 1400mg/RTV 100mg QD combined with RAL 400mg BID The severity of reported AEs was graded according to DAIDS criteria, Version 1.0 (National Institute of Allergy and Infectious Diseases (NIAID). Table for Grading the Severity of Adult and Pediatric Adverse Events, Version 1.0. Division of Acquired Immunodeficiency Syndrome (DAIDS), Washington D.C.; 2004)." (NCT00802074)
Timeframe: Day 0 through Day 49

Interventionparticipants (Number)
Group A3
Group B3
Group C4
Group D4
Group E5
Group F5

[back to top]

CL/F: Steady-state Plasma RTG PK Following Administration of RTG 400mg BID Alone and Following Co-administration With FPV 1400mg BID, FPV 700mg/RTV 100 mg BID, or FPV 1400mg/RTV 100mg QD

Amprenavir (APV) is the active ingredient/metabolite of Fosamprenavir (FPV). RAL minimum concentration (Cmin), maximum concentration (Cmax), area under the plasma concentration-time curve (AUC), and oral clearance (CL/F) as determined from RAL concentrations observed in blood samples obtained at baseline, and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours during the period when RAL 400mg BID was administered with the FPV-Containing BID regimens (FPV 1400mg BID, FPV 700mg/RTV 100 mg BID), and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 24 hours during the period when RAL 400mg BID was administered with the FPV QD regimen (FPV 1400mg/RTV 100mg QD). As Groups A and B received the same regimens (albeit in different order), PK data for these two groups were collated, then assessed. For the same reason, PK data from Groups C and D regimens were collated before assessment, as were the PK data from Groups E and F. (NCT00802074)
Timeframe: Day 7 of the RAL 400mg BID regimen and Day 14 of the RAL 400mg/FPV 1400mg BID, RAL 400mg/FPV 700mg/RTV 100mg BID, and RAL 400mg BID Plus FPV 1400mg/RTV 100mg QD regimens

InterventionL/h (Mean)
Group A & B1.46
Group C & D2.00
Group E & F1.18

[back to top]

Lopinavir Area Under the Curve (AUC)

Lopinavir Area Under the Plasma Concentration versus Time Curve (AUC) (NCT00810108)
Timeframe: pre-dose, 1,2,4,6,8, and 12 hours post-dose

Interventionmg*hr/L (Median)
All Subjects Taking Whole Tablets144
All Subjects Taking Crushed Tablets92

[back to top]

Change in Waist Circumference From Baseline

Change was calculated as the waist circumference (based on mid-waist circumference) at week (48, 96, and 144) minus the baseline waist circumference. (NCT00811954)
Timeframe: Study entry to weeks 48, 96, and 144

,,
Interventioncm (Mean)
week 48 (nA=555, nB=551, nC=547)week 96 (nA=512, nB=526, nC=517)week 144 (nA=425, nB=419, nC=409)
Arm A: ATV/RTV + FTC/TDF2.33.33.6
Arm B: RAL + FTC/TDF3.14.04.0
Arm C: DRV/RTV + FTC/TDF2.12.83.4

[back to top]

Change in Waist:Height Ratio From Baseline

Change was calculated as the waist:height ratio at week (48, 96, and 144) minus the baseline waist:height ratio. (NCT00811954)
Timeframe: Study entry to weeks 48, 96, and 144

,,
Interventioncm:cm (Mean)
week 48 (nA=555, nB=551, nC=547)week 96 (nA=512, nB=526, nC=517)week 144 (nA=425, nB=419, nC=409)
Arm A: ATV/RTV + FTC/TDF0.010.020.02
Arm B: RAL + FTC/TDF0.020.020.02
Arm C: DRV/RTV + FTC/TDF0.010.020.02

[back to top]

Self-reported Adherence

Self-reported percentage of anti-HIV medications participant had taken during the last month at weeks 4, 24, 48, 96, and 144. (NCT00811954)
Timeframe: At Weeks 4, 24, 48, 96, and 144

,,
Interventionpercentage of prescribed medication (Mean)
week 4 (nA=584, nB=590, nC=583)week 24 (nA=570, nB=568, nC=562)week 48 (nA=555, nB=547, nC=536)week 96 (nA=508, nB=525, nC=507)week 144 (nA=361, nB=376, nC=350)
Arm A: ATV/RTV + FTC/TDF9897969697
Arm B: RAL + FTC/TDF9797979697
Arm C: DRV/RTV + FTC/TDF9896969698

[back to top]

Incidence of Death or AIDS Defining Events (CDC Category C)

The incidence of death or AIDS defining events (CDC category C) was estimated as number of incident events over total person years of follow-up. Multiple new events for a single subject were counted toward events totals in estimation of event incidence; generalized estimating equations were used to estimation of robust standard errors for the incidence. (NCT00811954)
Timeframe: Study entry to off-study at any time throughout the study (up to 213 weeks), participant follow-up time was variable

Interventionevents per 100 person-years (Number)
Arm A: ATV/RTV + FTC/TDF1.55
Arm B: RAL + FTC/TDF1.64
Arm C: DRV/RTV + FTC/TDF2.14

[back to top]

Incidence of Targeted Serious Non-AIDS Defining Events (Renal Failure, Liver Disease, Serious Metabolic Disorder, and CVD)

The incidence of targeted serious non-AIDS defining events was estimated as number of incident events over total person years of follow-up. Multiple new events for a single subject were counted toward events totals in estimation of event incidence; generalized estimating equations were used to estimation of robust standard errors for the incidence. (NCT00811954)
Timeframe: Study entry to off-study at any time throughout the study (up to 213 weeks), participant follow-up time was variable

Interventionevents per 100 person-years (Number)
Arm A: ATV/RTV + FTC/TDF2.38
Arm B: RAL + FTC/TDF2.24
Arm C: DRV/RTV + FTC/TDF2.69

[back to top]

Presence of Mutations Associated With ATV/RTV or DRV/RTV Resistance

The number of participants with ATV/RTV or DRV/RTV resistance determined by the Stanford resistance scoring algorithm (Version 6.3). All sequencing was performed regardless of status on randomized treatment at the time of virologic failure; no sequencing was performed on subjects not meeting virologic failure. (NCT00811954)
Timeframe: At the virologic failure at any time throughout the study (up to 213 weeks)

Interventionparticipants (Number)
Arm A: ATV/RTV + FTC/TDF0
Arm B: RAL + FTC/TDF0
Arm C: DRV/RTV + FTC/TDF0

[back to top]

Presence of Mutations Associated With INI Resistance

The number of participants with INI resistance determined by the Stanford resistance scoring algorithm (Version 6.3). All sequencing was performed regardless of status on randomized treatment at the time of virologic failure; no sequencing was performed on subjects not meeting virologic failure. (NCT00811954)
Timeframe: At the virologic failure at any time throughout the study (up to 213 weeks)

Interventionparticipants (Number)
Arm A: ATV/RTV + FTC/TDF1
Arm B: RAL + FTC/TDF1
Arm C: DRV/RTV + FTC/TDF1

[back to top]

Presence of Mutations Associated With NRTI Resistance

The number of participants with NRTI resistance determined by the Stanford resistance scoring algorithm (Version 6.3). All sequencing was performed regardless of status on randomized treatment at the time of virologic failure; no sequencing was performed on subjects not meeting virologic failure. (NCT00811954)
Timeframe: At the virologic failure at any time throughout the study (up to 213 weeks)

Interventionparticipants (Number)
Arm A: ATV/RTV + FTC/TDF8
Arm B: RAL + FTC/TDF7
Arm C: DRV/RTV + FTC/TDF3

[back to top]

Change in Framingham 10-year Risk of MI or Coronary Death From Baseline

"Only risk score estimated with fasting lipid results were included. Change was calculated as the Framingham 10-year risk of MI or coronary death at week (48, 96, and 144) minus the baseline Framingham 10-year risk of MI or coronary death. Framingham 10-year risk of MI or coronary death was calculated using Hear Coronary Heart Disease (10-year risk) found at https://www.framinghamheartstudy.org/risk-functions/coronary-heart-disease/hard-10-year-risk.php.~Framingham 10-year risk of MI or coronary death was calculated according to age, laboratory values of total cholesterol and HDL cholesterol, smoking status, systolic blood pressure, and treatment for hypertension. The Framingham 10-year risk of MI or coronary death was calculated as: for males: <0 point (<1 percent risk) up to ≥17 points (≥30 percent risk); whereas for females: <9 points (<1 percent risk) up to ≥25 points (≥30 percent risk). Higher scores indicate high cardiovascular risk." (NCT00811954)
Timeframe: Study entry to weeks 48, 96, and 144

,,
Interventionpercent risk (Mean)
week 48 (nA=509, nB=537, nC=492)week 96 (nA=479, nB=493, nC=470)week 144 (nA=347, nB=383, nC=349)
Arm A: ATV/RTV + FTC/TDF0.40.50.6
Arm B: RAL + FTC/TDF0.00.20.4
Arm C: DRV/RTV + FTC/TDF0.40.40.9

[back to top]

CD4+ T-cell Count

The absolute levels of CD4+ T-cell counts (cells/mm3) (NCT00811954)
Timeframe: At Weeks 24, 48, 96, and 144

,,
Interventioncells/mm^3 (Mean)
week 24 (nA=582, nB=574, nC=579)week 48 (nA=564, nB=565, nC=559)week 96 (nA=523, nB=541, nC=525)week 144 (nA=395, nB=418, nC=394)
Arm A: ATV/RTV + FTC/TDF462524587622
Arm B: RAL + FTC/TDF460526596631
Arm C: DRV/RTV + FTC/TDF457509564596

[back to top]

CD4+ T-cell Count Changes From Baseline

Change was calculated as the CD4+ T-cell count at week (24, 48, 96, and 144) minus the baseline CD4+ T-cell count (NCT00811954)
Timeframe: Study entry to weeks 24, 48, 96, and 144

,,
Interventioncells/mm^3 (Mean)
week 24 (nA=582, nB=574, nC=579)week 48 (nA=564, nB=565, nC=559)week 96 (nA=523, nB=541, nC=525)week 144 (nA=395, nB=418, nC=394)
Arm A: ATV/RTV + FTC/TDF157218284324
Arm B: RAL + FTC/TDF153218288325
Arm C: DRV/RTV + FTC/TDF147201256288

[back to top]

Change in Fasting HDL Cholesterol Level From Baseline

Only fasting results are included. Change was calculated as the fasting HDL cholesterol at week (48, 96, and 144) minus the baseline fasting HDL cholesterol. (NCT00811954)
Timeframe: Study entry to weeks 48, 96, and 144

,,
Interventionmg/dL (Mean)
week 48 (nA=522, nB=542, nC=506)week 96 (nA=490, nB=505, nC=488)week 144 (nA=364, nB=397, nC=363)
Arm A: ATV/RTV + FTC/TDF678
Arm B: RAL + FTC/TDF566
Arm C: DRV/RTV + FTC/TDF557

[back to top]

Change in Fasting Plasma Glucose Level From Baseline

Only fasting results are included. Change was calculated as the fasting plasma glucose at week (48, 96, and 144) minus the baseline fasting plasma glucose. (NCT00811954)
Timeframe: Study entry to weeks 48, 96, and 144

,,
Interventionmg/dL (Mean)
week 48 (nA=517, nB=535, nC=506)week 96 (nA=489, nB=499, nC=481)week 144 (nA=353, nB=392, nC=358)
Arm A: ATV/RTV + FTC/TDF2.23.02.2
Arm B: RAL + FTC/TDF1.30.90.9
Arm C: DRV/RTV + FTC/TDF2.12.53.6

[back to top]

Change in Fasting Total Cholesterol Level From Baseline

Only fasting results are included. Change was calculated as the fasting total cholesterol at week (48, 96, and 144) minus the baseline fasting total cholesterol. (NCT00811954)
Timeframe: Study entry to weeks 48, 96, and 144

,,
Interventionmg/dL (Mean)
week 48 (nA=521, nB=542, nC=507)week 96 (nA=490, nB=505, nC=490)week 144 (nA=364, nB=397, nC=363)
Arm A: ATV/RTV + FTC/TDF131620
Arm B: RAL + FTC/TDF136
Arm C: DRV/RTV + FTC/TDF151419

[back to top]

Change in Fasting Triglycerides Level From Baseline

Only fasting results are included. Change was calculated as the fasting triglycerides at week (48, 96, and 144) minus the baseline fasting triglycerides. (NCT00811954)
Timeframe: Study entry to weeks 48, 96, and 144

,,
Interventionmg/dL (Mean)
week 48 (nA=522, nB=542, nC=507)week 96 (nA=490, nB=505, nC=490)week 144 (nA=364, nB=397, nC=363)
Arm A: ATV/RTV + FTC/TDF181912
Arm B: RAL + FTC/TDF-9-9-4
Arm C: DRV/RTV + FTC/TDF161620

[back to top]

Cumulative Incidence of Discontinuation of the RAL or PI Component of Randomized Treatment for Toxicity by Week 96

The cumulative incidence of discontinuation for toxicity by week 96 was estimated using competing risks with treatment discontinuation for other reasons considered as a competing event; participants completing the study on the RAL or PI component of their randomized regimen were considered censored at the earliest of the date of last patient contact and off study date. (NCT00811954)
Timeframe: From study entry to week 96

Interventioncumulative events per 100 persons (Number)
Arm A: ATV/RTV + FTC/TDF14
Arm B: RAL + FTC/TDF1
Arm C: DRV/RTV + FTC/TDF5

[back to top]

Cumulative Incidence of First Adverse Event by Week 96

"The cumulative incidence of first adverse event (with and without total bilirubin and creatine kinase and measured from study entry) by week 96 was estimated using methods for competing risks. Discontinuation of randomized treatment prior to an adverse event was considered a competing event.~The time to the first of any post-entry Grade 2, 3, or 4 sign or symptom, or Grade 3 or 4 laboratory abnormality while on randomization. The protocol required reporting of signs and symptoms and laboratory values as follow: all signs and symptoms grade ≥2 post-entry to week 48, signs and symptoms grade >3 after week 48, and laboratory values grade >3 and all signs, symptoms, and laboratory values that led to a change in treatment, regardless of grade throughout out all post-entry follow-up." (NCT00811954)
Timeframe: From study entry to week 96

Interventioncumulative events per 100 persons (Number)
Arm A: ATV/RTV + FTC/TDF81
Arm B: RAL + FTC/TDF59
Arm C: DRV/RTV + FTC/TDF65

[back to top]

Cumulative Probability of First Virologic Failure by Week 96

"The Kaplan-Meier estimate of the cumulative probability of virologic failure by week 96.~Time to virologic failure was defined as the first time from study entry to the first of two consecutive HIV-1 RNA >1000 copies/mL at or after week 16 and before week 24, or >200 copies/mL at or after week 24. Week 16 is defined to occur between 14 (98 days) and 18 weeks (126 days) after study entry, week 24 is defined to occur between 22 (154 days) and 26 (182 days) after study entry, and week 96 is defined to occur between 88 (616 days) and 104 (728 days) after study entry." (NCT00811954)
Timeframe: From study entry to week 96

Interventioncumulative probability per 100 persons (Number)
Arm A: ATV/RTV + FTC/TDF13
Arm B: RAL + FTC/TDF10
Arm C: DRV/RTV + FTC/TDF15

[back to top]

Cumulative Probability of Time to Loss of Virologic Response (TLOVR) by Week 96

"The Kaplan-Meier estimate of the cumulative probability of TROVR by week 96.~A composite TLOVR endpoint defined in the CDER of the FDA document Guidance for Industry - Antiretroviral Drugs Using Plasma HIV RNA Measurements - Clinical Consideration for Accelerated and Traditional Approval (Appendix B, pages 20) http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm070968.pdf.~If participants never achieved a confirmed HIV-1 RNA≤200 cp/mL (on two consecutive visits) prior to death, permanent discontinuation of randomized treatment, or time of last available HIV-1 RNA evaluation, TLOVR was equal to 0; otherwise, TLOVR was the earliest time of permanent discontinuation of randomized treatment prior to study close-out period, time to confirmed levels >200 cp/mL, or time to death. If TLOVR is immediately preceded by a single missing scheduled visit or multiple consecutive missing scheduled visits, TLOVR is replaced by the first such missing visit." (NCT00811954)
Timeframe: From study entry to week 96

Interventioncumulative probability per 100 persons (Number)
Arm A: ATV/RTV + FTC/TDF31
Arm B: RAL + FTC/TDF16
Arm C: DRV/RTV + FTC/TDF24

[back to top]

Time to Loss of Virological Response (TLOVR)

TLOVR (virological failure) was defined as the time from first dose of study treatment (Day 1) until the time of virologic failure using the time to loss of virologic response algorithm. (NCT00827112)
Timeframe: Baseline through Week 96

InterventionDays (Mean)
Maraviroc+ Atazanavir / Ritonavir436.2
Atazanavir / Ritonavir + Emtricitabine / Tenofovir463.8

[back to top]

Change From Baseline in Plasma log10 Viral Load at Weeks 16, 24, 48 and 96

(NCT00827112)
Timeframe: Baseline, Week 16, Week 24, Week 48, Week 96

,
Interventionlog10 copies/ml (Mean)
Baseline (n= 59, 61)Change at Week 16 (n= 54, 58)Change at Week 24 (n= 56, 58)Change at Week 48 (n= 53, 54)Change at Week 96 (n= 49, 51)
Atazanavir / Ritonavir + Emtricitabine / Tenofovir114827-107684.6-110498.1-115582.9-99662.6
Maraviroc+ Atazanavir / Ritonavir84982-89859.1-87241.2-82343.4-80117.7

[back to top]

Number of Participants With HIV-1 RNA Tropism Status Using Trofile Assay

Viral tropism was determined using the trofile assay with enhanced sensitivity for participants with HIV-1 RNA greater than equal to 1000 copies/mL. The enhanced trofile assay had the sensitivity to detect 100 percent of spiked samples when C-X-C chemokine receptor type 4 {CXCR4} [X4]-using HIV-1 RNA represented 0.3 percent of the total viral population. (NCT00827112)
Timeframe: Baseline to Week 96 or Time of treatment Failure

,
InterventionParticipants (Number)
BaselineWeek 96 or Time of treatment Failure
Atazanavir / Ritonavir + Emtricitabine / Tenofovir610
Maraviroc+ Atazanavir / Ritonavir600

[back to top]

Percentage of Participants With Less Than 400 Copies/mL of HIV-1 RNA

(NCT00827112)
Timeframe: Week 2, Week 4, Week 8, Week 12, Week 16, Week 20, Week 24, Week 32, Week 40, Week 48, Week 60, Week 72, Week 84, Week 96

,
InterventionPercentage of participants (Number)
Week 2 (n= 55, 60)Week 4 (n= 57, 60)Week 8 (n= 57, 59)Week 12 (n= 55, 59)Week 16 (n= 54, 58)Week 20 (n= 56, 57)Week 24 (n= 56, 58)Week 32 (n= 55, 57)Week 40 (n= 54, 55)Week 48 (n= 53, 54)Week 60 (n= 52, 53)Week 72 (n= 52, 53)Week 84 (n= 50, 52)Week 96 (n= 49, 51)
Atazanavir / Ritonavir + Emtricitabine / Tenofovir34.4352.4677.0588.5291.8093.4493.4493.4490.1686.8986.8985.2585.2583.61
Maraviroc+ Atazanavir / Ritonavir27.1250.8579.6689.8388.1489.8391.5389.8391.5389.8386.4486.4481.3677.97

[back to top]

Percentage of Participants With Less Than 50 Copies/mL of HIV-1 RNA

(NCT00827112)
Timeframe: Week 2, Week 4, Week 8, Week 12, Week 16, Week 20, Week 24, Week 32, Week 40, Week 48, Week 60, Week 72, Week 84, Week 96

,
InterventionPercentage of participants (Number)
Week 2 (n= 55, 60)Week 4 (n= 57, 60)Week 8 (n= 57, 59)Week 12 (n= 55, 59)Week 16 (n= 54, 58)Week 20 (n= 56, 57)Week 24 (n= 56, 58)Week 32 (n= 55, 57)Week 40 (n= 54, 55)Week 48 (n= 53, 54)Week 60 (n= 52, 53)Week 72 (n= 52, 53)Week 84 (n= 50, 52)Week 96 (n= 49, 51)
Atazanavir / Ritonavir + Emtricitabine / Tenofovir6.6021.3042.6062.3073.8083.6188.5288.5288.5283.6185.2581.9783.6181.97
Maraviroc+ Atazanavir / Ritonavir08.5047.5061.0072.9071.2081.3679.6681.3674.5867.8074.5876.2767.80

[back to top]

Number of Participants With Phenotypic Resistance

Phenotypic resistance was assessed for all participants at screening and was evaluated for PIs, NRTIs, and NNRTIs using Monogram GenoSeq and/or PhenoSenseGT assays. This was then repeated for all participants with HIV-1 viral load more than 500 copies/mL either at treatment failure or at early termination, up to Week 96. (NCT00827112)
Timeframe: Week 96 or Time of treatment failure

InterventionParticipants (Number)
Maraviroc+ Atazanavir / Ritonavir0
Atazanavir / Ritonavir + Emtricitabine / Tenofovir0

[back to top]

Time-Averaged Difference (TAD) in log10 Viral Load

TAD was calculated as area under the curve of HIV divided by time period minus baseline HIV where HIV was denoted as HIV-1 RNA (log10 copies/mL). (NCT00827112)
Timeframe: Week 16, Week 24, Week 48, Week 96

,
Interventionlog10 copies/mL (Mean)
Week 16Week 24Week 48Week 96
Atazanavir / Ritonavir + Emtricitabine / Tenofovir-2.402-2.626-2.868-3.001
Maraviroc+ Atazanavir / Ritonavir-2.459-2.663-2.897-2.998

[back to top]

Average Observed Plasma Concentration (Cavg) of Maraviroc

Cavg was described as area under the plasma concentration-time profile from time zero to time 24 hours (AUC24) divided by the dosing interval (AUC24/ 24). (NCT00827112)
Timeframe: Day 14 (0, 0.5, 1, 2, 3, 4, 6, 8, 12 and 24 hours post dose)

Interventionng/mL (Mean)
Maraviroc+ Atazanavir / Ritonavir185.10

[back to top]

HIV-1 RNA Levels at Baseline

(NCT00827112)
Timeframe: Baseline

Interventioncopies/mL (Mean)
Maraviroc+ Atazanavir / Ritonavir84982
Atazanavir / Ritonavir + Emtricitabine / Tenofovir114827

[back to top]

Number of Participants With Genotypic Resistance

Genotypic resistance was assessed for all participants at screening and was evaluated for protease inhibitors (PIs), Nucleotide reverse transcriptase inhibitors (NRTIs), and non-NRTIs (NNRTIs) using Monogram GenoSeq and/or PhenoSenseGT assays. This was then repeated for all participants with HIV-1 viral load more than 500 copies/mL either at treatment failure or at early termination, up to Week 96. (NCT00827112)
Timeframe: Week 96 or Time of treatment failure

InterventionParticipants (Number)
Maraviroc+ Atazanavir / Ritonavir0
Atazanavir / Ritonavir + Emtricitabine / Tenofovir0

[back to top]

Maximum Observed Plasma Concentration (Cmax) of Maraviroc

(NCT00827112)
Timeframe: Day 14 (0, 0.5, 1, 2, 3, 4, 6, 8, 12 and 24 hours post dose)

Interventionnanogram (ng)/mL (Median)
Maraviroc+ Atazanavir / Ritonavir650

[back to top]

Percentage of Participants With Plasma Human Immuno Deficiency Virus-1 Ribonucleic Acid (HIV-1 RNA) Levels Less Than 50 Copies/Milliliter (mL)

(NCT00827112)
Timeframe: Week 48

InterventionPercentage of participants (Number)
Maraviroc+ Atazanavir / Ritonavir74.60
Atazanavir / Ritonavir + Emtricitabine / Tenofovir83.60

[back to top]

Minimum Observed Plasma Concentration (Cmin) of Maraviroc

(NCT00827112)
Timeframe: Day 14 (0, 0.5, 1, 2, 3, 4, 6, 8, 12 and 24 hours post dose)

Interventionng/mL (Median)
Maraviroc+ Atazanavir / Ritonavir37.0

[back to top]

Change From Baseline in Cluster of Differentiation 4+T Lymphocyte (CD4) Cell Counts at Weeks 16, 24, 48 and 96

(NCT00827112)
Timeframe: Baseline, Week 16, Week 24, Week 48, Week 96

,
Interventioncells/microliter (cells/mcL) (Mean)
Baseline (n= 59, 61)Change at Week 16 (n= 54, 58)Change at Week 24 (n= 54, 57)Change at Week 48 (n= 52, 53)Change at Week 96 (n= 50, 51)
Atazanavir / Ritonavir + Emtricitabine / Tenofovir390.00139.80173.30226.60298.50
Maraviroc+ Atazanavir / Ritonavir357.70169.60188.90215.70287.50

[back to top]

Change From Baseline in Cluster of Differentiation 8+T Lymphocyte (CD8) Cell Count at Weeks 16, 24, 48 and 96

(NCT00827112)
Timeframe: Baseline, Week 16, Week 24, Week 48, Week 96

,
Interventioncells/mcL (Mean)
Baseline (n= 59, 61)Change at Week 16 (n= 54, 58)Change at Week 24 (n= 54, 57)Change at Week 48 (n= 52, 53)Change at Week 96 (n= 50, 51)
Atazanavir / Ritonavir + Emtricitabine / Tenofovir1125.60-153.80-178.00-267.60-231.40
Maraviroc+ Atazanavir / Ritonavir931.1063.706.20-76.80-63.00

[back to top]

Change From Baseline in HIV-1 RNA Levels of First 15 Participants at Days 4, 7, 10 and 14

Plasma HIV-1 RNA levels were evaluated for first 15 participants enrolled at United States (U.S) sites only. (NCT00827112)
Timeframe: Baseline , Days 4, 7, 10 and 14

,
Interventioncopies/mL (Mean)
Change at Day 4Change at Day 7Change at Day 10Change at Day 14
Atazanavir / Ritonavir + Emtricitabine / Tenofovir-46479.40-52137.10-54925.90-55449.90
Maraviroc+ Atazanavir / Ritonavir1800.00-36947.90-58595.80-47271.60

[back to top]

Proportion of Participants With Virologic Failure or Off Study Treatment Regimen or Death at or Prior to Week 24

The proportion of participants with virologic failure (see primary outcome measure for definition) and/or premature treatment discontinuation/modification and/or death was estimated using Kaplan-Meier method. An adaptation of Greenwood's variance estimate was used in constructing the confidence interval. (NCT00830804)
Timeframe: From start of study treatment to Week 24

InterventionProportion of participants (Number)
RAL+DRV/RTV0.21

[back to top]

Changes in Fasting Total Cholesterol, High-density Lipoprotein and Triglyceride at Week 24

Results report the week 24 change from week 0 (week 24 - week 0) fasting total cholesterol, high-density lipoprotein and triglyceride. (NCT00830804)
Timeframe: From start of study treatment through week 24

Interventionmg/dL (Median)
Fasting Total CholesterolFasting High-density LipoproteinFasting Triglyceride
RAL + DRV/RTV31.56.524.5

[back to top]

Changes in Fasting Total Cholesterol, High-density Lipoprotein and Triglyceride at Week 48

Results report the week 48 change from week 0 (week 48 - week 0) fasting total cholesterol, high-density lipoprotein and triglyceride. (NCT00830804)
Timeframe: From start of study treatment through week 48

Interventionmg/dL (Median)
Fasting Total CholesterolFasting High-density LipoproteinFasting Triglyceride
RAL + DRV/RTV30923

[back to top]

Number of Participants With Pretreatment Drug Resistance

Results report the number of participants who had resistance to non-nucleoside reverse transciptase inhibitors (NNRTI), nucleoside reverse transciptase inhibitors (NRTI) and protease inbitors (PI) based on genotypic resistance testing done prior to participant's entry into the study. Participants are classified into one (and only one category) based on the maximum number of drug class resistance seen for the participant. (NCT00830804)
Timeframe: At screening

Interventionparticipants (Number)
With NNRTI mutations onlyWith NRTI mutations onlyWith Both NNRTI and NRTI mutationsWith PI mutations onlyWith PI, NNRTI and NRTI mutationsNo Resistance Detected
RAL + DRV/RTV9812191

[back to top]

Proportion of Participants With Plasma HIV-1 RNA <50 Copies/ml or <200 Copies/ml at Week 48

Results report the percentage of participants with plasma HIV-1 RNA <50 copies/ml or <200 copies/ml at week 48. (NCT00830804)
Timeframe: From start of study treatment to week 48

Interventionproportion of participants (Number)
With HIV-1 RNA < 50 copies/mlWith HIV-1 RNA < 200 copies/ml
RAL+DRV/RTV0.710.86

[back to top]

Proportion of Participants With Plasma HIV-1 RNA < 50 Copies/ml or <200 Copies/ml at Week 24

Results report the percentage of participants with plasma HIV-1 RNA < 50 copies/ml or <200 copies/ml at week 24. (NCT00830804)
Timeframe: From start of study treatment to week 24

Interventionproportion of participants (Number)
With HIV-1 RNA < 50 copies/mlWith HIV-1 RNA < 200 copies/ml
RAL+DRV/RTV0.790.93

[back to top]

Change in CD4 Count at Week 48

Results report the week 48 change from baseline (week 48 - baseline) in CD4 count. Baseline CD4 count was computed as the mean of CD4 count values at pre-entry and study entry. (NCT00830804)
Timeframe: From start of study treatment through week 48

Interventioncells/mm3 (Median)
RAL + DRV/RTV200

[back to top]

Change in Fasting Low-density Lipoprotein at Week 24

Results report the week 24 change from week 0 (week 24 - week 0) fasting low-density lipoprotein (LDL). For participants whose calculated fasting LDL and direct fasting LDL were both reported, only the calculated fasting LDL was used. Direct fasting LDL was reported when the participant had high fasting triglyceride. (NCT00830804)
Timeframe: From start of study treatment through week 24

Interventionmg/dL (Median)
RAL + DRV/RTV16.0

[back to top]

Change in Fasting Low-density Lipoprotein at Week 48

Results report the week 48 change from week 0 (week 48 - week 0) fasting low-density lipoprotein (LDL). For participants whose calculated fasting LDL and direct fasting LDL were both reported, only the calculated fasting LDL was used. Direct fasting LDL was reported when the participant had high fasting triglyceride. (NCT00830804)
Timeframe: From start of study treatment through week 48

Interventionmg/dL (Median)
RAL + DRV/RTV17

[back to top]

Change in Plasma HIV-1 RNA From Baseline to Week 1

Results report the week 1 change from baseline (week 1 - baseline) in HIV-1 RNA. Baseline HIV-1 RNA was computed as the mean of the log10 HIV-1 RNA values at pre-entry and study entry. (NCT00830804)
Timeframe: Baseline and week 1

Interventionlog10 copies/ml (Median)
RAL+DRV/RTV-1.67

[back to top]

Number of Participants With Integrase Drug Resistance at Virologic Failure

Results report the number of participants who had integrase resistance mutation(s) detected at the time of virologic failure. (NCT00830804)
Timeframe: From 12 weeks after starting study treatment to week 52

Interventionparticipants (Number)
RAL+DRV/RTV5

[back to top]

Number of Participants With Perfect Overall Adherence by Self Report

"At each study visit, adherence was measured in terms of the number of missed doses each participant had over a 4-day recall for each drug. Adherence for all study visit weeks were combined for an overall measure of adherence. Participants who had zero missed doses on all weeks in all drugs while on study were classified as having an overall perfect adherence." (NCT00830804)
Timeframe: From one week after starting study treatment to week 52

Interventionparticipants (Number)
RAL + DRV/RTV95

[back to top]

Plasma Trough Concentration of Darunavir

Plasma trough concentrations (ng/ml) of Darunavir (DRV) below the detection limit (50 ng/ml) were replaced by half the corresponding lower limit of quantitation. Geometric mean of trough concentrations obtained within the prescribed trough time (within 20-28 hours after the last DRV dose) was computed for each participant. For participants who experienced virologic failure (see primary outcome measure definition), only those concentrations on or before virologic failure confirmation were used in the geometric mean computation. (NCT00830804)
Timeframe: From start of study treatment to week 52

Interventionng/ml (Median)
RAL+DRV/RTV1218

[back to top]

Plasma Trough Concentration of Raltegravir

Plasma trough concentrations (ng/ml) of Raltegravir (RAL) below the detection limit (10 ng/ml) were replaced by half the corresponding lower limit of quantitation. Geometric mean of trough concentrations obtained within the prescribed trough time (within 9-15 hours after the last RAL dose) was computed for each participant. For participants who experienced virologic failure (see primary outcome measure definition), only those concentrations on or before virologic failure confirmation were used in the geometric mean computation. (NCT00830804)
Timeframe: From start of study treatment to week 52

Interventionng/ml (Median)
RAL+DRV/RTV117

[back to top]

Proportion of Participants Who Experienced Signs/Symptoms or Laboratory Toxicities Grade 3 or Higher, or of Any Grade Which Led to a Permanent Change or Discontinuation of Study Treatment

Signs, symptoms and laboratory values were graded according to the Division of AIDS Adverse Event Grading System. Results report the percentage of participants who had grade 3 or higher events, or events of any grade which led to a permanent change or discontinuation of study treatment, which occurred any time from start of treatment to end of treatment. (NCT00830804)
Timeframe: From start of study treatment to week 52

Interventionproportion of participants (Number)
RAL+DRV/RTV0.20

[back to top]

Proportion of Participants With Virologic Failure After Initiating RAL Plus DRV/RTV at or Prior to Week 24

Virologic failure is defined as: at week 12, confirmed plasma HIV-1 RNA >= 1000 copies/ml or confirmed rebound from the week 4 value by >0.5 log10 copies/ml (for subjects with week 4 value <= 50 copies/ml, confirmed rebound to >50 copies/ml); at week 24 or later, confirmed value > 50 copies/ml. Viral load confirmation was scheduled 7-35 days after initial virologic failure. The proportion was estimated using Kaplan-Meier method. An adaptation of Greenwood's variance estimate was used in constructing the confidence interval. (NCT00830804)
Timeframe: From start of study treatment to week 24

InterventionProportion of participants (Number)
RAL+DRV/RTV0.16

[back to top]

Number of Participants With Protease Drug Resistance at Virologic Failure

Results report the number of participants who had protease resistance mutation(s) detected at the time of virologic failure. (NCT00830804)
Timeframe: From 12 weeks after starting study treatment to week 52

Interventionparticipants (Number)
RAL+DRV/RTV0

[back to top]

AUC(TAU) of Ritonavir, Administered As Atazanavir/Ritonavir With and Without Voriconazole, in EM Participants

AUC(TAU)=area under the plasma concentration-time curve in 1 dosing interval; EM=extensive metabolizers, or participants with functional CYP2C19 alleles. (NCT00833482)
Timeframe: Predose and at 1, 2, 3, 4, 5, 7, 9,13, and 24 hours postdose on Days 20 and 30 of a 30-day cycle

Interventionng.h/mL (Geometric Mean)
Atazanavir/Ritonavir, 300/100 QD (EM)9572
Atazanavir/Ritonavir, 300/100mgQD + Voriconazole, 200mgBID(EM)8280

[back to top]

Area Under the Plasma Concentration-time Curve in 1 Dosing Interval [AUC(TAU)] of Atazanavir Administered as Atazanavir/Ritonavir With and Without Voriconazole, in EM Participants

EM=extensive metabolizers, or participants with functional CYP2C19 alleles. (NCT00833482)
Timeframe: Predose and at 1, 2, 3, 4, 5, 7, 9,13, and 24 hours postdose on Days 20 and 30 of a 30-day cycle

Interventionng*h/mL (Geometric Mean)
Atazanavir/Ritonavir, 300/100 QD (EM)44634
Atazanavir/Ritonavir, 300/100mgQD + Voriconazole, 200mgBID(EM)38276

[back to top]

Tmax of Ritonavir, Administered As Atazanavir/Ritonavir With and Without Voriconazole, in EM Participants

Tmax=time to maximum concentration; EM=extensive metabolizers, or participants with functional CYP2C19 alleles. (NCT00833482)
Timeframe: Predose and at 1, 2, 3, 4, 5, 7, 9,13, and 24 hours postdose on Days 20 and 30 of a 30-day cycle

InterventionHours (Median)
Atazanavir/Ritonavir, 300/100 QD (EM)4.0
Atazanavir/Ritonavir, 300/100mgQD + Voriconazole, 200mgBID(EM)4.0

[back to top]

Number of Participants With Marked Abnormalities in Hematology Laboratory Test and Urinalysis Results

LLN=lower limit of normal; ULN=upper limit of normal; preRX=pretreatment. Safety criteria: Neutrophils + bands: If <.85*LLN or >1.15*ULN or ULN or if preRXULN; if preRX>ULN, use >1.15*preRX or 1.15*ULN, or if preRX ULN; if preRX >ULN, use >1.15*preRX or = 2+, or if preRX >=1+, use >=2*preRX. White blood cells, urine: If >=2+, or if preRX >=2+, use >=4+. Red blood cells, urine: If >=2+ or if preRX >=2+, use >=4+. Not all categories were evaluated for each arm. (NCT00833482)
Timeframe: Within 21 days of Day 1 and on Days 3, 10, 20, 26, and 31 (at discharge)

,,,,,
InterventionParticipants (Number)
Neutrophils + bands(absolute)(*10^3 cells/uL): LowLymphocytes, relative (*10*3 cells/uL): LowLymphocytes, relative (*10*3 cells/uL): HighBlood, urine: HighWhite blood cells (WBC), urine: HighRed blood cells (RBC), urine: High
Atanazivir/Ritonavir, 300/100 QD + Voriconazole, 50 BID (PM)0NANA0NANA
Atazanavir/Ritonavir, 300/100 QD (EM)011100
Atazanavir/Ritonavir, 300/100 QD (PM)0NANA0NANA
Atazanavir/Ritonavir, 300/100mgQD + Voriconazole, 200mgBID(EM)1112NANA
Voriconazole, 200 BID (EM)115111
Voriconazole, 50 mg BID (PM)0NANA0NANA

[back to top]

Number of Participants With Marked Abnormalities in Serum Chemistry Test Results

LLN=lower limit of normal; ULN=upper limit of normal; preRX=pretreatment. Safety criteria: AST and ALT: If >1.25*ULN, or if preRX>ULN, use >1.25*preRX. Total and direct bilirubin: If >1.1*ULN or if preRX>ULN, use >1.25*preRX. Creatinine: If >1.33*preRX. Serum glucose, fasting: If preRXULN; if preRX>ULN, use >2*preRX or 1.5*ULN or preRX>ULN, use >1.5*or preRX. Lactose dehydrogenase: If >1.25*ULN or preRX>ULN, use >1.5*preRX. (NCT00833482)
Timeframe: Within 21 days of Day 1 and on Days 3, 10, 20, 26, and 31 (at discharge)

,,,,,
InterventionParticipants (Number)
Aspartate aminotransferase (AST) (U/L): HighAlanine aminotransferase (ALT) (U/L): HighTotal bilirubin (mg/dL): HighDirect bilirubin (mg/dL): HighCreatinine (mg/dL): HighSerum glucose, fasting (mg/dL): LowCreatine kinase (U/L): HighLactase dehydrogenase (U/L): High
Atazanavir/Ritonavir, 300/100 QD (EM)012200011
Atazanavir/Ritonavir, 300/100 QD (PM)01721000
Atazanavir/Ritonavir, 300/100 QD + Voriconazole, 50 BID (PM)01820000
Atazanavir/Ritonavir, 300/100mgQD + Voriconazole, 200mgBID(EM)112100030
Voriconazole, 200 BID (EM)00400101
Voriconazole, 50 mg BID (PM)00000000

[back to top]

Tmax of Voriconazole, Administered With and Without Atazanavir/Ritonavir, in EM Participants

Tmax=time to maximum concentration; EM=extensive metabolizers, or participants with functional CYP2C19 alleles. (NCT00833482)
Timeframe: Predose and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours postdoseDays 3 and 30 of a 30-day cycle

InterventionHours (Median)
Voriconazole, 200 BID (EM)1.50
Atazanavir/Ritonavir, 300/100mgQD + Voriconazole, 200mgBID(EM)1.25

[back to top]

Time to Maximum Concentration (Tmax) of Atazanavir, Administered as Atazanavir/Ritonavir With and Without Voriconazole, in EM Participants

EM=extensive metabolizers, or participants with functional CYP2C19 alleles. (NCT00833482)
Timeframe: Predose and at 1, 2, 3, 4, 5, 7, 9,13, and 24 hours postdose on Days 20 and 30 of a 30-day cycle

InterventionHours (Median)
Atazanavir/Ritonavir, 300/100 QD (EM)3.0
Atazanavir/Ritonavir, 300/100mgQD + Voriconazole, 200mgBID(EM)2.07

[back to top]

AUC(TAU)of Voriconazole, Administered With and Without Atazanavir/Ritonavir, in EM Participants

AUC(TAU)=area under the plasma concentration-time curve in 1 dosing interval; EM=extensive metabolizers, or participants with functional CYP2C19 alleles. (NCT00833482)
Timeframe: Predose and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours postdoseDays 3 and 30 of a 30-day cycle

Interventionng.h/mL (Geometric Mean)
Voriconazole, 200 BID (EM)20284
Atazanavir/Ritonavir, 300/100mgQD + Voriconazole, 200mgBID(EM)12944

[back to top]

Maximum Observed Plasma Concentration (Cmax) and Minimum Observed Plasma Concentration (Cmin) of Atazanavir, Administered as Atazanavir/Ritonavir With and Without Voriconazole, in Participants Who Are Extensive Metabolizers (EM)

EM participants are those with functional CYP2C19 alleles. (NCT00833482)
Timeframe: Predose and at 1, 2, 3, 4, 5, 7, 9,13, and 24 hours postdose on Days 20 and 30 of a 30-day cycle

,
Interventionng/mL (Geometric Mean)
Atazanavir CminAtazanavir Cmax
Atazanavir/Ritonavir, 300/100 QD (EM)6744715
Atazanavir/Ritonavir, 300/100mgQD + Voriconazole, 200mgBID(EM)5254076

[back to top]

Number of Participants With Abnormalities in Vital Signs

(NCT00833482)
Timeframe: Within 21 days of Day 1 and on Days -1, 1, 3, 11, 21, and 31 (at discharge)

,,,,,
InterventionParticipants (Number)
Diastolic blood pressureSystolic blood pressureHeart rateRespiratory rateTemperature
Atanazivir/Ritonavir, 300/100 QD + Voriconazole, 50 BID (PM)00000
Atazanavir/Ritonavir, 300/100 QD (EM)00000
Atazanavir/Ritonavir, 300/100 QD (PM)00000
Atazanavir/Ritonavir, 300/100mgQD + Voriconazole, 200mgBID(EM)00000
Voriconazole, 200 BID (EM)00000
Voriconazole, 50 mg BID (PM)00000

[back to top]

Number of Participants With Death as Outcome, Serious Adverse Events (SAEs), Adverse Events (AEs) Leading to Discontinuation, and Any AE

AE=any new unfavorable symptom, sign, or disease or worsening of a preexisting condition that may not have a causal relationship with treatment. SAE=a medical event that at any dose results in death, persistent or significant disability/incapacity, or drug dependency/abuse; is life-threatening, an important medical event, or a congenital anomaly/birth defect; or requires or prolongs hospitalization. (NCT00833482)
Timeframe: Days 1 to 31 (discharge), continuously

,,,,,
InterventionParticipants (Number)
DeathsSAEsAEs leading to discontinuationAny AE
Atanazivir/Ritonavir, 300/100 QD + Voriconazole, 50 BID (PM)0003
Atazanavir/Ritonavir, 300/100 QD (EM)00224
Atazanavir/Ritonavir, 300/100 QD (PM)0002
Atazanavir/Ritonavir, 300/100mgQD + Voriconazole, 200mgBID(EM)00121
Voriconazole, 200 BID (EM)00022
Voriconazole, 50 mg BID (PM)0000

[back to top]

Number of Participants With Investigator-identified Abnormalities in Electrocardiogram Results Not Present Prior to Administration of Study Drug and Considered Not Relevant and Not AEs by Investigator

volt=voltage; LVH=left ventricular hypertrophy (NCT00833482)
Timeframe: Within 21 days of Day 1 and on Days -1, 21, and 31 (at discharge)

,,,,,
InterventionParticipants (Number)
Sinus bradycardiaST elevation and fusion complexesT-wave abnormality + minimum volt criteria for LVHNonspecific T-wave abnormality
Atanazivir/Ritonavir, 300/100 QD + Voriconazole, 50 BID (PM)2111
Atazanavir/Ritonavir, 300/100 QD (EM)0000
Atazanavir/Ritonavir, 300/100 QD (PM)0000
Atazanavir/Ritonavir, 300/100mgQD + Voriconazole, 200mgBID(EM)0000
Voriconazole, 200 BID (EM)0000
Voriconazole, 50 mg BID (PM)0000

[back to top]

Cmax and Cmin of Voriconazole, Administered With and Without Atazanavir/Ritonavir, in EM Participants

Cmax=maximum observed plasma concentration; Cmin=minimum observed plasma concentration; EM=extensive metabolizers, or participants with functional CYP2C19 alleles. (NCT00833482)
Timeframe: Predose and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours postdoseDays 3 and 30 of a 30-day cycle

,
Interventionng/mL (Geometric Mean)
Voriconazole CmaxVoriconazole Cmin
Atazanavir/Ritonavir, 300/100mg QD + Voriconazole, 200 mg BID3014439
Voriconazole, 200 BID3416776

[back to top]

Cmax and Cmin of Ritonavir, Administered As Atazanavir/Ritonavir With and Without Voriconazole, in EM Participants

Cmax=maximum observed plasma concentration; Cmin=minimum observed plasma concentration; EM=extensive metabolizers, or participants with functional CYP2C19 alleles. (NCT00833482)
Timeframe: Predose and at 1, 2, 3, 4, 5, 7, 9,13, and 24 hours postdose on Days 20 and 30 of a 30-day cycle

,
Interventionng/mL (Geometric Mean)
Ritonavir CmaxRitonavir Cmin
Atazanavir/Ritonavir, 300/100 QD (EM)159737.1
Atazanavir/Ritonavir, 300/100mgQD + Voriconazole, 200mgBID(EM)142928.3

[back to top]

Fluctuation Index of TMC310911

Fluctuation index, ie, percentage fluctuation: variation between maximum (Cmax) and minimum (Cmin) plasma concentration at steady-state, calculated as: 100 x ([Cmax-Cmin]/Css,av). Css,av is an average steady-state plasma concentration. (NCT00838162)
Timeframe: Day 14

InterventionPercent ng/mL (Mean)
TMC310911/Rtv 75/100 mg Twice Daily181.0
TMC310911/Rtv 150/100 mg Twice Daily161.3
TMC310911/Rtv 300/100 mg Twice Daily129.2
TMC310911/Rtv 300/100 mg Once a Day331.8

[back to top]

Average Steady-state Plasma Concentration (Css,av) of TMC310911

(NCT00838162)
Timeframe: Day 14

Interventionng/mL (Mean)
TMC310911/Rtv 75/100 mg Twice Daily336.2
TMC310911/Rtv 150/100 mg Twice Daily889.6
TMC310911/Rtv 300/100 mg Twice Daily1340
TMC310911/Rtv 300/100 mg Once a Day887.3

[back to top]

Time to Reach the Maximum Plasma Concentration (Tmax) of TMC310911

(NCT00838162)
Timeframe: Day 1 and Day 14

,,,
Interventionhours (Median)
Day 1Day 14
TMC310911/Rtv 150/100 mg Twice Daily5.003.00
TMC310911/Rtv 300/100 mg Once a Day5.003.02
TMC310911/Rtv 300/100 mg Twice Daily5.063.87
TMC310911/Rtv 75/100 mg Twice Daily5.024.98

[back to top]

Predose Plasma Concentration (C0h) of TMC310911

(NCT00838162)
Timeframe: Day 2, Day 3, Day 4, Day 6, Day 8, Day 10, Day 12 and Day 14

,,,
Interventionng/mL (Mean)
Day 2Day 3Day 4Day 6Day 8Day 10Day 12Day 14
TMC310911/Rtv 150/100 mg Twice Daily207.7448.0291.3443.0665.9394.4518.6420.9
TMC310911/Rtv 300/100 mg Once a Day45.48125.9191.0250.0239.6251.7250.2209.9
TMC310911/Rtv 300/100 mg Twice Daily231.9586.31035960.51028736.11110924.8
TMC310911/Rtv 75/100 mg Twice Daily79.9593.9888.94119.1134.578.51104.6152.5

[back to top]

Number of Participants With Virologic Response at Any Timepoint During the 14-day Treatment Period

Virologic response is a viral load test result below a chosen threshold value (less than 50 copies/mL, less than 400 copies/mL, or at least 1 log drop in viral load) at any timepoint during a 14-day treatment of 4 different dose regimens of TMC310911 coadministered with 100 mg ritonavir. (NCT00838162)
Timeframe: 14 days

,,,
InterventionParticipants (Number)
Viral load less than 50 copies/mLViral load less than 400 copies/mLAt least 1 log10 Viral Load Drop
TMC310911/Rtv 150/100 mg Twice Daily148
TMC310911/Rtv 300/100 mg Once a Day007
TMC310911/Rtv 300/100 mg Twice Daily018
TMC310911/Rtv 75/100 mg Twice Daily039

[back to top]

Mean Changes From Baseline in Plasma log10 Human Immunodeficiency Virus Type 1 Ribonucleic Acid (HIV-1 RNA)

The antiviral activity of TMC310911 is measured by the change in viral load from baseline in the 14 days of treatment following initiation of treatment with 4 different dosing regimens of TMC310911 coadministered with ritonavir. (NCT00838162)
Timeframe: Baseline (Day 1), Day 8, Day 15

,,,
Interventionlog10 copies/mL (Mean)
BaselineDay 8Day 15
TMC310911/Rtv 150/100 mg Twice Daily4.36-1.14-1.79
TMC310911/Rtv 300/100 mg Once a Day4.71-1.06-1.55
TMC310911/Rtv 300/100 mg Twice Daily4.78-1.07-1.69
TMC310911/Rtv 75/100 mg Twice Daily4.72-1.30-1.53

[back to top]

Mean Changes From Baseline in CD4+ Cell Count

(NCT00838162)
Timeframe: Baseline (Day 1), Day 8, Day 15

,,,
Interventionx 1000000 cells/L (Mean)
BaselineDay 8Day 15
TMC310911/Rtv 150/100 mg Twice Daily464.940.3-2.8
TMC310911/Rtv 300/100 mg Once a Day479.617.992.5
TMC310911/Rtv 300/100 mg Twice Daily494.0-14.8-2.0
TMC310911/Rtv 75/100 mg Twice Daily575.7-21.0-33.7

[back to top]

Maximum Plasma Concentration (Cmax) of TMC310911

(NCT00838162)
Timeframe: Day 1 and Day 14

,,,
Interventionng/mL (Mean)
Day 1Day 14
TMC310911/Rtv 150/100 mg Twice Daily623.01674
TMC310911/Rtv 300/100 mg Once a Day19242963
TMC310911/Rtv 300/100 mg Twice Daily706.32256
TMC310911/Rtv 75/100 mg Twice Daily487.9589.7

[back to top]

Area Under the Plasma Concentration-time Curve (AUC12) From the Time of Administration of TMC310911 up to 12 Hours After Dosing

(NCT00838162)
Timeframe: Day 1 and Day 14

,,,
Interventionng.h/mL (Mean)
Day 1Day 14
TMC310911/Rtv 150/100 mg Twice Daily273510680
TMC310911/Rtv 300/100 mg Once a Day1004017520
TMC310911/Rtv 300/100 mg Twice Daily299016010
TMC310911/Rtv 75/100 mg Twice Daily25194031

[back to top]

Percent Change in Bone Mineral Density (BMD) of the Hip From Study Entry to Week 96

Bone mineral density (BMD) of the hip was evaluated by dual-energy x-ray absorptiometry (DXA) scans at study entry and study week 96. The percent change was calculated as ((week 96 value - study entry value) / study entry value)) x 100. (NCT00851799)
Timeframe: Study entry, week 96

Interventionpercent (Median)
Cohort A: ATV/RTV + FTC/TDF-3.7
Cohort B: RAL + FTC/TDF-2.2
Cohort C: DRV/RTV + FTC/TDF-3.3

[back to top]

Percent Change in Visceral Abdominal Fat (VAT) From Study Entry to Week 96

Visceral abdominal fat (VAT) was evaluated by single slice abdominal computerized tomography scans at study entry and week 96. The percent change was calculated as as ((week 96 value - study entry value) / study entry value)) x 100. (NCT00851799)
Timeframe: Study entry, week 96

Interventionpercent (Median)
Cohort A: ATV/RTV + FTC/TDF10.7
Cohort B: RAL + FTC/TDF16.2
Cohort C: DRV/RTV + FTC/TDF9.5

[back to top]

Fold Change in High Sensitivity C-reactive Protein (hsCRP) From Study Entry to Weeks 48 and 96

hsCRP was measured at study entry and weeks 48 and 96 (unit of measure ug/ml). Fold change from study entry to week 48 or week 96 was calculated as (week 48 value or week 96 value) / (study entry value). Results identified above or below the limit of quantification were imputed at the quantification limit of the respective assay. (NCT00851799)
Timeframe: Study entry, weeks 48 and 96

,,
InterventionFold change (Median)
Fold change from study entry to week 48Fold change from study entry to week 96
Cohort A: ATV/RTV + FTC/TDF0.750.85
Cohort B: RAL + FTC/TDF0.880.78
Cohort C: DRV/RTV + FTC/TDF0.781.31

[back to top]

Fold Change in Interleukin-6 (IL-6) From Study Entry to Weeks 48 and 96

IL-6 was measured at study entry and weeks 48 and 96 (unit of measure pg/ml). Fold change from study entry to week 48 or week 96 was calculated as (week 48 value or week 96 value) / (study entry value). Results identified above or below the limit of quantification were imputed at the quantification limit of the respective assay. (NCT00851799)
Timeframe: Study entry, weeks 48 and 96

,,
InterventionFold change (Median)
Fold change from study entry to week 48Fold change from study entry to week 96
Cohort A: ATV/RTV + FTC/TDF0.620.89
Cohort B: RAL + FTC/TDF0.710.82
Cohort C: DRV/RTV + FTC/TDF0.750.89

[back to top]

Fold Change in Percent Expression of CD38+HLADR+ on CD4+ (Percent) From Study Entry to Weeks 24 and 96

Percent expression of CD38+HLADR+ on CD4+ was measured at study entry and weeks 24 and 96 (unit of measure percent). Fold change from study entry to week 24 or week 96 was calculated as (week 24 value or week 96 value) / (study entry value). (NCT00851799)
Timeframe: Study entry, weeks 24 and 96

,,
InterventionFold change (Median)
Fold change from study entry to week 24Fold change from study entry to week 96
Cohort A: ATV/RTV + FTC/TDF0.490.38
Cohort B: RAL + FTC/TDF0.510.34
Cohort C: DRV/RTV + FTC/TDF0.520.37

[back to top]

Change in CD4+ T-cell Count From Study Entry to Weeks 24, 48, 96 and 144

Change was calculated as (CD4+ T-cell count at week 24, 48, 96, or 144) - (CD4+ T-cell count at study entry). (NCT00851799)
Timeframe: Study entry to weeks 24, 48, 96, and 144

,,
Interventioncell/mm^3 (Median)
Change from study entry to week 24Change from study entry to week 48Change from study entry to week 96Change from study entry to week 144
Cohort A: ATV/RTV + FTC/TDF161209280305
Cohort B: RAL + FTC/TDF133191247279
Cohort C: DRV/RTV + FTC/TDF118194248227

[back to top]

Fold Change in Percent Expression of CD38+HLADR+ on CD8+ (Percent) From Study Entry to Weeks 24 and 96

Percent expression of CD38+HLADR+ on CD8+ was measured at study entry and weeks 24 and 96 (unit of measure percent). Fold change from study entry to week 24 or week 96 was calculated as (week 24 value or week 96 value) / (study entry value). (NCT00851799)
Timeframe: Study entry, weeks 24 and 96

,,
InterventionFold change (Median)
Fold change from study entry to week 24Fold change from study entry to week 96
Cohort A: ATV/RTV + FTC/TDF0.510.35
Cohort B: RAL + FTC/TDF0.560.36
Cohort C: DRV/RTV + FTC/TDF0.590.38

[back to top]

Fold Change in Soluble CD14 From Study Entry to Weeks 48 and 96

Soluble CD14 was measured at study entry and weeks 48 and 96 (unit of measure ng/ml). Fold change from study entry to week 48 or week 96 was calculated as (week 48 value or week 96 value) / (study entry value). Results identified above or below the limit of quantification were imputed at the quantification limit of the respective assay. (NCT00851799)
Timeframe: Study entry, weeks 48 and 96

,,
InterventionFold change (Median)
Fold change from study entry to week 48Fold change from study entry to week 96
Cohort A: ATV/RTV + FTC/TDF1.010.98
Cohort B: RAL + FTC/TDF0.910.90
Cohort C: DRV/RTV + FTC/TDF1.000.98

[back to top]

Annual Rate of Change in Right Common Carotid Artery Intima-media Thickness (CIMT)

"Right common carotid artery intima-media thickness was measured by ultrasound scan at study entry and weeks 48, 96 and 144.~The annual rate of change in right common carotid artery intima-media thickness (CIMT) was estimated over 144 weeks from study entry using mixed effects linear regression model that adjusted for screening HIV-1 RNA level and Framingham risk score stratification factors." (NCT00851799)
Timeframe: Study entry, week 144

Interventionmicron/year (Mean)
Cohort A: ATV/RTV + FTC/TDF8.2
Cohort B: RAL + FTC/TDF10.7
Cohort C: DRV/RTV + FTC/TDF12.9

[back to top]

Change in Brachial Artery (BA) Flow Mediated Dilation (FMD) From Study Entry to Week 24

"Brachial artery flow mediated dilation was measured by brachial artery reactivity tests. All results reflect measures captured from participants who reported fasting and not smoking for at least 8 hours prior to FMD assessments.~The change from study entry to week 24 in brachial artery FMD (%) was defined as the maximum FMD (%) calculated from resting heart rate (RH) 60 seconds and RH 90 seconds, relative to resting brachial artery diameter." (NCT00851799)
Timeframe: Study entry, week 24

Interventionpercent (Median)
Cohort A: ATV/RTV + FTC/TDF-0.05
Cohort B: RAL + FTC/TDF-0.27
Cohort C: DRV/RTV + FTC/TDF0.15

[back to top]

Fold Change in Soluble CD163 From Study Entry to Weeks 48 and 96

Soluble CD163 was measured at study entry and weeks 48 and 96 (unit of measure ng/ml). Fold change from study entry to week 48 or week 96 was calculated as (week 48 value or week 96 value) / (study entry value). Results identified above or below the limit of quantification were imputed at the quantification limit of the respective assay. (NCT00851799)
Timeframe: Study entry, weeks 48 and 96

,,
InterventionFold change (Median)
Fold change from study entry to week 48Fold change from study entry to week 96
Cohort A: ATV/RTV + FTC/TDF0.540.51
Cohort B: RAL + FTC/TDF0.620.56
Cohort C: DRV/RTV + FTC/TDF0.610.58

[back to top]

Percent Change in Bone Mineral Density (BMD) of the Lumber Spine From Study Entry to Week 96

Bone mineral density (BMD) of the lumber spine was evaluated by dual-energy x-ray absorptiometry (DXA) scans at study entry and study week 96. The percent change was calculated as ((week 96 value - study entry value) / study entry value)) x 100. (NCT00851799)
Timeframe: Study entry, week 96

Interventionpercent (Median)
Cohort A: ATV/RTV + FTC/TDF-4.0
Cohort B: RAL + FTC/TDF-1.6
Cohort C: DRV/RTV + FTC/TDF-3.1

[back to top]

Percent Change in Bone Mineral Density (BMD) of the Total Body From Study Entry to Week 96

Bone mineral density (BMD) of the total body was evaluated by dual-energy x-ray absorptiometry (DXA) scans at study entry and study week 96. The percent change was calculated as ((week 96 value - study entry value) / study entry value)) x 100. (NCT00851799)
Timeframe: Study entry, week 96

Interventionpercent (Median)
Cohort A: ATV/RTV + FTC/TDF-1.9
Cohort B: RAL + FTC/TDF-0.9
Cohort C: DRV/RTV + FTC/TDF-1.0

[back to top]

Percent Change in Lean Mass From Study Entry to Week 96

Lean mass was evaluated by dual-energy x-ray absorptiometry (DXA) scans at study entry and week 96. The percent change was calculated as ((week 96 value - study entry value) / study entry value)) x 100. (NCT00851799)
Timeframe: Study entry, week 96

Interventionpercent (Median)
Cohort A: ATV/RTV + FTC/TDF1.8
Cohort B: RAL + FTC/TDF1.7
Cohort C: DRV/RTV + FTC/TDF0.1

[back to top]

Percent Change in Subcutaneous Abdominal Fat (SAT) From Study Entry to Week 96

Subcutaneous abdominal fat (SAT) was evaluated by single slice abdominal computerized tomography scans at study entry and week 96. The percent change was calculated as ((week 96 value - study entry value) / study entry value)) x 100. (NCT00851799)
Timeframe: Study entry, week 96

Interventionpercent (Median)
Cohort A: ATV/RTV + FTC/TDF10.3
Cohort B: RAL + FTC/TDF11.8
Cohort C: DRV/RTV + FTC/TDF11.4

[back to top]

Percent Change in Total Limb Fat From Study Entry to Week 96

Total limb fat was evaluated by dual-energy x-ray absorptiometry (DXA) scans at study entry and week 96. The percent change was calculated as ((week 96 value - study entry value) / study entry value)) x 100. (NCT00851799)
Timeframe: Study entry, week 96

Interventionpercent (Median)
Cohort A: ATV/RTV + FTC/TDF9.8
Cohort B: RAL + FTC/TDF6.3
Cohort C: DRV/RTV + FTC/TDF7.9

[back to top]

Percent Change in Trunk Fat From Study Entry to Week 96

Trunk fat was evaluated by dual-energy x-ray absorptiometry (DXA) scans at study entry and week 96. The percent change was calculated as ((week 96 value - study entry value) / study entry value)) x 100. (NCT00851799)
Timeframe: Study entry, week 96

Interventionpercent (Median)
Cohort A: ATV/RTV + FTC/TDF10.8
Cohort B: RAL + FTC/TDF13.5
Cohort C: DRV/RTV + FTC/TDF9.7

[back to top]

Change in Fasting Calculated Low-density Lipoprotein Cholesterol (LDL-C) From Study Entry to Weeks 4, 24, 48 and 96

Calculated LDL-C (unit of measure mg/dL) was measured at study entry and weeks 4, 24, 48 and 96 from participants who reported fasting for at least 8 hours prior to assessment; all results were centrally laboratory tested in batch. Change in calculated LDL-C was calculated as (week 4, 24, 48 or 96 result) - (study entry result). (NCT00851799)
Timeframe: Study entry, weeks 4, 24, 48 and 96

,,
Interventionmg/dL (Median)
Change from study entry to week 4Change from study entry to week 24Change from study entry to week 48Change from study entry to week 96
Cohort A: ATV/RTV + FTC/TDF0212
Cohort B: RAL + FTC/TDF-3-2-1-1
Cohort C: DRV/RTV + FTC/TDF1356

[back to top]

CD4+ T-cell Count at Study Entry and Weeks 24, 48, 96 and 144

The absolute levels of CD4+ T-cell counts (cells/mm^3) measured at study entry and weeks 24, 48, 96 and 144. (NCT00851799)
Timeframe: Study entry, weeks 24, 48, 96 and 144

,,
Interventioncell/mm^3 (Median)
Study EntryWeek 24Week 48Week 96Week 144
Cohort A: ATV/RTV + FTC/TDF350509573634658
Cohort B: RAL + FTC/TDF343445496569613
Cohort C: DRV/RTV + FTC/TDF355464528567560

[back to top]

Change in Absolute Flow Mediated Dilation (FMD) From Study Entry to Weeks 4, 24 and 48

The change in absolute FMD was defined as the maximum absolute FMD from the RH 60 and 90 second measurements, from study entry to weeks 4, 24, and 48 (unit of measure millimeters). All results reflect measures captured from participants who reported fasting and not smoking for at least 8 hours prior to FMD assessments. (NCT00851799)
Timeframe: Study entry, weeks 4, 24 and 48

,,
Interventionmm (Mean)
Change from study entry to week 4Change from study entry to week 24Change from study entry to week 48
Cohort A: ATV/RTV + FTC/TDF0.002-0.0020.002
Cohort B: RAL + FTC/TDF0.012-0.0040.005
Cohort C: DRV/RTV + FTC/TDF-0.0050.008-0.001

[back to top]

Change in Brachial Artery Flow Mediated Dilation (FMD) From Study Entry to Weeks 4 and 48

"Brachial artery flow mediated dilation was measured by brachial artery reactivity tests. All results reflect measures captured from participants who reported fasting and not smoking for at least 8 hours prior to FMD assessments.~The change from study entry to weeks 4 and 48 in brachial artery FMD (%) was defined as the maximum FMD (%) calculated from resting heart rate (RH) 60 seconds and RH 90 seconds, relative to resting brachial artery diameter." (NCT00851799)
Timeframe: Study entry, weeks 4 and 48

,,
Interventionpercent (Mean)
Change from study entry to week 4Change from study entry to week 48
Cohort A: ATV/RTV + FTC/TDF-0.04-0.04
Cohort B: RAL + FTC/TDF0.22-0.08
Cohort C: DRV/RTV + FTC/TDF-0.15-0.11

[back to top]

Fold Change in D-dimer From Study Entry to Weeks 48 and 96

D-dimer was measured at study entry and weeks 48 and 96 (unit of measure ug/ml). Fold change from study entry to week 48 or week 96 was calculated as (week 48 value or week 96 value) / (study entry value). Results identified above or below the limit of quantification were imputed at the quantification limit of the respective assay. (NCT00851799)
Timeframe: Study entry, weeks 48 and 96

,,
InterventionFold change (Median)
Fold change from study entry to week 48Fold change from study entry to week 96
Cohort A: ATV/RTV + FTC/TDF0.570.52
Cohort B: RAL + FTC/TDF0.730.72
Cohort C: DRV/RTV + FTC/TDF0.650.65

[back to top]

Change in Fasting Glucose Level From Study Entry to Weeks 4, 24, 48 and 96

Glucose (unit of measure mg/dL) was measured at study entry and weeks 4, 24, 48 and 96; all results reflect measures captured from participants who reported fasting for at least 8 hours prior to assessment. Change was calculated as (fasting result during at week 4, 24, 48 or 96) - (fasting result at study entry). (NCT00851799)
Timeframe: Study entry, weeks 4, 24, 48 and 96

,,
Interventionmg/dL (Median)
Change from study entry to week 4Change from study entry to week 24Change from study entry to week 48Change from study entry to week 96
Cohort A: ATV/RTV + FTC/TDF3443
Cohort B: RAL + FTC/TDF3446
Cohort C: DRV/RTV + FTC/TDF2422

[back to top]

Change in Fasting High-density Lipoprotein Cholesterol (HDL-C) From Study Entry to Weeks 4, 24, 48 and 96

HDL cholesterol (unit of measure mg/dL) was measured at study entry and weeks 4, 24, 48 and 96 from participants who reported fasting for at least 8 hours prior to assessment; all results were centrally laboratory tested in batch. Change in HDL-C was calculated as (week 4, 24, 48 or 96 result) - (study entry result). (NCT00851799)
Timeframe: Study entry, weeks 4, 24, 48 and 96

,,
Interventionmg/dL (Median)
Change from study entry to week 4Change from study entry to week 24Change from study entry to week 48Change from study entry to week 96
Cohort A: ATV/RTV + FTC/TDF-1324
Cohort B: RAL + FTC/TDF-2324
Cohort C: DRV/RTV + FTC/TDF-3014

[back to top]

Change in Fasting Insulin Level From Study Entry to Weeks 4, 24, 48 and 96

Insulin (unit of measure uIU/dL) was measured at study entry and weeks 4, 24, 48 and 96; all results reflect measures captured from participants who reported fasting for at least 8 hours prior to assessment. Change was calculated as (fasting result during at week 4, 24, 48 or 96) - (fasting result at study entry). (NCT00851799)
Timeframe: Study entry, weeks 4, 24, 48 and 96

,,
InterventionuIU/dL (Median)
Change from study entry to week 4Change from study entry to week 24Change from study entry to week 48Change from study entry to week 96
Cohort A: ATV/RTV + FTC/TDF4.04.04.03.5
Cohort B: RAL + FTC/TDF3.03.03.03.0
Cohort C: DRV/RTV + FTC/TDF3.02.03.02.0

[back to top]

Change in Fasting Total Cholesterol (TC) From Study Entry to Weeks 4, 24, 48 and 96

Total cholesterol (TC, unit of measure mg/dL) was measured at study entry and weeks 4, 24, 48 and 96 from participants who reported fasting for at least 8 hours prior to assessment; all results were centrally laboratory tested in batch. Change in TC was calculated as (week 4, 24, 48 or 96 result) - (study entry result). (NCT00851799)
Timeframe: Study entry, weeks 4, 24, 48 and 96

,,
Interventionmg/dL (Median)
Change from study entry to week 4Change from study entry to week 24Change from study entry to week 48Change from study entry to week 96
Cohort A: ATV/RTV + FTC/TDF49812
Cohort B: RAL + FTC/TDF-7-4-11
Cohort C: DRV/RTV + FTC/TDF371214

[back to top]

Change in Fasting Triglyceride (TG) From Study Entry to Weeks 4, 24, 48 and 96

Triglyceride (TG, unit of measure mg/dL) was measured at study entry and weeks 4, 24, 48 and 96 from participants who reported fasting for at least 8 hours prior to assessment; all results were centrally laboratory tested in batch. Change in TG was calculated as (week 4, 24, 48 or 96 result) - (study entry result). (NCT00851799)
Timeframe: Study entry, weeks 4, 24, 48 and 96

,,
Interventionmg/dL (Median)
Change from study entry to week 4Change from study entry to week 24Change from study entry to week 48Change from study entry to week 96
Cohort A: ATV/RTV + FTC/TDF146910
Cohort B: RAL + FTC/TDF-12-16-13-7
Cohort C: DRV/RTV + FTC/TDF15280

[back to top]

Area Under the Plasma Concentration-Time Curve From Time of Administration to 24 Hours Post-dose (AUC0-24h)

The AUC (0-24) is the area under the plasma concentration-time curve from time zero to 24 hours post dose. The selected arms were based on the dosing frequency (once daily). (NCT00855335)
Timeframe: Between Predose and 24 hours postdose at Weeks 24-28 (Visit 4, 2nd trimester), 34-38 (visit 5, 3rd trimester) and 6-12 weeks postpartum (visit 8)

,,,,
Interventionnanogram*hour per milliliter (ng*h/mL) (Mean)
Postpartum (6-12 W)2nd trimester3rd trimester
Cobicistat 150 mg (Darunavir 800/Cobicistat 150 mg) Once Daily864338624736
Darunavir 800 mg (Darunavir 800/Cobicistat 150 mg) Once Daily996134729347991
Darunavir 800 mg (Darunavir 800/Ritonavir 100 mg) Once Daily921166228961112
Rilpivirine 25 mg Once Daily271417921762
Ritonavir 100 mg (Darunavir 800/Ritonavir 100 mg) Once Daily658439353821

[back to top]

Maximum Plasma Concentration (Cmax)

The Cmax is the maximum observed plasma concentration. (NCT00855335)
Timeframe: Between Predose and 24 hours postdose at Weeks 24-28 (Visit 4, 2nd trimester), 34-38 (visit 5, 3rd trimester) and 6-12 weeks postpartum (visit 8)

,,,,,,,
Interventionnanogram per milliliter (ng/mL) (Mean)
Postpartum (6-12 W)2nd Trimester3rd Trimester
Cobicistat 150 mg (Darunavir 800/Cobicistat 150 mg) Once Daily996571759
Darunavir 600 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily665946685328
Darunavir 800 mg (Darunavir 800/Cobicistat 150 mg) Once Daily791843404910
Darunavir 800 mg (Darunavir 800/Ritonavir 100 mg) Once Daily731049645132
Etravirine 200 mg Twice Daily569774785
Rilpivirine 25 mg Once Daily167121123
Ritonavir 100 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily1110546.8536.1
Ritonavir 100 mg (Darunavir 800/Ritonavir 100 mg) Once Daily742439397

[back to top]

Mean Change From Baseline in CD4+ Cell Count

Mean Change From Baseline in CD4+ Cell Count were assessed for immunology testing. (NCT00855335)
Timeframe: Baseline, 4 weeks after baseline, 2nd and 3rd trimesters of pregnancy and postpartum (2-5 weeks and 6-12 weeks)

Intervention10^6 Cells/Liter (Mean)
Baseline2nd trimester3rd trimesterPostpartum (2-5 weeks)Postpartum (6-12 weeks)
Darunavir 800 mg/Cobicistat 150 mg Once Daily594.1713.2972.17163244.67

[back to top]

Number of Participants With Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA) Plasma Viral Load (<) 50 Copies/Milliliter (mL)

Number of participants were assessed with a viral load (VL) lesser than (<) 50 HIV-1 RNA copies/ mL over time. (NCT00855335)
Timeframe: Up to postpartum (6-12 weeks)

,,,,
InterventionParticipants (Number)
2nd trimester Less than(<)50 copies/milliLiter(mL)3rd trimester: <50 copies/mLPostpartum (2-5 weeks): <50 copies/mLPostpartum (6-12 weeks): <50 copies/mL
Darunavir 600 mg /Ritonavir 100 Twice Daily6556
Darunavir 800 mg /Ritonavir 100 mg Once Daily9887
Darunavir 800 mg/Cobicistat 150 mg Once Daily6555
Etravirine 200 mg Twice Daily121098
Rilpivirine 25 mg Once Daily1313910

[back to top]

Predose (Trough) Plasma Concentration (C0h)

C0h is defined as the predose (trough) plasma concentration or concentration just prior to study drug administration. (NCT00855335)
Timeframe: Predose on Weeks 24-28 (Visit 4, 2nd trimester), 34-38 (visit 5, 3rd trimester) and 6-12 weeks postpartum (visit 8)

,,,,,,,
Interventionnanogram per milliliter (ng/mL) (Mean)
Postpartum (6-12 W)2nd Trimester3rd Trimester
Cobicistat 150 mg (Darunavir 800/Cobicistat 150 mg) Once Daily134NA30.1
Darunavir 600 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily360823233280
Darunavir 800 mg (Darunavir 800/Cobicistat 150 mg) Once Daily2811540824
Darunavir 800 mg (Darunavir 800/Ritonavir 100 mg) Once Daily248117931528
Etravirine 200 mg Twice Daily281439413
Rilpivirine 25 mg Once Daily12775.678.0
Ritonavir 100 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily491.4225.9236.0
Ritonavir 100 mg (Darunavir 800/Ritonavir 100 mg) Once Daily14794.274.6

[back to top]

Time to Reach the Maximum Plasma Concentration (Tmax)

The Tmax is defined as actual sampling time to reach maximum observed plasma concentration. (NCT00855335)
Timeframe: Between Predose and 24 hours postdose at Weeks 24-28 (Visit 4, 2nd trimester), 34-38 (visit 5, 3rd trimester) and 6-12 weeks postpartum (visit 8)

,,,,,,,
Interventionhour (h) (Median)
Postpartum (6-12 W)2nd Trimester3rd Trimester
Cobicistat 150 mg (Darunavir 800/Cobicistat 150 mg) Once Daily4.004.033.50
Darunavir 600 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily3.003.003.00
Darunavir 800 mg (Darunavir 800/Cobicistat 150 mg) Once Daily4.004.003.50
Darunavir 800 mg (Darunavir 800/Ritonavir 100 mg) Once Daily4.004.003.05
Etravirine 200 mg Twice Daily4.003.053.00
Rilpivirine 25 mg Once Daily4.004.004.00
Ritonavir 100 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily5.044.174.07
Ritonavir 100 mg (Darunavir 800/Ritonavir 100 mg) Once Daily4.185.926.00

[back to top]

Plasma Concentration of Drug in the Cord Plasma and Maternal Plasma Samples Collected at the Time of Delivery

The drug concentrations were evaluated in the cord plasma and maternal plasma samples collected at the time of delivery. (NCT00855335)
Timeframe: On day of delivery - Intrapartum (Visit 6)

,,,,,,,
Interventionnanogram per milliliter (ng/mL) (Mean)
Cord PlasmaMaternal Plasma
Cobicistat 150 mg (Darunavir 800/Cobicistat 150 mg) Once DailyNA74.5
Darunavir 600 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily348.42149
Darunavir 800 mg (Darunavir 800/Cobicistat 150 mg) Once Daily125857
Darunavir 800 mg (Darunavir 800/Ritonavir 100 mg) Once Daily2281663
Etravirine 200 mg Twice Daily147421
Rilpivirine 25 mg Once Daily32.859.0
Ritonavir 100 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily17.07316.7
Ritonavir 100 mg (Darunavir 800/Ritonavir 100 mg) Once DailyNA154

[back to top]

Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)

An adverse event (AE) was any untoward medical occurrence in a participant who received study drug without regard to possibility of causal relationship. A serious adverse event (SAE) is an AE resulting in any of the following outcomes or deemed significant for any other reason: death; initial or prolonged inpatient hospitalization; life-threatening experience (immediate risk of dying); persistent or significant disability/incapacity; congenital anomaly. (NCT00855335)
Timeframe: Up to follow up period (16 weeks after postpartum)

,,,,
InterventionParticipants (Number)
Any AEAny SAE
Darunavir 600 mg /Ritonavir 100 Twice Daily146
Darunavir 800 mg /Ritonavir 100 mg Once Daily176
Darunavir 800 mg/Cobicistat 150 mg Once Daily51
Etravirine 200 mg Twice Daily124
Rilpivirine 25 mg Once Daily94

[back to top]

Mean Change From Baseline in CD4+ Cell Count

Mean Change From Baseline in CD4+ Cell Count were assessed for immunology testing. (NCT00855335)
Timeframe: Baseline, 4 weeks after baseline, 2nd and 3rd trimesters of pregnancy and postpartum (2-5 weeks and 6-12 weeks)

,,,
Intervention10^6 Cells/Liter (Mean)
Baseline4 Weeks after Baseline2nd trimester3rd trimesterPostpartum (2-5 weeks)Postpartum (6-12 weeks)
Darunavir 600 mg /Ritonavir 100 Twice Daily466.3-14.837.183.5127.9174.5
Darunavir 800 mg /Ritonavir 100 mg Once Daily497.9116.3154.1274.9186.0323.0
Etravirine 200 mg Twice Daily417.476.2513.7777.30115.36154.90
Rilpivirine 25 mg Once Daily495.7924.0039.2189.46139.42168.18

[back to top]

Area Under the Plasma Concentration-Time Curve From Time of Administration to 12 Hours Post-dose (AUC0-12h)

The AUC (0-12) is the area under the plasma concentration-time curve from time zero to 12 hours post dose. The selected arms were based on the dosing frequency (twice daily). (NCT00855335)
Timeframe: Between Predose and 24 hours postdose at Weeks 24-28 (Visit 4, 2nd trimester), 34-38 (visit 5, 3rd trimester) and 6-12 weeks postpartum (visit 8)

,,
Interventionnanogram*hour per milliliter (ng*h/mL) (Mean)
Postpartum (6-12 W)2nd Trimester3rd Trimester
Darunavir 600 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily568903937045880
Etravirine 200 mg Twice Daily500466176846
Ritonavir 100 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily740637753750

[back to top]

Number of Infants With Human Immunodeficiency Virus (HIV) Positive Test Result

The infants were evaluated for HIV positive tests using HIV polymerase chain reaction test (PCR). (NCT00855335)
Timeframe: Birth to age 16 weeks

Interventioninfants (Number)
Darunavir 600 mg /Ritonavir 100 Twice Daily0
Darunavir 800 mg /Ritonavir 100 mg Once Daily0
Etravirine 200 mg Twice Daily0
Rilpivirine 25 mg Once Daily0
Darunavir 800 mg/Cobicistat 150 mg Once Daily0

[back to top]

Mean Change From Baseline in Log10 Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA) Viral Load Value

Mean change from baseline in log 10 HIV-1 RNA VL was assessed up to postpartum (6-12 weeks). (NCT00855335)
Timeframe: Baseline, 4 weeks after baseline, 2nd and 3rd trimesters of pregnancy and postpartum (2-5 weeks and 6-12 weeks)

InterventionLog 10 copies per milliliter (copies/mL) (Mean)
Baseline2nd trimester3rd trimesterPostpartum (2-5 weeks)Postpartum (6-12 weeks)
Darunavir 800 mg/Cobicistat 150 mg Once Daily1.770.10.210.180.23

[back to top]

Mean Change From Baseline in Log10 Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA) Viral Load Value

Mean change from baseline in log 10 HIV-1 RNA VL was assessed up to postpartum (6-12 weeks). (NCT00855335)
Timeframe: Baseline, 4 weeks after baseline, 2nd and 3rd trimesters of pregnancy and postpartum (2-5 weeks and 6-12 weeks)

,,,
InterventionLog 10 copies per milliliter (copies/mL) (Mean)
Baseline4 Weeks after Baseline2nd trimester3rd trimesterPostpartum (2-5 weeks)Postpartum (6-12 weeks)
Darunavir 600 mg /Ritonavir 100 Twice Daily2.12-0.26-0.19-0.31-0.180.09
Darunavir 800 mg /Ritonavir 100 mg Once Daily1.88-0.27-0.16-0.23-0.040.11
Etravirine 200 mg Twice Daily2.060.180.160.170.130.05
Rilpivirine 25 mg Once Daily1.840.200.160.250.200.08

[back to top]

Minimum Plasma Concentration (Cmin)

The Cmin is the minimum observed plasma concentration. (NCT00855335)
Timeframe: Between Predose and 24 hours postdose at Weeks 24-28 (Visit 4, 2nd trimester), 34-38 (visit 5, 3rd trimester) and 6-12 weeks postpartum (visit 8)

,,,,,,,
Interventionnanogram per milliliter (ng/mL) (Mean)
Postpartum (6-12 W)2nd Trimester3rd Trimester
Cobicistat 150 mg (Darunavir 800/Cobicistat 150 mg) Once Daily41.4NANA
Darunavir 600 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily285119222661
Darunavir 800 mg (Darunavir 800/Cobicistat 150 mg) Once Daily1538168184
Darunavir 800 mg (Darunavir 800/Ritonavir 100 mg) Once Daily147312481075
Etravirine 200 mg Twice Daily269383349
Rilpivirine 25 mg Once Daily84.054.352.9
Ritonavir 100 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily264.7141.1148.1
Ritonavir 100 mg (Darunavir 800/Ritonavir 100 mg) Once Daily40.532.228.0

[back to top]

Number of Participants With Resistance at Virological Failure

Resistance analysis was determined using genotypic and phenotypic analysis at the time of virological failure. For participants with a baseline viral load greater than (>) 200 copies/mL, virologic failure was defined as follows: HIV ribonucleic acid (RNA) levels that did not fall by at least 0.5 log 4 weeks after Baseline; viral load >1000 copies/mL (at 2 successive visits) by gestational weeks 34-38; or viral load >200 copies/mL (at 2 successive visits) after reaching a viral load less than or equal to (<=) 200 copies/mL. For participants with a baseline viral load <=200 copies/mL, virologic failure was defined as viral load of >200 copies/mL (at 2 successive visits) at any point during the study. (NCT00855335)
Timeframe: Up to follow-up phase (16 weeks after postpartum)

InterventionParticipants (Number)
Darunavir 600 mg /Ritonavir 100 Twice Daily0
Darunavir 800 mg /Ritonavir 100 mg Once Daily0
Etravirine 200 mg Twice Daily0
Rilpivirine 25 mg Once Daily0
Darunavir 800 mg/Cobicistat 150 mg Once Daily0

[back to top] [back to top]

Overall Neurocognitive Impairment Score at Week 2 or 4

Neuropsychological performance was assessed at Week 2 or 4, Week 24 and Week 48 in the following measures: Premorbid/language (Wide Range Achievement Test 4 -Reading Subtest), Learning (HVLT-R, Hopkins Verbal Learning Test-Revised), Memory (HVLT-R), Speed of Processing (Trailmaking A (Army Individual Test Battery, 1944), 1974, Stroop color, Attention (WAIS-III Symbol Search; Stroop word, Fine motor, Executive (Trailmaking B, Stroop interference, Letter, Category Fluency. An overall summary score was created by averaging all tests. Participants also completed the self-reported functional status Patient's Assessment of Own Functioning Inventory (PAOFI) and the Activities of Daily Living Scale (ADLS). Best available demographically corrected normative data were utilized to create z scores. A negative z score denotes below-average performance relative to a US normative comparison population. A higher z score is a better outcome. (NCT00855413)
Timeframe: Week 2 or 4

Interventionz score (Mean)
Acute HIV Infection Treatment Group-0.69

[back to top]

Overall Neurocognitive Impairment at Week 48

Neuropsychological performance was assessed at baseline (week 2 or 4), week 24 and week 48 in the following domain (measures): Premorbid/language (Wide Range Achievement Test (WRAT) 4 - Reading Subtest), Learning (HVLT-R, Hopkins Verbal Learning Test - Revised), Memory (HVLT-R), Speed of Processing (Trailmaking A (Army Individual Test Battery, 1944), 1974, Stroop color, Attention (WAIS-III Symbol Search; Stroop word, Fine motor, Executive (Trailmaking B, Stroop interference, Letter, Category Fluency. An overall summary score of neurocognitive functioning was created by averaging all tests. Participants also completed the self-reported functional status Patient's Assessment of Own Functioning Inventory (PAOFI) and the Activities of Daily Living Scale (ADLS). Best available demographically corrected normative data were utilized to create z scores. A negative z score denotes below-average performance relative to a US normative comparison population. A higher z score is a better outcome. (NCT00855413)
Timeframe: Week 48

Interventionz score (Mean)
Acute HIV Infection Treatment Group-.45

[back to top]

Overall Neurocognitive Impairment at Week 24

Neuropsychological performance was assessed at week 2 or 4, week 24 and week 48 in the following measures: Premorbid/language (Wide Range Achievement Test 4 -Reading Subtest), Learning (Hopkins Verbal Learning Test - Revised), Memory (HVLT-R), Speed of Processing (Trailmaking A (Army Individual Test Battery, 1944), 1974, Stroop color, Attention (WAIS-III Symbol Search; Stroop word, Fine motor, Executive (Trailmaking B, Stroop interference, Letter, Category Fluency. An overall summary score of neurocognitive functioning was created by averaging all tests. Participants also completed Patient's Assessment of Own Functioning Inventory (PAOFI) and the Activities of Daily Living Scale (ADLS). Best available demographically corrected normative data were utilized to create z scores. A negative z score denotes below-average performance relative to a US normative comparison population. A higher z score is a better outcome. (NCT00855413)
Timeframe: Week 24

Interventionz score (Mean)
Acute HIV Infection Treatment Group-0.40

[back to top]

Number of Participants With Virologic Response

Virologic response to study treatment defined as plasma HIV RNA measurement <50 copies/mL at week 48 (NCT00855413)
Timeframe: 48 weeks from enrollment

InterventionParticipants (Count of Participants)
Acute HIV Infection Treatment Group9

[back to top]

Number of Participants With Virologic Response

Virologic response defined as plasma HIV RNA measurement <200 copies/mL at week 24 (NCT00855413)
Timeframe: 24 weeks

InterventionParticipants (Count of Participants)
Acute HIV Infection Treatment Group13

[back to top]

Maximum Ritonavir Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure

(NCT00855413)
Timeframe: between week 4-12 and between Weeks 36-48

Interventionng/g (Number)
Acute HIV Infection Treatment Group14,698

[back to top]

Number of Participants With Neurocognitive Impairment at Week 48

(NCT00855413)
Timeframe: Week 48

InterventionParticipants (Count of Participants)
Acute HIV Infection Treatment Group3

[back to top]

Number of Participants With Neurocognitive Impairment at Week 24

(NCT00855413)
Timeframe: Week 24

InterventionParticipants (Count of Participants)
Acute HIV Infection Treatment Group3

[back to top]

Number of Participants With Neurocognitive Impairment at Baseline

(NCT00855413)
Timeframe: Week 2 or 4

InterventionParticipants (Count of Participants)
Acute HIV Infection Treatment Group8

[back to top]

Number of Participants With HIV RNA Measurement Above the Limits of Detection in Cerebrospinal Fluid

(NCT00855413)
Timeframe: Week 4 and Week 48

Interventionparticipants (Number)
Acute HIV Infection Treatment Group0

[back to top]

Number of Participants Who Stopped Study Treatment Due to Adverse Event or Intolerance

(NCT00855413)
Timeframe: Enrollment to Week 48

InterventionParticipants (Count of Participants)
Acute HIV Infection Treatment Group1

[back to top]

Minimum Ritonavir Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure

(NCT00855413)
Timeframe: between week 4-12 and between weeks 36-48

Interventionng/g (Number)
Acute HIV Infection Treatment Group96

[back to top]

Minimum Ritonavir Exposure Range in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture

(NCT00855413)
Timeframe: Week 4 and week 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group2

[back to top]

Minimum Ritonavir Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen

(NCT00855413)
Timeframe: Weeks 0-4 and Weeks 12, 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group2

[back to top]

Minimum Etravirine Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure

(NCT00855413)
Timeframe: between Week 4-12 and between Weeks 36-48

Interventionng/g (Number)
Acute HIV Infection Treatment Group34

[back to top]

Minimum Etravirine Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen

(NCT00855413)
Timeframe: Weeks 0-4 and weeks 12, 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group12

[back to top]

Minimum Etravirine Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture

(NCT00855413)
Timeframe: Week 4 and week 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group4

[back to top]

Minimum Darunavir Exposure Range in Semen Among Participants Who Consented to an Optional Collection of Semen

(NCT00855413)
Timeframe: Weeks 0-4 and weeks 12, 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group9

[back to top]

Minimum Darunavir Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure

(NCT00855413)
Timeframe: between week 4-12 and between weeks 36-48

Interventionng/g (Number)
Acute HIV Infection Treatment Group987

[back to top]

Minimum Darunavir Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture

(NCT00855413)
Timeframe: Week 4 and week 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group10

[back to top]

Median Time to HIV RNA Suppression to <200 Copies/mL

(NCT00855413)
Timeframe: From enrollment to the date of HIV RNA suppression, assessed up to Week 48

Interventiondays (Median)
Acute HIV Infection Treatment Group59

[back to top]

Median Change in CD4 Cell Count From Week 0 to Week 48.

(NCT00855413)
Timeframe: 48 weeks from enrollment

Interventioncells/mm^3 (Median)
Acute HIV Infection Treatment Group349

[back to top]

Median Change in CD4 Cell Count From Week 0 to Week 24.

(NCT00855413)
Timeframe: week 0, week 24

Interventioncells/mm^3 (Median)
Acute HIV Infection Treatment Group158

[back to top]

Maximum Ritonavir Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen

(NCT00855413)
Timeframe: Weeks 0-4 and Weeks 12, 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group22

[back to top]

Maximum Ritonavir Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture

(NCT00855413)
Timeframe: Week 4 and Week 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group22

[back to top]

Maximum Etravirine Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen

(NCT00855413)
Timeframe: Weeks 0-4 and weeks 12, 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group185

[back to top]

Maximum Etravirine Exposure in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure

(NCT00855413)
Timeframe: between Week 4-12 and between Weeks 36-48

Interventionng/g (Number)
Acute HIV Infection Treatment Group83,749

[back to top]

Maximum Etravirine Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture

(NCT00855413)
Timeframe: Week 4 and week 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group25

[back to top]

Maximum Darunavir Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure

(NCT00855413)
Timeframe: between Week 4-12 and between Weeks 36-48

Interventionng/g (Number)
Acute HIV Infection Treatment Group83,749

[back to top]

Maximum Darunavir Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen

(NCT00855413)
Timeframe: Weeks 0-4 and weeks 12, 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group1296

[back to top]

Maximum Darunavir Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture

(NCT00855413)
Timeframe: Week 4 and week 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group240

[back to top]

HIV RNA Levels Immediately Prior to Initiating Study Treatment.

(NCT00855413)
Timeframe: HIV RNA level at enrollment

Interventioncopies/mL (Median)
Acute HIV Infection Treatment Group1,000,000

[back to top]

HIV RNA Detection in Semen

Total cumulative levels of HIV RNA detected in the semen of participants from enrollment through week 48. (NCT00855413)
Timeframe: From enrollment through 48 weeks

Interventioncopies/mL (Mean)
Acute HIV Infection Treatment Group2541

[back to top]

HIV RNA Detection in Ileal Biopsy Specimens

Average HIV RNA detected in the ileal biopsy specimens per participant over weeks 4 and 48. (NCT00855413)
Timeframe: Weeks 4 and 48

Interventioncopies/mL (Mean)
Acute HIV Infection Treatment Group40

[back to top]

Correlation of Time to HIV RNA Levels <200 Copies/mL With Improvement in Neurocognitive Functioning From Baseline to Week 24 and 48

(NCT00855413)
Timeframe: Baseline to Week 24 and 48

Interventionr value (Number)
Acute HIV Infection Treatment Group-.82

[back to top]

Correlation of HIV RNA Levels in CSF and Drug Levels With Neurocognitive Functioning

(NCT00855413)
Timeframe: From enrollment through Week 48

Interventionr value (Number)
Acute HIV Infection Treatment GroupNA

[back to top]

Change in Overall Neurocognitive Impairment From Baseline to Week 24 or 48

Change in overall z score from baseline to week 24 or 48. A negative z score denotes below-average performance relative to a US normative comparison population. A higher z score is a better outcome. (NCT00855413)
Timeframe: Baseline to Week 24 or 48

Interventionz score (Mean)
Acute HIV Infection Treatment Group4.23

[back to top]

Ritonavir Systemic Exposure During the Follicular Phase (Days 6-10 After Menses)

Systemic exposure determined by area under the concentration time curve was measured by blood drawn for PK assessment at the following times: 0 (time of dose), 0.5, 1, 2, 4,6, 8, 12 and 24 hours. The Follicular phase starts on day 1 of the menstrual cycle when estrogen and progesterone levels are lowest. this lasts 14 days. Dose administration and PK would have been drawn on day 6, 7, 8, 9, or 10 after Day 1 (start of Follicular phase). (NCT00869960)
Timeframe: Between time of dosing to 24 hours after dose administration

Interventionmg*h/L (Mean)
Antiretroviral Therapy7.2

[back to top]

Tenofovir Systemic Exposure During the Luteal Phase (Days 20-25 After Menses)

Systemic exposure determined by area under the concentration time curve was measured by blood drawn for PK assessment at the following times: 0 (time of dose), 0.5, 1, 2, 4,6, 8, 12 and 24 hours. The Luteal phase starts on day 14 of the menstrual cycle when estrogen and progesterone levels are beginning to increase. This lasts 14 days or until Day 1 of the Follicular phase. Dose administration and PK during the Luteal phase, would have been drawn on day 20, 21, 22, 23, 24 and 25 start of Follicular phase). (NCT00869960)
Timeframe: between time of dosing tp 24 hours after dose administration

Interventionmg*h/L (Mean)
Antiretroviral Therapy2.2

[back to top]

Tenofovir Systemic Exposure During the Follicular Phase (Days 6-10 After Menses)

Systemic exposure determined by area under the concentration time curve was measured by blood drawn for PK assessment at the following times: 0 (time of dose), 0.5, 1, 2, 4,6, 8, 12 and 24 hours. The Follicular phase starts on day 1 of the menstrual cycle when estrogen and progesterone levels are lowest. this lasts 14 days. Dose administration and PK would have been drawn on day 6, 7, 8, 9, or 10 after Day 1 (start of Follicular phase). (NCT00869960)
Timeframe: between time of dosing to 24 hours after dose administered

Interventionmg*h/L (Mean)
Antiretroviral Therapy2.4

[back to top]

Ritonavir Systemic Exposure During the Luteal Phase (Days 20-25 After Menses)

Systemic exposure determined by area under the concentration time curve was measured by blood drawn for PK assessment at the following times: 0 (time of dose), 0.5, 1, 2, 4,6, 8, 12 and 24 hours. The Luteal phase starts on day 14 of the menstrual cycle when estrogen and progesterone levels are beginning to increase. This lasts 14 days or until Day 1 of the Follicular phase. Dose administration and PK during the Luteal phase, would have been drawn on day 20, 21, 22, 23, 24 and 25 start of Follicular phase). (NCT00869960)
Timeframe: Between time of dosing to 24 hours after dose administration

Interventionmg*h/L (Mean)
Antiretroviral Therapy6.7

[back to top]

Emtricitabine Systemic Exposure During the Luteal Phase (Days 20-25 After Menses)

Systemic exposure determined by area under the concentration time curve was measured by blood drawn for PK assessment at the following times: 0 (time of dose), 0.5, 1, 2, 4,6, 8, 12 and 24 hours. The Luteal phase starts on day 14 of the menstrual cycle when estrogen and progesterone levels are beginning to increase. This lasts 14 days or until Day 1 of the Follicular phase. Dose administration and PK during the Luteal phase, would have been drawn on day 20, 21, 22, 23, 24 and 25 start of Follicular phase). (NCT00869960)
Timeframe: Between time of dosing to 24 hours after dose administration

Interventionmg*h/L (Mean)
Antiretroviral Therapy10.2

[back to top]

Emtricitabine Systemic Exposure During the Follicular Phase (Days 6-10 After Menses)

Systemic exposure determined by area under the concentration time curve was measured by blood drawn for PK assessment at the following times: 0 (time of dose), 0.5, 1, 2, 4,6, 8, 12 and 24 hours. The Follicular phase starts on day 1 of the menstrual cycle when estrogen and progesterone levels are lowest. this lasts 14 days. Dose administration and PK would have been drawn on day 6, 7, 8, 9, or 10 after Day 1 (start of Follicular phase). (NCT00869960)
Timeframe: Between time of dosing to 24 hours after dose administration

Interventionmg*h/L (Mean)
Antiretroviral Therapy11.2

[back to top]

Atazanavir Systemic Exposure During the Luteal Phase (Days 20-25 After Menses)

Systemic exposure determined by area under the concentration time curve was measured by blood drawn for PK assessment at the following times: 0 (time of dose), 0.5, 1, 2, 4,6, 8, 12 and 24 hours. The Luteal phase starts on day 14 of the menstrual cycle when estrogen and progesterone levels are beginning to increase. This lasts 14 days or until Day 1 of the Follicular phase. Dose administration and PK during the Luteal phase, would have been drawn on day 20, 21, 22, 23, 24 and 25 start of Follicular phase). (NCT00869960)
Timeframe: Between time of dosing to 24 hours after dose administration

Interventionmg*h/L (Mean)
Antiretroviral Therapy22.4

[back to top]

Atazanavir Systemic Exposure During the Follicular Phase (Days 6-10 After Menses)

Systemic exposure determined by area under the concentration time curve was measured by blood drawn for PK assessment at the following times: 0 (time of dose), 0.5, 1, 2, 4,6, 8, 12 and 24 hours. The Follicular phase starts on day 1 of the menstrual cycle when estrogen and progesterone levels are lowest. this lasts 14 days. Dose administration and PK would have been drawn on day 6, 7, 8, 9, or 10 after Day 1 (start of Follicular phase). (NCT00869960)
Timeframe: Between time of dosing to 24 hours after dose administration

Interventionmg*h/L (Mean)
Antiretroviral Therapy23.9

[back to top]

"Average Change in Activated (CD38+HLADR+) CD8+ T Cells in the Ileum"

Average of changes(week 0-week 12) in the % of CD8+ T cells that are CD38+HLA-DR+, by flow cytometry (NCT00884793)
Timeframe: 12 weeks

Interventionpercentage change (Mean)
Intensification Arm-5.4

[back to top]

Number of Subjects Who Experienced an Increase in CD4% in the Ileum.

Number of subjects who experienced an increase from week 0 to week 12 in CD4+ T cells (as a % of T cells, by flow cytometry) in the ileum (NCT00884793)
Timeframe: 12 weeks

Interventionparticipants (Number)
Intensification Arm5

[back to top]

Number of Subjects Who Experienced an Increase in CD4+ T Cells (as a % of All Cells) in the Ileum.

Number of subjects who experienced an increase in CD4+ T cells (as a % of all cells) in the ileum (by flow cytometry) from week 0 to week 12. (NCT00884793)
Timeframe: 12 weeks

Interventionparticipants (Number)
Intensification Arm6

[back to top]

Number of Subjects Who Had a Decrease in HIV RNA Per Million CD4+ T Cells in the Ileum

Number of subjects who had a decrease from week 0 to week 12 in unspliced cell-associated HIV RNA per million CD4+ T cells in the ileum (NCT00884793)
Timeframe: 12 weeks

Interventionparticipants (Number)
Intensification Arm5

[back to top]

Number of Patients With Virological Failure (Two Consecutive Measures of HIV-RNA Higher Than 50 Copies/mL or a Single Measure Higher Than 1000 Copies/mL) Within 48 Weeks at intention-to.Treat Analysis

(NCT00885482)
Timeframe: 48 weeks

Interventionpatients (Number)
Single Arm1

[back to top]

AUC of Rosuvastatin

(NCT00885495)
Timeframe: 7 days

Interventionng*hr/mL (Geometric Mean)
Rosuvastatin108.96
Rosuvastatin-Darunavir-Ritonavir161.24

[back to top]

Cmax of Rosuvastatin

(NCT00885495)
Timeframe: 7 days

Interventionng/mL (Geometric Mean)
Rosuvastatin6.70
Rosuvastatin-Darunavir-Ritonavir16.32

[back to top]

To Compare the Change in Low-density Lipoprotein (LDL) Cholesterol With Rosuvastatin Therapy Alone, Darunavir/Ritonavir Therapy Alone and With the Co-administration of Rosuvastatin and Darunavir/Ritonavir.

LDL values (NCT00885495)
Timeframe: 45 days

Interventionmg/dL (Median)
Rosuvastatin Alone85
Darunavir/Ritonavir115
Rosuvastatin+Darunavir/Ritonavir80

[back to top]

To Investigate the Effect of Rosuvastatin on the Steady State Pharmacokinetics of Darunavir/Ritonavir.

Geometric mean of the Concentration minimum of darunavir and ritonavir in the presence and absence of rosuvastatin. (NCT00885495)
Timeframe: 45 days

Interventionng/mL (Geometric Mean)
Darunavir Cmin with RosuvastatinDarunavir Cmin absent RosuvastatinRitonavir Cmin absent RousuvastatinRitonavir Cmin with Rousuvastatin present
Darunavir/Ritonavir+Rosuvastatin27443235194148

[back to top]

Symptom Score

AIDS Clinical Trials Group Symptom Summary Score (20 item scale with severity from 0-4); Severity scale, 0=absent, 1=is least severe and 4 is most severe. Minimum score = 0 units on scale. Maximum score = 80 units on scale. (NCT00885664)
Timeframe: Week 4

Interventionunits on a scale (Median)
CD4<10010
CD4>/=1008

[back to top]

IL-7

Interleukin 7 measured by Luminex multiplex assay in picograms/mL, dynamic range 0.13-2000 pg/mL (NCT00885664)
Timeframe: 4 weeks

Interventionpg/mL (Median)
CD4<10017.50
CD4>/=10010.53

[back to top]

IL-8

Interleukin 8 measured by Luminex multiplex assay in picograms/mL, dynamic range 0.13-2000 pg/mL (NCT00885664)
Timeframe: 4 weeks

Interventionpg/mL (Median)
CD4<1007.58
CD4>/=1005.18

[back to top]

INF Gamma

Interferon gamma measured by Luminex multiplex assay in picograms/mL, dynamic range 0.13-2000 pg/mL (NCT00885664)
Timeframe: 4 weeks

Interventionpg/mL (Median)
CD4<1002.24
CD4>/=1001.06

[back to top]

SF-12 Mental Capacity Score

Measure of mental functioning where lower is better out of a scale of 100. (NCT00885664)
Timeframe: 4 weeks

Interventionscore on a scale (Median)
CD4<10046
CD4>/=10050

[back to top]

SF-12 Physical Capacity Score

Measure of physical function out of 100. Lower score means less physical capacity. (NCT00885664)
Timeframe: 4 weeks

Interventionunits on a scale (Median)
CD4<10043
CD4>/=10054

[back to top]

TNF Alpha

Tumor Necrosis Factor Alpha - measured by Luminex multiplex assay in picograms/mL, dynamic range 0.13-2000 pg/mL (NCT00885664)
Timeframe: 4 weeks

Interventionpg/mL (Median)
CD4<10013.07
CD4>/=1009.07

[back to top]

IL-1 Beta

Cytokine IL-1 beta measurement by Luminex multiplex assay in picograms/mL, dynamic range 0.13-2000 pg/mL (NCT00885664)
Timeframe: 4 weeks

Interventionpg/mL (Median)
CD4<1000.03
CD4>/=1000.03

[back to top]

IL-10

Interleukin 10 measured by Luminex multiplex assay in picograms/mL, dynamic range 0.13-2000 pg/mL (NCT00885664)
Timeframe: 4 weeks

Interventionpg/mL (Median)
CD4<10021.32
CD4>/=10010.30

[back to top]

IL-6

Interleukin 6 measured by Luminex multiplex assay in picograms/mL, dynamic range 0.13-2000 pg/mL (NCT00885664)
Timeframe: 4 weeks

Interventionpg/mL (Median)
CD4<1004.41
CD4>/=1004.01

[back to top]

IL-4

Interleukin-4 measured by Luminex multiplex assay in picograms/mL, dynamic range 0.13-2000 pg/mL (NCT00885664)
Timeframe: 4 weeks

Interventionpg/mL (Median)
CD4<1000.07
CD4>/=1000.07

[back to top]

Pharmacokinetic Results of Etravirine (ETR) (Results for AUC12hr)

The table below shows pharmacokinetic (PK) results of ETR in the current study expressed as the area under the plasma concentration-time curve from time of intake to 12 hours after dosing (AUC12hr). (NCT00896051)
Timeframe: Week 2

Interventionng.h/mL (Mean)
ATV/Rtv 300/100 mg (Treatment A)7629
ATV/Rtv 400/100 mg (Treatment B)5171

[back to top]

Pharmacokinetic Results of Low-Dose Ritonavir (Rtv): Treatment A: Atazanavir (ATV)/Rtv 300/100 mg (Results for AUC24hr)

The table below shows the pharmacokinetic (PK) results of low-dose ritonavir (rtv) when administered as atazanavir (ATV)/rtv 300/100 mg pretreatment (Reference) and at Week 2 after treatment (Test). Results are expressed as the area under the plasma concentration-time curve from time of intake to 24 hours after dosing (AUC24hr). (NCT00896051)
Timeframe: Day -1 (Reference); Week 2 (Test)

Interventionng.h/ml (Mean)
ATV/Rtv 300/100 mg (Reference)12560
ATV/Rtv 300/100 mg (Test)11120

[back to top]

Pharmacokinetic Results of Low-Dose Ritonavir (Rtv): Treatment B: Atazanavir (ATV)/Rtv 400/100 mg (Results for AUC24hr)

The table below shows pharmacokinetic (PK) results of low-dose ritonavir (rtv) when administered as atazanavir (ATV)/ritonavir (rtv) 300/100 mg pretreatment (Reference) and when administered as ATV/rtv 400/100 mg at Week 2 after treatment (Test). Results are expressed as the area under the plasma concentration-time curve from time of intake to 24 hours after dosing (AUC24hr). (NCT00896051)
Timeframe: Day -1 (Reference); Week 2 (Test)

Interventionng.h/ml (Mean)
ATV/Rtv 300/100 mg (Reference)13880
ATV/Rtv 300/100 mg (Test)13660

[back to top]

Change From Pre-Baseline in Log10 Viral Load Over Time

The table below shows the mean change from prebaseline over time in log10 (Copies/mL) plasma viral load using the Non-Completing = Failure (NC=F) imputation method. (NCT00896051)
Timeframe: Pre-Baseline, Baseline, Weeks 4, 12, 24, 48

,
Interventionlog10 (Copies/mL) (Mean)
BaselineWeek 4Week 12Week 24Week 48
ATV/Rtv 300/100 mg (Treatment A)-1.4-1.9-1.7-1.8-1.4
ATV/Rtv 400/100 mg (Treatment B)-1.4-1.8-2.0-1.8-1.4

[back to top]

Change From Prebaseline in CD4+ Cell Count Over Time

The table below shows the mean change from prebaseline over time in CD4+ cell count using the Non-Completing = Failure (NC=F) imputation method. (NCT00896051)
Timeframe: Prebaseline, Baseline, Weeks 4, 12, 24, 48

,
InterventionCD4+ cell count (Mean)
BaselineWeek 4Week 12Week 24Week 48
ATV/Rtv 300/100 mg (Treatment A)16553154105
ATV/Rtv 400/100 mg (Treatment B)8467283132

[back to top]

Pharmacokinetic Results of Atazanavir (ATV): Treatment A: ATV/Low-Dose Ritonavir (Rtv) 300/100 mg (Results for C0h, Cmin, and Cmax)

The table below shows pharmacokinetic (PK) results of atazanavir (ATZ) when administered as ATV/rtv 300/100 mg pretreatment (Reference) and at Week 2 after treatment (Test). Results are expressed as the predose plasma concentration (C0h), minimum plasma concentration (Cmin), and maximum plasma concentration (Cmax). (NCT00896051)
Timeframe: Day -1 (Pretreatment); Week 2 (Test)

,
Interventionng/ml (Mean)
C0h, ng/ml (Reference, n=21; Test, n=19)Cmin, ng/ml (Reference, n=20; Test, n=18)Cmax, ng/ml (Reference, n=20; Test, n=19)
ATV/Rtv 300/100 mg (Reference)133911045652
ATV/Rtv 300/100 mg (Test)845.7758.65232

[back to top]

Pharmacokinetic Results of Atazanavir (ATV): Treatment B: ATV/Low-Dose Ritonavir (Rtv) 400/100 mg (Results for C0h, Cmin, and Cmax)

The table below shows pharmacokinetic (PK) results of atazanavir (ATV) when administered as ATV/ritonavir (rtv) 300/100 mg pretreatment (Reference) and when administered as ATV/rtv 400/100 mg at Week 2 after treatment (Test). Results are expressed as the predose plasma concentration (C0h), minimum plasma concentration (Cmin), maximum plasma concentration (Cmax), and area under the plasma concentration-time curve from time of intake to 24 hours after dosing (AUC24hr). (NCT00896051)
Timeframe: Day -1 (Reference); Week 2 (Test)

,
Interventionng/ml (Mean)
C0h, ng/ml (Reference, n=22; Test, n=20)Cmin, ng/ml (Reference, n=21;Test, n=18)Cmax, ng/ml (Reference, n=22; Test, n=20)
ATV/Rtv 300/100 mg (Reference)189816716419
ATV/Rtv 400/100 mg (Test)154511076950

[back to top]

Pharmacokinetic Results of Etravirine (ETR) (Results for C0h, Cmin, and Cmax)

The table below shows pharmacokinetic (PK) results of ETR in the current study expressed as the predose plasma concentration (C0h), minimum plasma concentration (Cmin) and maximum plasma concentration (Cmax). (NCT00896051)
Timeframe: Week 2

,
Interventionng/ml (Mean)
C0h (Treatment B, n=19)Cmin (Treatment A, n=16; Treatment B, n=18)Cmax (Treatment A, n=18; Treatment B, n=18)
ATV/Rtv 300/100 mg (Treatment A)422.2425.1773.0
ATV/Rtv 400/100 mg (Treatment B)316.6286.5628.7

[back to top]

Pharmacokinetic Results of Atazanavir (ATV): Treatment B: ATV/Low-Dose Ritonavir (Rtv) 400/100 mg (Results for AUC24hr)

The table below shows pharmacokinetic (PK) results of atazanavir (ATV) when administered as ATV/ritonavir (rtv) 300/100 mg pretreatment (Reference) and when administered as ATV/rtv 400/100 mg at Week 2 after treatment (Test). Results are expressed as the area under the plasma concentration-time curve from time of intake to 24 hours after dosing (AUC24hr). (NCT00896051)
Timeframe: Day -1 (Reference); Week 2 (Test)

Interventionng.h/mL (Mean)
ATV/Rtv 300/100 mg (Reference)74210
ATV/Rtv 300/100 mg (Test)72220

[back to top]

Pharmacokinetic Results of Low-Dose Ritonavir (Rtv): Treatment B: Atazanavir (ATV)/Rtv 400/100 mg (Results for C0h, Cmin, and Cmax)

The table below shows pharmacokinetic (PK) results of low-dose ritonavir (rtv) when administered as atazanavir (ATV)/ritonavir (rtv) 300/100 mg pretreatment (Reference) and when administered as ATV/rtv 400/100 mg at Week 2 after treatment (Test). Results are expressed as the predose plasma concentration (C0h), minimum plasma concentration (Cmin), maximum plasma concentration (Cmax), and area under the plasma concentration-time curve from time of intake to 24 hours after dosing (AUC24hr). (NCT00896051)
Timeframe: Day -1 (Reference); Week 2 (Test)

,
Interventionng/ml (Mean)
C0h, ng/ml (Reference, n=22; Test, n=20)Cmin, ng/mlCmax, ng/ml (Reference, n=22)
ATV/Rtv 300/100 mg (Reference)109.264.701882
ATV/Rtv 400/100 mg (Test)163.475.681847

[back to top]

The Percentage of Participants With a Virologic Response (Plasma Viral Load < 50 Copies/mL) at Week 48 Using the Snapshot Analysis Method

The table below provides the results from the snapshot analysis method that includes the percentage of participants with virologic response (<50 copies/mL), the percentage of participants who were virologic failures (VF) (>50 copies/mL, discontinued prior to time X for reasons of VF or for other reasons, except for VF or adverse event, with a last viral load >50 copies/mL), and the percentage of participants with no viral load (VL) data available at Week 48. (NCT00896051)
Timeframe: Week 48

,
InterventionPercentage of Participants (Number)
Virologic ResponseVirologic FailureNo VL Data Available
ATV/Rtv 300/100 mg (Treatment A)50.031.818.2
ATV/Rtv 400/100 mg (Treatment B)45.536.418.2

[back to top]

Percentage of Participants With Undetectable Plasma Viral Load (VL) Values (<50 Copies/mL) at Week 48

The table below shows the percentage of participants wih undetectable plasma viral load (VL) values (<50 copies/mL) at Week 48 using the Non-Completing = Failure (NC=F) imputation method (ie, participants who discontinued early were counted as nonresponders by having their VL values after discontinuation imputed with their baseline value, thus resulting in a 0 change). (NCT00896051)
Timeframe: Week 48

InterventionPercentage of Participants (Number)
ATV/Rtv 300/100 mg (Treatment A)50.0
ATV/Rtv 400/100 mg (Treatment B)45.5

[back to top]

The Percentage of Participants With a Virologic Response Using the Non-Completing = Failure (NC=F) Imputation Method

The table below shows the percentage of participants per time point with a virologic response defined as having a plasma viral load (VL) <50 copies/mL, and with plasma VL <400 copies/mL using the Non-Completing = Failure (NC=F) imputation method (ie, participants who discontinued early were counted as nonresponders by having their VL values after discontinuation imputed with their Baseline value, thus resulting in a 0 change). (NCT00896051)
Timeframe: Baseline, Weeks 4, 12, 24, 48

,
InterventionPercentage of Participants (Number)
<50 copies/mL, Baseline<50 copies/mL, Week 4<50 copies/mL, Week 12<50 copies/mL, Week 24<50 copies/mL, Week 48<400 copies/mL, Baseline<400 copies/mL, Week 4<400 copies/mL, Week 12<400 copies/mL, Week 24<400 copies/mL, Week 48
ATV/Rtv 300/100 mg (Treatment A)9.131.859.163.650.040.977.368.272.750.0
ATV/Rtv 400/100 mg (Treatment B)9.136.459.163.645.540.977.381.872.759.1

[back to top]

Pharmacokinetic Results of Low-Dose Ritonavir (Rtv): Treatment A: Atazanavir (ATV)/Rtv 300/100 mg (Results for C0h, Cmin, and Cmax)

The table below shows the pharmacokinetic (PK) results of low-dose ritonavir (rtv) when administered as atazanavir (ATV)/rtv 300/100 mg pretreatment (Reference) and at Week 2 after treatment (Test). Results are expressed as the predose plasma concentration (C0h), minimum plasma concentration (Cmin), and maximum plasma concentration (Cmax). (NCT00896051)
Timeframe: Day -1 (Reference); Week 2 (Test)

,
Interventionng/ml (Mean)
C0h, ng/ml (Reference, n=21; Test, n=19)Cmin, ng/ml (Reference, n=20; Test, n=18)Cmax, ng/ml (Reference, n=20; Test, n=19)
ATV/Rtv 300/100 mg (Reference)143.460.421834
ATV/Rtv 300/100 mg (Test)102.543.971740

[back to top]

Pharmacokinetic Results of Atazanavir (ATV): Treatment A: ATV/Low-Dose Ritonavir (Rtv) 300/100 mg (Results for AUC24hr)

The table below shows pharmacokinetic (PK) results of atazanavir (ATZ) when administered as ATV/rtv 300/100 mg pretreatment (Reference) and at Week 2 after treatment (Test). Results are expressed as the area under the plasma concentration-time curve from time of intake to 24 hours after dosing (AUC24hr). (NCT00896051)
Timeframe: Day -1 (Pretreatment); Week 2 (Test)

Interventionng.h/mL (Mean)
ATV/Rtv 300/100 mg (Reference)60030
ATV/Rtv 300/100 mg (Test)55070

[back to top]

The Percentage of Participants With a Virologic Response Using the Time to Loss of Virologic Response (TLOVR) Imputation Method

The table below shows the percentage of participants with a virologic response defined as a viral load <50 Copies/mL and <400 Copies/mL per time point calculated using the time to loss of virologic response (TLOVR) imputation method. (NCT00896051)
Timeframe: Baseline, Weeks 4, 12, 24, 48

,
InterventionPercentage of Particpants (Number)
<50 copies/mL, Baseline<50 copies/mL, Week 4<50 copies/mL, Week 12<50 copies/mL, Week 24<50 copies/mL, Week 48<400 copies/mL, Baseline<400 copies/mL, Week 4<400 copies/mL, Week 12<400 copies/mL, Week 24<400 copies/mL, Week 48
ATV/Rtv 300/100 mg (Treatment A)9.131.859.163.645.536.477.368.268.259.1
ATV/Rtv 400/100 mg (Treatment A)4.536.454.559.150.040.977.386.468.254.5

[back to top]

Time to Virologic Failure

The table below shows the number of days to virologic failure defined as a plasma viral load (VL) > 50 copies/mL for participants who had been virologic responders (ie, having a plasma VL <50, and <400 copies/mL according to the time to loss of virologic response [TLOVR] imputation method). Time to virologic failure was the time to subsequent loss of virologic response, and the time was calculated from Prebaseline (Week -2). Participants who never achieved a virologic response were defined as nonresponders and counted as virologic failures on Day 1. (NCT00896051)
Timeframe: Prebaseline to Week 48

,
InterventionDays (Median)
Virologic Responders (Plasma VL < 50 copies/mL)Virologic Responders (Plasma VL < 400 copies/mL)
ATV/Rtv 300/100 mg (Treatment A)318.0NA
ATV/Rtv 400/100 mg (Treatment B)NANA

[back to top]

Time to Confirmed Virologic Response

The table below provides the time in days it took participants to reach a confirmed virologic response defined as a plasma viral load (VL) <50 copies/mL, and plasma VL <400 copies/mL analyzed according to the Time to Loss of Virologic Response (TLOVR) imputation method. (NCT00896051)
Timeframe: Prebaseline to Week 48

,
InterventionDays (Median)
Plasma VL < 50 copies/mLPlasma VL < 400 copies/mL
ATV/Rtv 300/100 mg (Treatment A)71.028.0
ATV/Rtv 400/100 mg (Treatment B)76.028.0

[back to top]

Virological Response [Viral Load <50 Copies/mL, FDA-SNAPSHOT]

The analysis is based on the last observed viral load (VL) data within the Week 24 window. Virologic response is defined as a VL<50 copies/mL (observed case). Virologic Failure includes a) patients who had >=50 copies/mL in the Week-24 window, b) patients who discontinued prior to Week 24 for lack or loss of efficacy, c) patients who had a switch in their background regimen that was not permitted by the protocol, and d) patients who discontinued for reasons other than adverse events (AEs)/death, and lack or loss of efficacy (provided their last available viral load was detectable). (NCT00915655)
Timeframe: Week 24

InterventionParticipants (Number)
DRV/Rtv12

[back to top]

Virological Response[Viral Load <50 Copies/mL, TLOVR]

The analysis is based on virologic response defined as percentage of patients with confirmed plasma viral load <50 HIV-1 RNA copies/mL at Week 24 calculated according to the Food and Drug Administration (FDA) Time to Loss of Virologic Response (TLOVR) algorithm. (NCT00915655)
Timeframe: Week 24

InterventionParticipants (Number)
YesNo
DRV/Rtv111

[back to top]

Number of Participants With Virological Response (Viral Load Less Than 50 Copies/mL) at Week 24 - Time to Loss of Virologic Response (TLOVR)

The TLOVR algorithm was used to derive response, ie, response and loss of response needed to be confirmed at 2 consecutive visits and participants who permanently discontinued were considered nonresponders after discontinuation. Participants with intermittent missing viral load values were considered responders if the preceeding and succeeding visits indicated response. In all other cases, intermittent values were imputed with nonresponse. Resuppression after confirmed virologic failure was considered as failure in this algorithm. (NCT00919854)
Timeframe: Week 24

InterventionParticipants (Number)
DRV/Rtv13

[back to top]

Mean Change From Baseline to Week 24 and Week 48 in CD4+ Percentage

(NCT00919854)
Timeframe: Baseline, Week 24 and Week 48

InterventionPercentage of lymphocytes (Mean)
Week 24Week 48
DRV/Rtv44

[back to top]

Mean Change From Baseline to Week 24 and Week 48 in Plasma log10 Viral Load

(NCT00919854)
Timeframe: Baseline, Week 24 and Week 48

Interventionlog10 copies/mL (Mean)
Week 24Week 48
DRV/Rtv-2.04-2.14

[back to top]

Number of Participants With Virological Response (Viral Load Less Than 50 Copies/mL) at Week 48

(NCT00919854)
Timeframe: Week 48

InterventionParticipants (Number)
DRV/Rtv17

[back to top]

Number of Participants With Virological Response (Viral Load Less Than 400 Copies/mL) at Week 24 and Week 48

(NCT00919854)
Timeframe: Week 24 and Week 48

InterventionParticipants (Number)
Week 24Week 48
DRV/Rtv1718

[back to top]

Number of Participants With Less Than or Equal to 1 log10 Decrease in Plasma Viral Load at Week 24 and Week 48

(NCT00919854)
Timeframe: Week 24 and Week 48

InterventionParticipants (Number)
Week 24Week 48
DRV/Rtv1719

[back to top] [back to top]

Number of Patients With Resistance Mutations

number of patients with resistance mutations after second line treatment failure (HIV RNA> 1000 copies/ml) (NCT00928187)
Timeframe: between W12 and W48

Interventionparticipants (Number)
Arm A0
Arm B0
Arm C0

[back to top]

Number of Patients With Plasma HIV RNA < 50 Copies/mL

(NCT00928187)
Timeframe: 48 weeks

Interventionparticipants (Number)
Arm A105
Arm B92
Arm C97

[back to top]

Number of Patients With HIV Plasma Viral Load < 200 Copies/ml

number of patients having a plasma viral load below 200 copies/ml at week 24 (NCT00928187)
Timeframe: Week 24

InterventionParticipants (Count of Participants)
Arm A127
Arm B117
Arm C129

[back to top]

Number of Patients With HIV Plasma Viral Load < 50 Copies/ml

Snapshot of patients with HIV viral load less then 50 copies/ml at week 24 (NCT00928187)
Timeframe: Week 24

InterventionParticipants (Count of Participants)
Arm A90
Arm B81
Arm C97

[back to top]

Number of Patients Discontinuing Study Treatment

number of patients discounting treatment because of adverse events (NCT00928187)
Timeframe: between baseline and W48

Interventionparticipants (Number)
Arm A0
Arm B4
Arm C1

[back to top]

Gain in CD4 Cells Between Baseline and W48

median gain in circulating CD4 cells between baseline and W48 (NCT00928187)
Timeframe: between baseline and 48 weeks

Interventioncell/mm3 (Median)
Arm A133
Arm B136
Arm C115

[back to top]

Development of Metabolic Syndrome

number of patients developing metabolic syndrome over a period of 48 weeks (NCT00928187)
Timeframe: from baseline to week 48

InterventionParticipants (Count of Participants)
Arm A12
Arm B21
Arm C9

[back to top]

Adherence

number of patients in different categories of adherence as measured by questionnaire (NCT00928187)
Timeframe: between baseline and W48

,,
Interventionparticipants (Number)
Always above 95%At least once 80-95%At least once < 80%
Arm A508911
Arm B547214
Arm C67784

[back to top]

Tolerance: Neuropathies (Grade 1 to 4)

any symptom of peripheral neuropathy (NCT00928187)
Timeframe: between baseline and W48

Interventionparticipants (Number)
Arm A5
Arm B11
Arm C8

[back to top]

Tolerance: Gastrointestinal Complains

Gastrointestinal complaints (grade 1 to 4) between baseline and W48. (NCT00928187)
Timeframe: between baseline and 48 weeks

Interventionparticipants (Number)
Arm A50
Arm B48
Arm C26

[back to top]

Tolerance: Equal or Superior to a 25% Reduction in eGFR (Glomerular Filtration Rate)

evaluation of estimated glomerular filtration rate and number of participant with a decrease equal or superior to 25% of the baseline value (NCT00928187)
Timeframe: between baseline and W48

Interventionparticipants (Number)
Arm A28
Arm B14
Arm C19

[back to top]

Patients With Plasma HIV RNA < 200 Copies/ml

number of patients with plasma HIV RNA below 200 copies/ml (NCT00928187)
Timeframe: 48 weeks

Interventionparticipants (Number)
Arm A130
Arm B118
Arm C127

[back to top]

Participants With Plasma HIV RNA < 200 Copies/mL 48 Weeks After Randomization, Per-protocol Population, VL Less Than or Equal to 100,000 Copies Per mL

The per- protocol population includes those participants who fulfil the protocol in the terms of the eligibility, interventions, and outcome assessment (NCT00931463)
Timeframe: 48 weeks

Interventionparticipants (Number)
Ritonavir-boosted Lopinavir and 2N(t)RTI188
Ritonavir-boosted Lopinavir and Raltegravir184

[back to top]

Participants With Plasma HIV RNA < 200 Copies/mL 48 Weeks After Randomization, Per-protocol Population, Non-completer Classed as Failure

"The non-completer classed as failure analysis will include all randomised participants; participants who meet the following criteria will be defined as failures:~i. week 48 HIV RNA being above each threshold ii. has missing HIV-1 RNA data for any reason iii. stops randomly assigned therapy" (NCT00931463)
Timeframe: 48 weeks

Interventionparticipants (Number)
Ritonavir-boosted Lopinavir and 2N(t)RTI208
Ritonavir-boosted Lopinavir and Raltegravir210

[back to top]

Participants With Plasma HIV RNA < 200 Copies/mL 48 Weeks After Randomization

(NCT00931463)
Timeframe: 48 weeks following randomization

Interventionparticipants (Number)
Ritonavir-boosted Lopinavir and 2N(t)RTI219
Ritonavir-boosted Lopinavir and Raltegravir223

[back to top]

Participants With Plasma HIV RNA < 200 Copies/mL 48 Weeks After Randomization, Per-protocol Population

The per- protocol population includes those participants who fulfil the protocol in the terms of the eligibility, interventions, and outcome assessment (NCT00931463)
Timeframe: 48 weeks

Interventionparticipants (Number)
Ritonavir-boosted Lopinavir and 2N(t)RTI211
Ritonavir-boosted Lopinavir and Raltegravir211

[back to top]

Participants With Plasma HIV RNA < 200 Copies/mL 48 Weeks After Randomization, Per-protocol Population, Baseline VL >100,000 Copies Per mL

The difference between treatment arms in proportion of participants with plasma HIV RNA < 200 copies/mL 48 weeks after randomization, per-protocol population: stratified analysis by baseline plasma viral load (less than or equal to 100,000 copies per mL or >100,000 copies per mL) on those participants who fulfil the protocol in the terms of the eligibility, interventions, and outcome assessment (NCT00931463)
Timeframe: 48 weeks

Interventionparticipants (Number)
Ritonavir-boosted Lopinavir and 2N(t)RTI31
Ritonavir-boosted Lopinavir and Raltegravir39

[back to top]

Change From Baseline to Nadir in Plasma HIV-1 RNA

The quantification of plasma HIV-1 RNA, was conducted for the change from baseline to on treatment nadir (maximum change) before starting HAART or Kaletra monotherapy on Day 8. The quantification was done using a PCR. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose Day 1) to Day 8

Interventionlog10 copies/mL (Mean)
GSK2248761 30 mg-1.019
Placebo-0.580

[back to top]

Change From Baseline Through Day 8 in Plasma HIV-1 RNA

The quantitative analysis of plasma was done to evaluate the amount of HIV-1 RNA at Day 1,2,3,4,5,6,7, 8 and End of treatment visit. The quantification was done using a Polymerase chain reactor (PCR). The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose Day 1) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose Day 1) to Day 8

Interventionlog 10 copies per milliliter (mL) (Mean)
GSK2248761 30 mg-0.967
Placebo-0.036

[back to top]

Change From Baseline in Hematology Paramaters- Basophils, Eosinophils, Lymphocytes, Monocytes, White Blood Cell Count

The data for hematology parameters for Basophils, eosinophils, lymphocytes, monocytes, and white blood cell count from the change from baseline was reported. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 2, 4 , 7 , Day 8 and Follow-up (Day 14)

,
Interventionthousand cells per microliter (Mean)
Basophils, Day 2Basophils, Day 4Basophils, Day 7Basophils, Day 8Basophils, Follow-UpEosinophils, Day 2Eosinophils, Day 4Eosinophils, Day 7Eosinophils, Day 8Eosinophils, Follow-UpLymphocytes, Day 2Lymphocytes, Day 4Lymphocytes, Day 7Lymphocytes, Day 8Lymphocytes, Follow-UpMonocytes, Day 2Monocytes, Day 4Monocytes, Day 7Monocytes, Day 8Monocytes, Follow-UpWhite blood cell, Day 2White blood cell, Day 4White blood cell, Day 7White blood cell, Day 8White blood cell, Follow-Up
GSK2248761 30 mg-1.78.31.73.3-3.3-1.7-60.0-38.3-96.7-63.3-120.0268.3126.7273.3-110.028.333.3-15.023.310.0148.3758.345.0500.0-90.0
Placebo-5.05.0-10.0-10.0-15.0-120.0-20.0-50.0-80.0-135.0315.0260.0180.0480.050.0125.0120.025.075.0-115.01295.0-10.0-600.0-240.0-110.0

[back to top]

GSK2248761 PK Parameters Following Dose Administration on Day 1: Apparent Clearance (CL/F)

The Clearance factor was defined as the volume of plasma cleared of the drug GSK2248761, per unit time. Serial blood samples were collected on Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours postdose on Day 1 and used for analysis. (NCT00945282)
Timeframe: Day 1 (Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours post-dose)

Interventionliter per hour (Geometric Mean)
GSK2248761 30 mg13.53

[back to top]

GSK2248761 PK Parameters Following Dose Administration on Day 7: AUC(0-τ)

AUC(0-τ) is the AUC to the end of dosing period. For Day 7, it is the AUC measured at the end of the dosing period at Day 7. Serial blood samples were collected on Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours postdose on Day 7 and used for analysis. (NCT00945282)
Timeframe: Day 7 (Pre-dose, 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours postdose)

Interventionhour*ng/mL (Geometric Mean)
GSK2248761 30 mg9679.71

[back to top]

GSK2248761 PK Parameters Following Dose Administration on Day 7: Tmax

Tmax is defined as the, time of maximum measured GSK2248761 concentration in the plasma, on Day 7. Serial blood samples were collected on Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours postdose on Day 7 and used for analysis (NCT00945282)
Timeframe: Day 7 (Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours post dose)

Interventionhours (Median)
GSK2248761 30 mg4.01

[back to top]

HIV-1 Rate of Decline by Treatment

The rate of decrease in the viral load of HIV-1 virus in response to individual treatment was measured. The viral load data was assumed to have a log normal prior distribution and followed linear decline with non-informative conjugate prior densities. The rate of decline (slope of the day) for each treatment was measured using a PCR from Day 1 to Day 8. The slope has been reported as mean. (NCT00945282)
Timeframe: Day 1 to Day 8

Interventionlog10 copies/mL (Mean)
GSK2248761 30 mg-0.1243
Placebo0.0189

[back to top]

Accumulation Ratio for AUC , Cmax, Cτ, and Time Invariance Ratio Following Repeat Administration

The accumulation ratio is based on the parameters, Cmax, AUC(0-tau), AUC(0-24), C(tau), C24, AND AUC(0-inf). The accumulation ratio Ro was the ratio of AUC(0-tau) on Day 7 to that of AUC(0-24) on Day 1; the accumulation ratio R (Cmax) was the ratio of Cmax on Day 7 to that of Cmax on Day 1; the accumulation ratio R(Ctau) was the ratio of Ctau on Day 7 to the ratio of C24 on Day 1 and the Time Invariance Ratio Rs was defined as the ratio of AUC(0-tau) on Day 7 to that of AUC(0-inf) on Day 1. The ratio has been reported as number. (NCT00945282)
Timeframe: (Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours post-dose ) on Day 1 and Day 7

Interventionratio (Number)
Accumulation Ratio RoAccumulation Ratio R [Cmax]Accumulation Ratio R[Ctau]Time Invariance Ratio Rs
GSK2248761 30 mg1.5761.2121.7501.309

[back to top]

Assessment of the Achievement of Pre-dose GSK2248761 Steady State Concentration Following Repeat Dose Administration on Day 2 Through 7

The pre-dose GSK2248761 steady state concentration, following repeated dose administration from Day 2 through Day 7 was assessed. Serial dose sampling was done on each day of Day 2, 3, 4, 5 and Day 6 and for Day 7 (Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours postdose), before the administration of the study drug daily. (NCT00945282)
Timeframe: Day 7 (Pre - dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours post-dose) and Days 2, 3, 4. 5 and 6: pre-dose only

Interventionng/mL (Mean)
Days 4, 5, 6 and 7Days 5, 6 and 7Days 6 and 7
GSK2248761 30 mg0.052-0.019-0.056

[back to top]

Change From Baseline in CD4+ and CD8+ T-lymphocyte Cell Count at Day1 and Day 8.

Whole venous blood samples were obtained from each participant for the analysis of lymphocyte subsets by flow cytometry (total lymphocyte counts, percentage, CD4+ cell counts, and CD8+ cell counts) at Screening, Day 1 and Day 8. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values (Day 1 and Day 8). Baseline was defined as Screening. (NCT00945282)
Timeframe: Baseline (Screening), Day 1 and Day 8

,
Interventionper cubic millimeter (Mean)
CD4+ cells, Day 1CD4+ cells, Day 8CD8+ cells, Day 1CD8+ cells, Day 8
GSK2248761 30 mg1.287.5123.5313.3
Placebo76.0102.0526.0531.5

[back to top]

Change From Baseline in Clinical Chemistry Paramaters- Albumin and Total Protein

The data for clinical chemistry parameters Albumin and total protein, the change from baseline was reported. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 2, 4 , 7 , Day 8 and Follow-up (Day 14)

,
Interventiongram per deciliter (Mean)
Albumin, Day 2Albumin, Day 4Albumin, Day 7Albumin, Day 8Albumin, Follow-upTotal protein, Day 2Total protein, Day 4Total protein, Day 7Total protein, Day 8Total protein, Follow-up
GSK2248761 30 mg-0.17-0.08-0.10-0.05-0.07-0.45-0.100.100.00-0.30
Placebo-0.200.00-0.05-0.100.05-0.55-0.100.05-0.25-0.20

[back to top]

Change From Baseline in Clinical Chemistry Paramaters- Alkaline Phosphatase, Alanine Amino Transferase, Aspartate Amino Transferase

The data for clinical chemistry paramaters- alkaline phosphatase, alanine amino transferase, aspartate amino transferase, the change from baseline was reported. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 2, 4 , 7 , Day 8 and Follow-up (Day 14)

,
InterventionInternational units (IU) per liter (Mean)
Alkaline phosphatase, Day 2Alkaline phosphatase, Day 4Alkaline phosphatase, Day 7Alkaline phosphatase, Day 8Alkaline phosphatase, Follow-upAlanine amino transferase, Day 2Alanine amino transferase, Day 4Alanine amino transferase, Day 7Alanine amino transferase, Day 8Alanine amino transferase, Follow-upAspartate amino transferase, Day 2Aspartate amino transferase, Day 4Aspartate amino transferase, Day 7Aspartate amino transferase, Day 8Aspartate amino transferase, Follow-up
GSK2248761 30 mg-5.8-1.51.83.34.2-3.0-2.01.73.7-7.20.8-0.2-0.31.7-0.2
Placebo-16.0-16.0-7.5-14.5-9.50.5-3.5-3.50.0-1.51.0-4.5-3.00.02.5

[back to top]

Change From Baseline in Clinical Chemistry Paramaters- Phosphorus

The data for clinical chemistry paramaters- phosphorous, the change from baseline was reported. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 2, 4 , 7 , Day 8 and Follow-up (Day 14)

,
Interventionmillimole per liter (Mean)
Phosphorus, Day 2Phosphorus, Day 4Phosphorus, Day 7Phosphorus, Day 8Phosphorus, Follow-up
GSK2248761 30 mg-0.050.07-0.17-0.27-0.58
Placebo-0.800.25-0.00-0.35-0.95

[back to top]

Change From Baseline in Clinical Chemistry Paramaters- Thyroxine Total, Thyroxine Binding Globulin, Total T3.

The data for clinical chemistry parameters Thyroxine total, thyroxine binding globulin, Total T3 the change from baseline was reported. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 2, 4 , 7 , Day 8 and Follow-up (Day 14)

,
InterventionNanomoles per liter (Mean)
Thyroxine total, Day 2Thyroxine total, Day 4Thyroxine total, Day 7Thyroxine total, Day 8Thyroxine total, Follow-upThyroxine binding globulin, Day 2Thyroxine binding globulin, Day 4Thyroxine binding globulin, Day 7Thyroxine binding globulin, Day 8Thyroxine binding globulin, Follow-upTotal T3, Day 2Total T3, Day 4Total T3, Day 7Total T3, Day 8Total T3, Follow-up
GSK2248761 30 mg-0.60-0.08-0.280.00-0.37-0.33-5.00-0.670.17-1.83-0.0130.068-0.055-0.035-0.005
Placebo-0.400.500.100.000.052.00-6.50-3.00-1.00-1.000.1700.3100.2250.1900.375

[back to top]

Change From Baseline in Clinical Chemistry Paramaters- Thyroxine, Free

The data for clinical chemistry parameters Thyroxine, free the change from baseline was reported. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 2, 4 , 7 , Day 8 and Follow-up (Day 14)

,
InterventionPicomole per liter (Mean)
Thyroxine free, Day 2Thyroxine free, Day 4Thyroxine free, Day 7Thyroxine free, Day 8Thyroxine free, Follow- up
GSK2248761 30 mg-0.0180.1080.0650.1170.072
Placebo0.0150.1900.1200.1300.135

[back to top]

Change From Baseline in Clinical Chemistry Paramaters- Uric Acid

The data for clinical chemistry parameters Uric acid, the change from baseline was reported. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 2, 4 , 7 , Day 8 and Follow-up (Day 14)

,
InterventionMicromole per liter (Mean)
Uric acid, Day 2Uric acid, Day 4Uric acid, Day 7Uric acid, Day 8Uric acid, Follow-up
GSK2248761 30 mg0.270.17-0.15-0.60-0.53
Placebo-0.55-0.70-0.80-1.05-0.80

[back to top]

Change From Baseline in Clinical Chemistry Paramaters-sodium, Potassium and Carbondioxide or Bicarbonate

The data for clinical chemistry parameters- sodium, potassium and carbon dioxide or bicarbonate, the change from baseline was reported. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 2, 4 , 7 , Day 8 and Follow-up (Day 14)

,
Interventionmilliequivalents per liter (Mean)
Sodium, Day 2Sodium, Day 4Sodium, Day 7Sodium, Day 8Sodium, Follow-upPotassium, Day 2Potassium, Day 4Potassium, Day 7Potassium, Day 8Potassium, Follow-upCarbondioxide, Day 2Carbondioxide, Day 4Carbondioxide, Day 7Carbondioxide, Day 8Carbondioxide, Follow-up
GSK2248761 30 mg-1.2-0.8-1.5-1.72.3-0.020.10-0.17-0.18-0.08-0.803.172.930.872.03
Placebo-0.51.00.0-0.51.0-0.250.10-0.10-0.00-0.30-1.352.703.601.65-0.30

[back to top]

Change From Baseline in Clinical Chemistry Parameters- Blood Urea Nitrogen, Triglycerides, Glucose, Creatinine, Calcium, Cholesterol, Total Bilirubin, and Direct Bilirubin.

The data for clinical chemistry parameters- Blood urea nitrogen, triglycerides, glucose, creatinine, calcium, cholesterol, total bilirubin, and direct bilirubin. The change from baseline was reported. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 2, 4 , 7 , Day 8 and Follow-up (Day 14)

,
Interventionmilligram per deciliter (Mean)
Blood urea nitrogen, Day 2Blood urea nitrogen, Day 4Blood urea nitrogen, Day 7Blood urea nitrogen, Day 8Blood urea nitrogen, Follow-upTriglycerides, Day 2Triglycerides, Day 4Triglycerides, Day 7Triglycerides, Day 8Triglycerides, Follow-upGlucose, Day 2Glucose, Day 4Glucose, Day 7Glucose, Day 8Glucose, Follow-upCreatinine, Day 2Creatinine, Day 4Creatinine, Day 7Creatinine, Day 8Creatinine, Follow-upCalcium, Day 2Calcium, Day 4Calcium, Day 7Calcium, Day 8Calcium, Follow-upCholesterol, Day 2Cholesterol, Day 4Cholesterol, Day 7Cholesterol, Day 8Cholesterol, Follow-upTotal bilirubin, Day 2Total bilirubin, Day 4Total bilirubin, Day 7Total bilirubin, Day 8Total bilirubin, Follow-upDirect bilirubin, Day 2Direct bilirubin, Day 4Direct bilirubin, Day 7Direct bilirubin, Day 8Direct bilirubin, Follow-up
GSK2248761 30 mg1.54.31.02.7-2.2-17.8-19.85.220.8-3.01.30.0-1.8-5.23.0-0.0480.025-0.003-0.060-0.003-0.25-0.13-0.40-0.470.08-11.8-15.3-12.0-8.3-5.5-0.12-0.08-0.03-0.080.08-0.08-0.08-0.05-0.050.02
Placebo-1.04.03.55.51.05.05.520.534.0114.06.0-0.5-1.0-5.010.00.0000.0250.035-0.0400.060-0.250.05-0.20-0.450.05-12.0-17.5-20.5-13.5-0.5-0.10-0.15-0.05-0.100.000.000.000.050.000.10

[back to top]

Change From Baseline in Hematology Paramaters- Hematocrit

The data for hematology parameter Hematocrit, the change from baseline was reported. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 2, 4 , 7 , Day 8 and Follow-up (Day 14)

,
Interventionpercentage of red blood cells (Mean)
Day 2Day 4Day 7Day 8Follow-up
GSK2248761 30 mg-1.22-0.92-0.80-1.45-2.78
Placebo-2.151.70-1.85-1.55-3.55

[back to top]

Change From Baseline in Hematology Paramaters- Hemoglobin

The data for hematology parameter hemoglobin from the change from baseline was reported. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 2, 4 , 7 , Day 8 and Follow-up (Day 14)

,
Interventiongram per decilitre (Mean)
Day 2Day 4Day 7Day 8Follow-up
GSK2248761 30 mg-0.40-0.28-0.25-0.48-0.93
Placebo-0.500.60-0.45-0.45-1.00

[back to top]

Change From Baseline in Hematology Paramaters- Mean Corpuscle Hemoglobin (MCH)

The data for hematology parameter MCH, the change from baseline was reported. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 2, 4 , 7 , Day 8 and Follow-up (Day 14)

,
Interventionpicogram (Mean)
Day 2Day 4Day 7Day 8Follow-up
GSK2248761 30 mg0.070.080.030.070.13
Placebo0.450.050.200.100.25

[back to top]

GSK2248761 PK Parameters Following Dose Administration on Day 1: Time to Maximum Observed Concentration (Tmax), Terminal Half-life (t1/2), Absorption Lag Time (Tlag)

Tmax is defined as the, time of maximum measured GSK2248761 concentration in the plasma, on Day 1. The t1/2 was defined as the time measured for plasma concentration to decrease by one half. The tlag was defined as the time taken for the drug GSK2248761, to appear in the systemic circulation following administration. Serial blood samples were collected on Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours postdose on Day 1 and used for analysis. (NCT00945282)
Timeframe: Day 1 (Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours postdose)

Interventionhour (Median)
tmaxt1/2tlag
GSK2248761 30 mg4.007.990.49

[back to top]

Change From Baseline in Hematology Paramaters- Mean Corpuscle Volume (MCV)

The change from baseline data for hematology parameter MCV, was reported. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 2, 4 , 7 , Day 8 and Follow-up (Day 14)

,
Interventionfemtoliters (Mean)
Day 2Day 4Day 7Day 8Follow-up
GSK2248761 30 mg0.270.15-0.100.130.47
Placebo-0.10-0.20-0.40-0.15-0.50

[back to top]

Change From Baseline in Hematology Paramaters- Platelet Count

The data for hematology parameter platelet count, for the change from baseline was reported. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 2, 4 , 7 , Day 8 and Follow-up (Day 14)

,
Interventionper cubic millimeter (Mean)
Day 2Day 4Day 7Day 8Follow-up
GSK2248761 30 mg-5.75.018.529.323.3
Placebo3.0-23.52.52.0-1.0

[back to top]

Change From Baseline in Hematology Paramaters- Red Blood Cell Count

The data for hematology parameter red blood cell count, for the change from baseline was reported. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 2, 4 , 7 , Day 8 and Follow-up (Day 14)

,
Interventionmillion cells per microliter (Mean)
Day 2Day 4Day 7Day 8Follow-up
GSK2248761 30 mg-0.153-0.112-0.083-0.170-0.343
Placebo-0.2400.200-0.190-0.170-0.375

[back to top]

Change From Baseline in Hematology Paramaters-Mean Corpuscle Hemoglobin Concentration

The data for hematology parameter Mean Corpuscle Hemoglobin concentration, the change from baseline was reported. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 2, 4 , 7 , Day 8 and Follow-up (Day 14)

,
Interventionpercentage of red blood cells (Mean)
Day 2Day 4Day 7Day 8Follow-up
GSK2248761 30 mg0.020.050.070.02-0.03
Placebo0.550.150.400.150.45

[back to top]

Change From Baseline in Hematology Parameters- Total Neutrophil

The data for hematology parameter total neutrophil count, for the change from baseline was reported. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 2, 4 , 7 , Day 8 and Follow-up (Day 14)

,
Interventiongiga cells per liter (Mean)
Day 2Day 4Day 7Day 8Follow-up
GSK2248761 30 mg243.3508.3-30.0296.776.7
Placebo980.0-375.0-745.0-705.0105.0

[back to top]

Change From Baseline in HR

Vital sign measurements for HR after sitting for 5 minutes were measured. The average mean values were measured. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 1 (4-hour), Day 4 (Pre-dose and 4 hour), Day 7 (pre-dose and 4-hour), Day 8 and follow up (Day 14)

,
InterventionBeats per minute (Mean)
HR , Day 1, 4 hourHR, Day 4, Pre-doseHR, Day 4, 4 hourHR, Day 7, Pre-doseHR, Day 7, 4 hourHR, Day 8HR, Follow-up
GSK2248761 30 mg-2.5-2.7-2.23.0-0.33.7-0.3
Placebo-3.0-6.0-6.0-1.0-2.50.05.0

[back to top]

Change From Baseline in Vital Signs-systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)

Vital sign measurements for SBP and DBP after sitting for 5 minutes were measured. The change from baseline was calculated by subtracting the baseline values from the individual post-randomization values. Baseline (pre-dose [Screening, Day -1 or Day 1]) was defined as last available scheduled assessment prior to time of the first dose unless it is specified otherwise. (NCT00945282)
Timeframe: Baseline (pre-dose at Day -1 or Day 1) and Day 1, 4, 7 , Day 8 and Follow-up (Day 14)

,
Interventionmillimeters of mercury (Mean)
SBP, Day 1, 4 hourSBP, Day 4, Pre-doseSBP, Day 4, 4 hourSBP, Day 7, Pre-doseSBP, Day 7, 4 hourSBP, Day 8SBP, Follow-upDBP, Day 1, 4 hourDBP, Day 4, Pre-doseDBP, Day 4, 4 hourDBP, Day 7, Pre-doseDBP, Day 7, 4 hourDBP, Day 8DBP, Follow-up
GSK2248761 30 mg4.3-0.73.06.29.88.55.88.25.89.78.26.05.24.5
Placebo-4.50.512.56.514.513.514.51.01.03.07.08.015.08.0

[back to top]

GSK2248761 Pharmacokinetic (PK) Parameters Following Dose Administration on Day 1: Area Under the Plasma Concentration Time Curve 0 to Infinite (AUC[0-∞]) and Area Under the Plasma Concentration Time Curve (AUC [0-24])

AUC (0-24), measured the plasma concentration of GSK2248761 against time, from time zero (pre-dose) to 24 hrs post-dose AUC (0-24) and from time zero to extrapolated infinite time AUC (0-∞). Serial blood samples were collected on Predose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours postdose on Day 1 and used for analysis. (NCT00945282)
Timeframe: Day 1 (Predose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours post-dose)

Interventionhours*nanograms (ng)/mL (Geometric Mean)
AUC(0-inf)AUC(0-24)
GSK2248761 30 mg2217.731842.19

[back to top]

GSK2248761 PK Parameters Following Dose Administration on Day 1: Maximum Observed Concentration (Cmax) and Concentration at 24 Hours Post Dose (C24)

Cmax represents the maximum concentration of GSK2248761 in the plasma. C24 is defined as the measure of plasma drug concentration of GSK2248761, 24 hours post dose, determined on Day 1. Serial blood samples were collected on Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours postdose on Day 1 and used for analysis.Data for dose normalized Cmax and C24 was reported. (NCT00945282)
Timeframe: Day 1 (Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours)

Interventionng/mL (Geometric Mean)
CmaxC24
GSK2248761 30 mg585.43103.40

[back to top]

GSK2248761 PK Parameters Following Dose Administration on Day 7: t1/2

The t1/2 was defined as the time measured for plasma concentration to decrease by one half. Serial blood samples were collected on Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours postdose on Day 7 and used for analysis (NCT00945282)
Timeframe: Day 7 (Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours post dose)

Interventionhour (Geometric Mean)
GSK2248761 30 mg9.69

[back to top]

GSK2248761 PK Parameters Following Dose Administration on Day 7: Predose Concentration (C0), Concentration at End of Dosing Interval (Cτ), Minimum Observed Concentration During One Dosing Interval (Cmin) and Cmax

The C0 was defined as the concentration of drug in plasma, before dose administration on Day 7. Cτ, was defined as the concentration of drug in the plasma at the end of dosing interval. The Cmin was defined as the minimum concentration of the drug in plasma during one dosing interval on Day 7. Cmax represents the maximum concentration of GSK2248761 in the plasma on Day 7. Serial blood samples were collected on Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours postdose on Day 7 and used for analysis. (NCT00945282)
Timeframe: Day 7 (Pre -dose, 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours post-dose)

Interventionng/mL (Geometric Mean)
C0, Day 7Cτ, Day 7Cmin, Day 7Cmax, Day 7
GSK2248761 30 mg57.4754.2746.37212.93

[back to top]

Number of Participants With Abnormal Electrocardiogram (ECG) Findings

Triplicate 12-lead ECGs were collected at different timepoints, after participants were supine for 5 minutes, during the study using an ECG machine that automatically calculated the heart rate (HR) and measures PR, QRS, QT, and QTc intervals. The three consecutive determinations were collected 5 plus or minus 2 minutes apart and all three tracings were recorded. The participants with abnormal values categorized as abnormal clinically significant (CS) and not clinically significant (NCS) were reported. (NCT00945282)
Timeframe: Day 1, Day 4, Day 7, Day 8 and follow-up

,
InterventionParticipants (Count of Participants)
Day 1, Pre-dose, NCSDay 1, 4 hour, NCSDay 1, 8 hour, NCSDay 4, Pre-dose, NCSDay 4, 4 hour, NCSDay 4, 8 hour, NCSDay 7, Pre-dose, NCSDay 7, 4 hour, NCSDay 7, 8 hour, NCSDay 8, Pre-dose, NCSFollow-up, NCS
GSK2248761 30 mg32222333233
Placebo00000000000

[back to top]

Number of Participants With Serious Adverse Events (SAEs) and Adverse Events (AEs)

An AE is defined as any untoward medical occurrence in a clinical investigation participant, temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product. An AE can therefore be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease (new or exacerbated) temporally associated with the use of a medicinal product. An SAE is defined as any untoward medical occurrence that, at any dose, results in death, is life threatening, requires hospitalization or prolongation of existing hospitalization, results in disability/incapacity, or is a congenital anomaly/birth defect, or is an important medical events that jeopardize the participants or may require medical or surgical intervention to prevent one of the other outcomes listed in the above definition, or a drug-induced liver injury. (NCT00945282)
Timeframe: Up to 38 days

,
InterventionParticipants (Count of Participants)
Any AEAny SAE
GSK2248761 30 mg40
Placebo10

[back to top]

PK Data of Cmax and Ctau at Different Doses for the Assessment of Dose Proportionality

Data for IDX899 100 mg, IDX899 200 mg, IDX899 400 mg and IDX899 800 mg for Day 1 and Day 2 were taken from the Idenix NV-05A-002 study which were combined with GSK2248761 30 mg once daily data from this study, to assess the dose proportionality. The dose proportionality occurred when increase in the administered doses were accompanied by proportional increases in measure of exposure of the drug in the plasma PK parameters like AUC, Cmax, Ctau and other factors. Data for Ctau on Day 1 is presented for concentration at 24 hours post-dose on Day 1. (NCT00945282)
Timeframe: (Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours post-dose ) From Day 1 to Up to Day 7

,,,,
Interventionng/mL (Geometric Mean)
Cmax, Day 1Cmax, Day 7Ctau, Day 1Ctau, Day 7
GSK2248761 30 mg175.63212.9331.0254.27
IDX899 100 mg797.8960.1128.9204.7
IDX899 200 mg1686.22158.9325.6469.2
IDX899 400 mg2625.94140.7422.9864.5
IDX899 800 mg3406.45394.5364.5540.3

[back to top]

Percent Change From Baseline in CD4+ and CD8+ T-lymphocyte Cell Count at Day 1 and Day 8

Data for CD4+ and CD8+ cells was collected at Screening, Day 1 and Day 8. The percent change from baseline was reported at Day 1 and Day 8. Baseline was defined as Screening. The percent change from baseline was calculated as post-randomization value minus the baseline value. (NCT00945282)
Timeframe: Baseline (Screening), Day 1 and Day 8

,
InterventionPercent change (Mean)
CD4+, Day 1CD4+, Day 8CD8+, Day 1CD8+, Day 8
GSK2248761 30 mg-3.2-2.4-2.8-0.4
Placebo-2.9-1.72.80.4

[back to top]

PK Data of Day 1 AUC(0-inf) and Day 7 AUC(0-tau) at Different Doses for the Assessment of Dose Proportionality

Data for IDX899 100 mg, IDX899 200 mg, IDX899 400 mg and IDX899 800 mg for Day 1 and Day 2 were taken from the Idenix NV-05A-002 study which were combined with GSK2248761 30 mg once daily data from this study, to assess the dose proportionality. The dose proportionality occurred when increase in the administered doses were accompanied by proportional increases in measure of exposure of the drug in the plasma PK parameters like AUC, Cmax, Ctau and other factors. The dose proportionality effects of IDX899 100 mg, IDX899 200 mg, IDX899 400 mg and IDX899 800 mg, following repeat dose administration on Day 7 for the PK parameter AUC(0-tau) has been reported. (NCT00945282)
Timeframe: (Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours post-dose ) From Day 1 to Up to Day 7

,,,,
Interventionhour*ng/mL (Geometric Mean)
AUC(0-inf), Day 1AUC(0-tau), Day 7
GSK2248761 30 mg2217.732903.91
IDX899 100 mg990811650
IDX899 200 mg2381727209
IDX899 400 mg3382049649
IDX899 800 mg3781253683

[back to top]

Virologic Response

Virologic response is defined as HIV viral load of < 50 copies/mL before week 48 and without subsequent rebound or change of ARV therapy prior to week 48. A rebound is defined by two consecutive measurements of VL >= 50 copies/ml, at least two weeks apart, after two consecutive measurements of VL< 50 copies/ml. Because of many missing data concerning the viral load, the virologic response could be determined only for four patients. (NCT00976950)
Timeframe: 48 weeks

InterventionNumber of participants (Number)
Virologic response - YesVirologic response - NoVirologic response - Missing
Aptivus and Ritonavir0438

[back to top]

Change in CD4+ Cell Count From Baseline at Week 48

(NCT00976950)
Timeframe: 48 weeks

Interventioncells/mm^3 (Mean)
Aptivus and Ritonavir85.8

[back to top]

Number of Patients Reporting Adverse Events (AE)

Any type of adverse events (NCT00976950)
Timeframe: 48 weeks

InterventionParticipants (Number)
Patients with any adverse eventsPatients with any serious adverse eventsPatients with AE leading to discontinuationPatients with hepatic adverse eventsPatients with dyslipidemia
Aptivus and Ritonavir61412

[back to top]

Percentage of Uncomplicated Malaria Episodes With Accompanying Adverse Events That Occurred in the 28 Days Following Antimalarial Therapy

The rates of adverse events, defined as severity grade 2 or higher that are possibly, probably or definitely related to study drugs over the course of the 28-day period after antimalarial therapy with artemether-lumefantrine (AL). (NCT00978068)
Timeframe: 28 days after antimalarial therapy

Intervention% uncomplicated malaria episodes w/ AEs (Number)
LPV/r + 2 NRTIs71.0
NVP or EFV + 2 NRTIs79.3

[back to top]

Incidence-density of Malaria Defined as the Number of Incident Episodes of Malaria Per Time at Risk.

(NCT00978068)
Timeframe: Time from randomization to at least 24 months of follow up or until end of the study

InterventionEpisodes/ Person-Yr at Risk (Number)
Group 1: LPV/r + 2 NRTIs1.32
Group 2: Nevirapine (NVP) or Efavirenz (EFV) + 2 NRTIs2.25

[back to top]

Incidence-density of Malaria Defined as the Number of Incident Episodes of Complicated Malaria Per Time at Risk.

(NCT00978068)
Timeframe: Time from randomization to at least 24 months of follow up or until end of the study

InterventionEpisodes/ Person-Yr at Risk (Number)
Group 10.024
Group 20.026

[back to top]

Estimates of the 6-month Risk of a First Episode of Malaria

To assess the effect of ART independently of potential interactions with antimalarial therapy after treatment for malaria, we compared the two groups with respect to the time to the first episode of malaria. Cumulative risk was estimated using the Kaplan-Meier product-limit formula. (NCT00978068)
Timeframe: Enrollment to 6 months follow up

InterventionCumulative Risk Percentage (Number)
LPV/r + 2 NRTIs40.7
NVP or EFV + 2 NRTIs52.5

[back to top]

63-day Risk of Recurrent Malaria

To assess the effect of potential interactions between ART and artemether-lumefantrine, the risks of recurrent malaria at 63 days were compared between the two groups. (NCT00978068)
Timeframe: 28 days after antimalarial therapy

InterventionCumulative Risk Percentage (Number)
LPV/r + 2 NRTIs28.1
NVP or EFV + 2 NRTIs54.2

[back to top]

28-day Risk of Recurrent Parasitemia

To assess the effect of potential interactions between ART and artemether-lumefantrine, the risks of recurrent parasitemia at 28 days were compared between the two groups. (NCT00978068)
Timeframe: 28 days after antimalarial therapy

InterventionCummulative Risk Percentage (Number)
LPV/r + 2 NRTIs14.0
NVP or EFV + 2 NRTIs40.8

[back to top]

Maximum Plasma Concentration (Cmax)

The maximum or peak concentration that colchicine reaches in the plasma. (NCT00983515)
Timeframe: serial pharmacokinetic blood samples drawn within 1 hour prior to colchicine dosing (0 hour) on Days 1 and 19, and then 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, 24, 36, 48, 72, and 96 hours after colchicine dose administration

Interventionpg/mL (Mean)
Colchicine Alone1,869.50
Colchicine With Ritonavir (at Steady-state)4,993.06

[back to top]

Area Under the Concentration Versus Time Curve From Time 0 to Time t [AUC(0-t)]

The area under the plasma concentration versus time curve, from time 0 to the time of the last measurable colchicine concentration (t), as calculated by the linear trapezoidal rule. (NCT00983515)
Timeframe: serial pharmacokinetic blood samples drawn within 1 hour prior to colchicine dosing (0 hour) on Days 1 and 19, and then 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, 24, 36, 48, 72, and 96 hours after colchicine dose administration

Interventionpg-hr/mL (Mean)
Colchicine Alone8,412.04
Colchicine With Ritonavir (at Steady-state)29,048.45

[back to top]

Area Under the Concentration Versus Time Curve From Time 0 Extrapolated to Infinity [AUC(0-∞)]

The area under the plasma concentration versus time curve from time 0 to infinity. AUC(0-∞) was calculated as the sum of AUC(0-t) plus the ratio of the last measurable colchicine plasma concentration to the elimination rate constant. (NCT00983515)
Timeframe: serial pharmacokinetic blood samples drawn within 1 hour prior to colchicine dosing (0 hour) on Days 1 and 19, and then 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, 24, 36, 48, 72, and 96 hours after colchicine dose administration

Interventionpg-hr/mL (Mean)
Colchicine Alone10,410.73
Colchicine With Ritonavir (at Steady-state)35,276.22

[back to top]

Adverse Events

Number of reported adverse events, severity of adverse events and relationship to study drug was assessed by questions, physical examination and laboratory parameters. Adverse event data was used to assess the safety and tolerability of low lopinavir/ritonavir doses. (NCT00985543)
Timeframe: Up to 11 weeks from screening to final study visit

Interventionnumber of adverse events (Number)
LPV/r 400/100 mg27
LPV/r 200/150 mg2
LPV/r 200/50 mg4

[back to top]

Plasma Lopinavir/Ritonavir Concentrations as Measured by the Area Under the Curve (AUC 0-12h).

Pharmacokinetics of plasma lopinavir/ritonavir over a 12-hour dosing interval following administration of lopinavir/ritonavir 400/100mg, 200/150mg and 200/50mg twice daily. (NCT00985543)
Timeframe: at the end of each 7-day dosing phase

Interventionng.h/mL (Geometric Mean)
LPV/r 400/100 mg99599
LPV/r 200/150 mg73603
LPV/r 200/50 mg45146

[back to top]

Placental Malaria Defined as Positive Placental RDT

Number of participants with positive placental RDT for malaria. Malaria rapid diagnostic tests (RDTs) assist in the diagnosis of malaria by detecting evidence of malaria parasites (antigens) in human blood. RDTs permit a reliable detection of malaria infections particularly in remote areas with limited access to good quality microscopy services. (NCT00993031)
Timeframe: Delivery

Interventionparticipants (Number)
With Protease Inhibitor6
Without Protease Inhibitor7

[back to top]

Incidence of Pre-eclampsia Defined by Hypertension > 140/90 on Two Occasions Measured > 6 Hours Apart With ≥1+ Proteinuria on Clean Catch Urine Dipstick

Pre-eclampsia Defined by Hypertension > 140/90 on Two Occasions Measured > 6 Hours Apart With ≥1+ Proteinuria on Clean Catch Urine Dipstick (NCT00993031)
Timeframe: Time from randomization until delivery

InterventionParticipants (Count of Participants)
Without Protease Inhibitor0
With Protease Inhibitor0

[back to top]

ART Levels in Hair Samples at Delivery

antiretroviral hair concentrations (per doubling) (NCT00993031)
Timeframe: delivery

Interventionantiretroviral hair concentration(ng/mg) (Mean)
Without Protease Inhibitor5.7
With Protease Inhibitor6.6

[back to top]

Maternal Malaria Defined as the Number of Treatments for New Episodes of Malaria Per Time at Risk After Pregnancy

(NCT00993031)
Timeframe: Number of treatments given for clinical malaria based on postive blood smear from time from delivery until 24 months after delivery or cessation of breastfeeding

Interventiontreatments (Number)
Group A21
Group B13

[back to top]

Maternal Malaria Defined as the Number of Treatments for New Episodes of Malaria Per Time at Risk During Pregnancy

(NCT00993031)
Timeframe: Number of treatments given for clinical malaria based on postive blood smear from time from randomization until 24 months after delivery or cessation of breastfeeding

Interventiontreatments (Number)
With Protease Inhibitor17
Without Protease Inhibitor17

[back to top]

Number of Participants With Maternal HIV RNA Suppression of <400 Copies/mL

Virologic suppression was defined as plasma HIV-1 RNA 400 copies/ml or less based on the lower limit of detection of the available test. (NCT00993031)
Timeframe: Time from randomization until delivery, an average of 20 weeks

InterventionParticipants (Count of Participants)
Without Protease Inhibitor166
With Protease Inhibitor153

[back to top]

Number of Participants With Maternal to Child Transmission of HIV, Measured by Infant HIV DNA PCR

HIV tested by DNA PCR (NCT00993031)
Timeframe: Delivery to 48 weeks postpartum

InterventionParticipants (Count of Participants)
Without Protease Inhibitor0
With Protease Inhibitor2

[back to top]

Number of Participants With Severe Maternal Anemia Defined by Hemoglobin < 8g/dl at Any Point During the Trial in Each Treatment Group

Proportion of women with severe maternal Anemia (hemoglobin < 8g/dl by hemacue or CBC) at any point during the trial in Each Treatment Group (NCT00993031)
Timeframe: Time from randomization until one year follow up

InterventionParticipants (Count of Participants)
Without Protease Inhibitor11
With Protease Inhibitor11

[back to top]

Prevalence of Composite Clinical Outcome Defined by LBW, Stillbirth(Intrauterine Fetal Demise >20wks GA), Late Spontaneous Abortion(Miscarriage 12-20wks GA), Preterm Delivery(<37wks Gestation), Neonatal Death(Death of Liveborn Infant Within First 28days)

Percent of evaluated participants with composite clinical outcome defined by LBW, stillbirth (intrauterine fetal demise >20wks GA), late spontaneous abortion(miscarriage 12-20wks GA), preterm delivery(<37wks gestation), neonatal death(death of live-born infant within first 28 days) (NCT00993031)
Timeframe: Time from randomization until 24 months postpartum or cessation of breastfeeding

Intervention% of evaluated participants with outcome (Number)
With Protease Inhibitor33.9
Without Protease Inhibitor27.8

[back to top]

Prevalence of Malaria Defined as Positive Placental Blood PCR

Number of participants with positive placental blood PCR for malaria (NCT00993031)
Timeframe: Delivery

Interventionparticipants (Number)
With Protease Inhibitor6
Without Protease Inhibitor7

[back to top]

Number of Participants With Grade 3 or 4 Toxicity in the Two Treatment Groups in Women

(NCT00993031)
Timeframe: Randomization to one month postpartum

InterventionParticipants (Count of Participants)
Without Protease Inhibitor12
With Protease Inhibitor8

[back to top]

Placental Malaria Defined Placental Histopathologic Analysis

Number of participants with positive placental histopathology slide for malaria (NCT00993031)
Timeframe: Delivery

Interventionparticipants (Number)
With Protease Inhibitor62
Without Protease Inhibitor47

[back to top]

Change in Maternal CD4 Cell Counts

CD4 cell count recovery efavirenz at delivery (NCT00993031)
Timeframe: Time of randomization to delivery, an average of 20 weeks

InterventionCD4 cell count (Median)
Without Protease Inhibitor-7
With Protease Inhibitor57

[back to top]

Prevalence of Malaria Defined as Positive Placental Blood Smear

Number of participants with positive placental blood smear for malaria (NCT00993031)
Timeframe: Delivery

Interventionparticipants (Number)
With Protease Inhibitor5
Without Protease Inhibitor6

[back to top]

Drug Adherence, Number of Participants With Missed Doses

Drug adherence, assessed as number of participants with missed doses over four-day recall (NCT00993148)
Timeframe: Week 24

Interventionparticipants (Number)
Maraviroc + Darunavir/Ritonavir0

[back to top]

Percentage of Participants With Plasma HIV-1 RNA >50 Copies/mL

Percentage of participants with confirmed plasma HIV-1 RNA level >50 copies/mL (NCT00993148)
Timeframe: 48 weeks

Interventionpercentage of participants (Number)
Maraviroc + Darunavir/Ritonavir8.3

[back to top]

Signs/Symptoms or Laboratory Toxicities of Grade 3 or Higher

Signs/symptoms or laboratory toxicities of Grade 3 or higher, or of any grade which led to a permanent change or discontinuation of study treatment regimen (NCT00993148)
Timeframe: 96 weeks

Interventionparticipants (Number)
Maraviroc + Darunavir/Ritonavir1

[back to top]

Median CD4 Count Change From Baseline

Median changes from baseline in peripheral CD4+ T-cell count (NCT00993148)
Timeframe: 96 weeks

Interventioncells per mm^3 (Median)
Maraviroc + Darunavir/Ritonavir247

[back to top]

Trough Concentrations (Ctrough) of Maraviroc

Average trough concentration (Ctrough) of maraviroc (NCT00993148)
Timeframe: 24 hours

Interventionng/mL (Mean)
Maraviroc + Darunavir/Ritonavir39.3

[back to top]

Proportion of Participants With Plasma HIV-1 RNA >50 Copies/mL

Proportion of participants with confirmed plasma HIV-1 RNA level >50 copies/mL (NCT00993148)
Timeframe: 96 weeks

Interventionpercentage of participants (Number)
Maraviroc + Darunavir/Ritonavir10

[back to top]

Drug Resistance Mutations and Co-receptor Tropism Assessed by Trofile ES

(NCT00993148)
Timeframe: At study entry and at the time of virologic failure

Interventionparticipants (Number)
Maraviroc + Darunavir/Ritonavir0

[back to top]

Percentage of Participants With Virologic Failure or Off Study Treatment Regimen

Percentage of participants with virologic failure (confirmed plasma HIV-1 RNA > 50 copies/mL) or off study treatment regimen (composite end point) (NCT00993148)
Timeframe: 24 weeks

Interventionpercentage of participants (Number)
Maraviroc + Darunavir/Ritonavir12.5

[back to top]

Percentage of Participants With Plasma HIV-1 RNA >50

Percentage of participants with confirmed plasma HIV-1 RNA > 50 copies/mL (NCT00993148)
Timeframe: 24 weeks

Interventionpercentage of participants (Number)
Maraviroc + Darunavir/Ritonavir12.5

[back to top] [back to top]

AUC (0-24) of Ritonavir Following Q12H Dosing

Blood samples were collected at indicated time points to assess AUC (0-24) of ritonavir following Q12H dosing. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 8. (NCT01009814)
Timeframe: Day 8: pre-morning dose, 1,2,3,4,5,6,8,12, 13,14,15,16,17,18,20 hours

InterventionHour*micrograms per milliliter (Geometric Mean)
BMS-663068 600 mg Q12H + RTV 100 mg Q12H18.9
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H19.5
BMS-663068 1200 mg Q12H + RTV 100 mg QAM6.76

[back to top]

AUC (0-24) of Ritonavir Following QHS Dosing

Blood samples were collected at indicated time points to assess AUC (0-24) of ritonavir following QHS dosing. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 8. (NCT01009814)
Timeframe: Day 8: pre-evening dose, 1,2,3,4,5,6,8 hours

InterventionHour*micrograms per milliliter (Geometric Mean)
BMS-663068 1200 mg QHS + RTV 100 mg QHS6.13

[back to top]

Inhibitory Quotient of BMS-626529 by Ctrough Following Q12H Dosing

Blood samples were collected at indicated time points to assess IQ of BMS-626529. Inhibitory quotient was calculated as the ratio of BMS-626529 in vivo exposure to in vitro measured protein binding adjusted EC90 (PBA-EC90). The following in vivo exposure measure was used in evaluating IQ: Ctrough. Geometric mean and geometric coefficient of variation are presented. (NCT01009814)
Timeframe: Days 1, 8: pre-morning dose, 1,2,3,4,5,6,8,12 hours; Day 8: 13,14,15,16,17,18,20 hours; Day 9: pre-dose, 4,12 hours; Day 10: pre-dose, 12 hours; Day 11: pre-dose; Days 5,6,7: pre-morning dose

InterventionRatio (Geometric Mean)
BMS-663068 600 mg Q12H + RTV 100 mg Q12H12.2
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H120
BMS-663068 1200 mg Q12H + RTV 100 mg QAM87.3
BMS-663068 1200 mg Q12H4.61

[back to top]

Inhibitory Quotient of BMS-626529 by Ctrough Following QHS Dosing

Blood samples were collected at indicated time points to assess IQ of BMS-626529. Inhibitory quotient was calculated as the ratio of BMS-626529 in vivo exposure to in vitro measured protein binding adjusted EC90 (PBA-EC90). The following in vivo exposure measure was used in evaluating IQ: Ctrough. Geometric mean and geometric coefficient of variation are presented. (NCT01009814)
Timeframe: Days 1, 8: pre-evening dose, 1,2,3,4,5,6,8 hours; Days 2: pre-evening dose, 12,16 hours; Day 9: pre-dose, 12,16 hours; Day 10: pre-dose, 12 hours; Day 11: pre-dose; Days 5,6,7: pre-evening dose

InterventionRatio (Geometric Mean)
BMS-663068 1200 mg QHS + RTV 100 mg QHS4.77

[back to top]

Mean Logarithm With Base 10 (Log10) Change From Baseline in Human Immunodeficiency Virus (HIV) Ribonucleic Acid (RNA) at Day 9

The primary assessment of the antiviral activity of BMS-663068 was assessed on the log10 change from Baseline in HIV RNA to Day 9. Baseline was the last non-missing observation before first dose (Day 1 pre-dose) and change from Baseline was calculated by subtracting Baseline value from post-Baseline visit value. An analysis of covariance (ANCOVA) model correcting for Baseline HIV viral load and treatment group was used to test the differences in mean log10 decrease in HIV RNA at Day 9 between 2 regimen groups by antiretroviral treatment history (ARV [antiretroviral] naive, ARV experienced, and combined [ARV naive + ARV experienced]). For the combined group (ARV naive +ARV experienced) an additional ANCOVA was used correcting also for treatment history as an additional covariate. Only Clade B participants were included in the population. (NCT01009814)
Timeframe: Baseline and Day 9

InterventionLog10 copies per milliliter (c/mL) (Mean)
BMS-663068 600 mg Q12H + RTV 100 mg Q12H-1.3445
BMS-663068 1200 mg QHS + RTV 100 mg QHS-1.2532
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H-1.2381
BMS-663068 1200 mg Q12H + RTV 100 mg QAM-1.1888
BMS-663068 1200 mg Q12H-0.8760

[back to top]

Number of Participants With Any Abnormality in Physical Examination

A complete physical examination included, at a minimum, assessment of the Cardiovascular, Respiratory, Gastrointestinal and Neurological systems. A brief physical examination included, at a minimum assessments of the skin, lungs, cardiovascular system, and abdomen (liver and spleen). Any abnormality found by investigator during physical examination were recorded. Number of participants with any abnormality in physical examination during study have been reported. (NCT01009814)
Timeframe: Up to 50 days

InterventionParticipants (Count of Participants)
BMS-663068 600 mg Q12H + RTV 100 mg Q12H1
BMS-663068 1200 mg QHS + RTV 100 mg QHS0
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H2
BMS-663068 1200 mg Q12H + RTV 100 mg QAM1
BMS-663068 1200 mg Q12H1

[back to top]

Number of Participants With Clinically Significant Abnormalities in Laboratory Parameters

Laboratory parameters included hematology, clinical chemistry and urine parameters. Clinically significant abnormal laboratory findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. Any abnormal laboratory test results which were considered clinically significant by the investigator were recorded on the case report form. Number of participants with any clinically significant abnormalities in laboratory parameters have been presented. (NCT01009814)
Timeframe: Up to 50 days

InterventionParticipants (Count of Participants)
BMS-663068 600 mg Q12H + RTV 100 mg Q12H0
BMS-663068 1200 mg QHS + RTV 100 mg QHS0
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H0
BMS-663068 1200 mg Q12H + RTV 100 mg QAM0
BMS-663068 1200 mg Q12H0

[back to top]

Area Under the Concentration-time Curve in One Dosing Interval (AUC [Tau]) of BMS-626529 Following Q12H Dosing

Blood samples were collected at indicated time points to assess AUC (tau) of BMS-626529 following Q12H dosing. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 1, Day 8 morning dose (AM) and Day 8 evening dose (PM). (NCT01009814)
Timeframe: Days 1, 8: pre-morning dose, 1,2,3,4,5,6,8,12 hours; Day 8: 13,14,15,16,17,18,20 hours

,,,
InterventionHour*micrograms per milliliter (Geometric Mean)
Day 1, n=10,10,10,10Day 8, AM, n=9,10,10,10Day 8, PM, n=9,10,10,10
BMS-663068 1200 mg Q12H13.819.522.5
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H18.329.930.6
BMS-663068 1200 mg Q12H + RTV 100 mg QAM20.627.627.0
BMS-663068 600 mg Q12H + RTV 100 mg Q12H9.1613.113.1

[back to top]

AUC (Tau) of BMS-626529 Following QHS Dosing

Blood samples were collected at indicated time points to assess AUC (tau) of BMS-626529 following QHS dosing. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 1 and Day 8 evening dose (PM). (NCT01009814)
Timeframe: Days 1, 8: pre-evening dose, 1,2,3,4,5,6,8 hours

InterventionHour*micrograms per milliliter (Geometric Mean)
Day 1, n=10Day 8 PM, n=9
BMS-663068 1200 mg QHS + RTV 100 mg QHS25.523.0

[back to top]

AUC (Tau) of Ritonavir Following Q12H Dosing

Blood samples were collected at indicated time points to assess AUC (tau) of ritonavir following Q12H dosing. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 1, Day 8 morning dose (AM) and Day 8 evening dose (PM). (NCT01009814)
Timeframe: Days 1, 8: pre-morning dose, 1,2,3,4,5,6,8,12 hours; Day 8: 13,14,15,16,17,18,20 hours

InterventionHour*micrograms per milliliter (Geometric Mean)
Day 8, AM, n=9,10,10
BMS-663068 1200 mg Q12H + RTV 100 mg QAM6.76

[back to top]

AUC (Tau) of Ritonavir Following Q12H Dosing

Blood samples were collected at indicated time points to assess AUC (tau) of ritonavir following Q12H dosing. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 1, Day 8 morning dose (AM) and Day 8 evening dose (PM). (NCT01009814)
Timeframe: Days 1, 8: pre-morning dose, 1,2,3,4,5,6,8,12 hours; Day 8: 13,14,15,16,17,18,20 hours

,
InterventionHour*micrograms per milliliter (Geometric Mean)
Day 1, n=10,10,0Day 8, AM, n=9,10,10Day 8, PM, n=9,10,0
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H2.059.0510.4
BMS-663068 600 mg Q12H + RTV 100 mg Q12H2.379.439.23

[back to top]

AUC (Tau) of Ritonavir Following QHS Dosing

Blood samples were collected at indicated time points to assess AUC (tau) of ritonavir following QHS dosing. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 1 and Day 8 evening dose (PM). (NCT01009814)
Timeframe: Days 1, 8: pre-evening dose, 1,2,3,4,5,6,8 hours

InterventionHour*micrograms per milliliter (Geometric Mean)
Day 1, n=10Day 8 PM, n=9
BMS-663068 1200 mg QHS + RTV 100 mg QHS2.916.13

[back to top]

Change From Baseline in Cluster of Differentiation 4 Positive (CD4+) Cell Count and Cluster of Differentiation 8 Positive (CD8+) Cell Count

Blood samples were collected for evaluation of CD4+ and CD8+ cells at Baseline (Day 1, pre-dose), Day 8, Day 15 (Follow up) and Day 50 (study discharge). Baseline was the last non-missing observation before first dose (Day 1 pre-dose) and change from Baseline was calculated by subtracting Baseline value from post-Baseline visit value. (NCT01009814)
Timeframe: Baseline (Day 1 pre-dose), Day 8, Day 15 and Day 50

,,,,
InterventionCells per microliter (cells/μL) (Mean)
CD4+, Day 8, n=9, 9, 10, 10, 10CD4+, Day 15, n=9, 9, 10, 10, 10CD4+, Day 50, n=9, 9, 10, 10, 9CD8+, Day 8, n=9, 9, 10, 10, 10CD8+, Day 15, n=9, 9, 10, 10, 10CD8+, Day 50, n=9, 9, 10, 10, 9
BMS-663068 1200 mg Q12H32.010.4-19.294.6-4.4-48.2
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H109.8117.671.1218.485.6139.7
BMS-663068 1200 mg Q12H + RTV 100 mg QAM38.544.6-48.4114.9-99.66.0
BMS-663068 1200 mg QHS + RTV 100 mg QHS93.4-6.1-50.3189.0-153.2-292.4
BMS-663068 600 mg Q12H + RTV 100 mg Q12H114.258.422.0384.264.8-37.6

[back to top]

Change From Baseline in Percent CD4+ Cell Count and Percent CD8+ Cell Count

Blood samples were collected for evaluation of percent CD4+ and percent CD8+ cells at Baseline (Day 1, pre-dose), Day 8, Day 15 (Follow up) and Day 50 (study discharge). Baseline was the last non-missing observation before first dose (Day 1 pre-dose) and change from Baseline was calculated by subtracting Baseline visit value from post-Baseline visit value. (NCT01009814)
Timeframe: Baseline (Day 1 pre-dose), Day 8, Day 15 and Day 50

,,,,
InterventionPercent cells per microliter (Mean)
Percent CD4+, Day 8, n=9, 9, 10, 10, 10Percent CD4+, Day 15, n=9, 9, 10, 10, 10Percent CD4+, Day 50, n=9, 9, 10, 10, 9Percent CD8+, Day 8, n=9, 9, 10, 10, 10Percent CD8+, Day 15, n=9, 9, 10, 10, 10Percent CD8+, Day 50, n=9, 9, 10, 10, 9
BMS-663068 1200 mg Q12H1.40.90.6-0.2-2.9-2.4
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H0.61.40.30.1-2.9-0.9
BMS-663068 1200 mg Q12H + RTV 100 mg QAM0.12.4-1.7-0.3-4.01.6
BMS-663068 1200 mg QHS + RTV 100 mg QHS1.21.21.1-0.4-2.8-0.7
BMS-663068 600 mg Q12H + RTV 100 mg Q12H-1.01.61.41.0-3.4-2.2

[back to top]

Cmax of BMS-626529 Following QHS Dosing

Blood samples were collected at indicated time points to assess Cmax of BMS-626529 following QHS dosing. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 1, Day 8 evening dose (PM) and Day 8 morning (AM) + evening dose (PM). (NCT01009814)
Timeframe: Days 1, 8: pre-evening dose, 1,2,3,4,5,6,8 hours

Interventionμg/mL (Geometric Mean)
Day 1, n=10Day 8, PM, n=9Day 8, AM+PM, n=9
BMS-663068 1200 mg QHS + RTV 100 mg QHS3.853.473.47

[back to top]

Cmax of Ritonavir Following Q12H Dosing

Blood samples were collected at indicated time points to assess Cmax of ritonavir following Q12H dosing. Geometric mean and geometric coefficient of variation are presented for Day 1, Day 8 morning dose (AM), Day 8 evening dose (PM) and Day 8 morning + evening dose (AM+PM). Pharmacokinetic parameter values were derived by non-compartmental methods. PK Population was used which comprised of all participants who receive ritonavir and provided pharmacokinetic samples. (NCT01009814)
Timeframe: Days 1, 8: pre-morning dose, 1,2,3,4,5,6,8,12 hours; Day 8: 13,14,15,16,17,18,20 hours

InterventionMicrograms per milliliter (μg/mL) (Geometric Mean)
Day 1, n=10,10,10Day 8, AM, n=9,10,10Day 8, AM+PM, n=9,10,10
BMS-663068 1200 mg Q12H + RTV 100 mg QAM0.2940.8410.841

[back to top]

Cmax of Ritonavir Following Q12H Dosing

Blood samples were collected at indicated time points to assess Cmax of ritonavir following Q12H dosing. Geometric mean and geometric coefficient of variation are presented for Day 1, Day 8 morning dose (AM), Day 8 evening dose (PM) and Day 8 morning + evening dose (AM+PM). Pharmacokinetic parameter values were derived by non-compartmental methods. PK Population was used which comprised of all participants who receive ritonavir and provided pharmacokinetic samples. (NCT01009814)
Timeframe: Days 1, 8: pre-morning dose, 1,2,3,4,5,6,8,12 hours; Day 8: 13,14,15,16,17,18,20 hours

,
InterventionMicrograms per milliliter (μg/mL) (Geometric Mean)
Day 1, n=10,10,10Day 8, AM, n=9,10,10Day 8, PM, n=9,10,0Day 8, AM+PM, n=9,10,10
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H0.3141.301.481.39
BMS-663068 600 mg Q12H + RTV 100 mg Q12H0.3301.311.241.27

[back to top]

Cmax of Ritonavir Following QHS Dosing

Blood samples were collected at indicated time points to assess Cmax of ritonavir following QHS dosing. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 1, Day 8 evening dose (PM) and Day 8 morning (AM) + evening dose (PM). (NCT01009814)
Timeframe: Days 1, 8: pre-evening dose, 1,2,3,4,5,6,8 hours

Interventionμg/mL (Geometric Mean)
Day 1, n=10Day 8, PM, n=9Day 8, AM+PM, n=9
BMS-663068 1200 mg QHS + RTV 100 mg QHS0.2570.6280.628

[back to top]

Ctrough of BMS-626529 Following QHS Dosing

Blood samples were collected at indicated time points to assess Ctrough of BMS-626529 following QHS dosing. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 1, Days 5, 6, 7, 8 and Day 8 evening dose (PM). (NCT01009814)
Timeframe: Days 1, 8: pre-evening dose, 1,2,3,4,5,6,8 hours; Days 5,6,7: pre-evening dose

Interventionμg/mL (Geometric Mean)
Day 1, n=10Day 5, n=9Day 6, n=9Day 7, n=9Day 8, n=9Day 8, PM, n=9
BMS-663068 1200 mg QHS + RTV 100 mg QHS0.6990.07050.07430.07190.05460.0544

[back to top]

Ctrough of Ritonavir Following Q12H Dosing

Blood samples were collected at indicated time points to access Ctrough of ritonavir following Q12H dosing. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 1, Days 5, 6, 7, 8, Day 8 morning dose (AM) and Day 8 evening dose (PM). (NCT01009814)
Timeframe: Days 1, 8: pre-morning dose, 1,2,3,4,5,6,8,12 hours; Day 8: 13,14,15,16,17,18,20 hours; Days 5,6,7: pre-morning dose

InterventionMicrograms per milliliter (μg/mL) (Geometric Mean)
Day 5, n=9,10,10Day 6, n=9,10,10Day 7, n=9,10,10Day 8, n=9,10,10Day 8, AM, n=9,10,10
BMS-663068 1200 mg Q12H + RTV 100 mg QAM0.08310.08200.08600.08910.0912

[back to top]

Ctrough of Ritonavir Following Q12H Dosing

Blood samples were collected at indicated time points to access Ctrough of ritonavir following Q12H dosing. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 1, Days 5, 6, 7, 8, Day 8 morning dose (AM) and Day 8 evening dose (PM). (NCT01009814)
Timeframe: Days 1, 8: pre-morning dose, 1,2,3,4,5,6,8,12 hours; Day 8: 13,14,15,16,17,18,20 hours; Days 5,6,7: pre-morning dose

,
InterventionMicrograms per milliliter (μg/mL) (Geometric Mean)
Day 1, n=10,10,0Day 5, n=9,10,10Day 6, n=9,10,10Day 7, n=9,10,10Day 8, n=9,10,10Day 8, AM, n=9,10,10Day 8, PM, n=9,10,0
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H0.1310.5540.5940.5870.5030.3930.476
BMS-663068 600 mg Q12H + RTV 100 mg Q12H0.1170.5020.5260.4770.4700.3450.453

[back to top]

Ctrough of Ritonavir Following QHS Dosing

Blood samples were collected at indicated time points to assess Ctrough of ritonavir following QHS dosing. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 1, Days 5, 6, 7, 8 and Day 8 evening dose (PM). (NCT01009814)
Timeframe: Days 1, 8: pre-evening dose, 1,2,3,4,5,6,8 hours; Days 5,6,7: pre-evening dose

Interventionμg/mL (Geometric Mean)
Day 1, n=10Day 5, n=9Day 6, n=9Day 7, n=9Day 8, n=9Day 8, PM, n=9
BMS-663068 1200 mg QHS + RTV 100 mg QHS0.1570.1070.08450.08440.08080.0830

[back to top]

Inhibitory Quotient (IQ) of BMS-626529 by Css,Avg and by Lowest Concentration of a Drug During Dosing Interval (Cmin) Following Q12H Dosing

Blood samples were collected at indicated time points to assess IQ of BMS-626529. Inhibitory quotient was calculated as the ratio of BMS-626529 in vivo exposure to in vitro measured protein binding adjusted EC90 (PBA-EC90). The following in vivo exposure measures were used in evaluating IQ: Cmin and Css,avg. Geometric mean and geometric coefficient of variation are presented. (NCT01009814)
Timeframe: Days 1, 8: pre-morning dose, 1,2,3,4,5,6,8,12 hours; Day 8: 13,14,15,16,17,18,20 hours; Day 9: pre-dose, 4,12 hours; Day 10: pre-dose, 12 hours; Day 11: pre-dose; Days 5,6,7: pre-morning dose

,,,
InterventionRatio (Geometric Mean)
IQ CminIQ Css,avg
BMS-663068 1200 mg Q12H4.5415.4
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H97.5327
BMS-663068 1200 mg Q12H + RTV 100 mg QAM67.2252
BMS-663068 600 mg Q12H + RTV 100 mg Q12H11.029.7

[back to top]

Inhibitory Quotient (IQ) of BMS-626529 by Css,Avg and by Lowest Concentration of a Drug During Dosing Interval (Cmin) Following QHS Dosing

Blood samples were collected at indicated time points to assess IQ of BMS-626529. Inhibitory quotient was calculated as the ratio of BMS-626529 in vivo exposure to in vitro measured protein binding adjusted EC90 (PBA-EC90). The following in vivo exposure measures were used in evaluating IQ: Cmin and Css,avg. Geometric mean and geometric coefficient of variation are presented. (NCT01009814)
Timeframe: Days 1, 8: pre-evening dose, 1,2,3,4,5,6,8 hours; Day 2: pre-evening dose, 12,16 hours; Day 9: pre-dose, 12,16 hours; Day 10: pre-dose, 12 hours; Day 11: pre-dose; Days 5,6,7: pre-evening dose

InterventionRatio (Geometric Mean)
IQ CminIQ Css,avg
BMS-663068 1200 mg QHS + RTV 100 mg QHS3.8667.8

[back to top]

Maximum Observed Plasma Concentration (Cmax) of BMS-626529 Following Q12H Dosing

Blood samples were collected at indicated time points to assess Cmax of BMS-626529 following Q12H dosing. Geometric mean and geometric coefficient of variation are presented for Day 1, Day 8 morning dose (Anti Meridiem [AM]), Day 8 evening dose (Post Meridiem [PM]) and Day 8 morning + evening dose (AM+PM). Pharmacokinetic parameter values were derived by non-compartmental methods. Pharmacokinetic (PK) Population was used which comprised of all participants who receive BMS-663068 and provided pharmacokinetic samples. (NCT01009814)
Timeframe: Days 1, 8: pre-morning dose, 1,2,3,4,5,6,8,12 hours; Day 8: 13,14,15,16,17,18,20 hours

,,,
InterventionMicrograms per milliliter (μg/mL) (Geometric Mean)
Day 1, n=10,10,10,10Day 8, AM, n=9,10,10,10Day 8, PM, n=9,10,10,10Day 8, AM+PM, n=9,10,10,10
BMS-663068 1200 mg Q12H2.553.183.613.39
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H3.525.554.605.05
BMS-663068 1200 mg Q12H + RTV 100 mg QAM4.055.104.774.93
BMS-663068 600 mg Q12H + RTV 100 mg Q12H1.742.222.252.24

[back to top]

Number of Participants With Treatment Emergent Non-serious Adverse Event (Non-SAE) and Serious AE (SAE)

An AE is any new untoward medical occurrence or worsening of a pre-existing medical condition in a participant or clinical investigation participant administered an investigational (medicinal) product and that does not necessarily have a causal relationship with this treatment. Any untoward medical occurrence resulting in death, life threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, congenital anomaly/birth defect, any other situation according to medical or scientific judgment that may not be immediately life-threatening or result in death or hospitalization but may jeopardize the participant or may require medical or surgical intervention to prevent serious outcomes were categorized as SAE. Treatment emergent adverse events (occurred after start of treatment) have been presented.Safety Population comprised of all randomized participants who used the trial medication at least once. (NCT01009814)
Timeframe: Up to 50 days

,,,,
InterventionParticipants (Count of Participants)
Non-SAESAE
BMS-663068 1200 mg Q12H90
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H90
BMS-663068 1200 mg Q12H + RTV 100 mg QAM80
BMS-663068 1200 mg QHS + RTV 100 mg QHS50
BMS-663068 600 mg Q12H + RTV 100 mg Q12H80

[back to top]

Number of Participants With Worst-case Abnormalities in Diastolic Blood Pressure (DBP) and Systolic Blood Pressure (SBP)

Vital signs including DBP and SBP were recorded. Normal ranges were as following: For DBP, lower limit: value <55 millimeter of mercury (mmHg) and change <-20 mmHg; upper limit: value >90 mmHg and change >20 mmHg). For SBP, lower limit: value <90 mmHg and change <-10 mmHg; upper limit: value >140 mmHg and change >10 mmHg. Number of participants with worst-case abnormalities are presented. Worst-case abnormality was defined as: (1) Below Normal: at least one post-Baseline assessment was below normal range, and no post-Baseline assessment was above normal range. (2) Above Normal: at least one post-Baseline assessment was above normal range, and no post-Baseline assessment was below normal range. (3) Within Normal: all post-Baseline assessments were within normal range. It was to be considered as 'Missing' when there were no post-Baseline assessments. (NCT01009814)
Timeframe: Up to 50 days

,,,,
InterventionParticipants (Count of Participants)
SBP, Above NormalSBP, Below NormalSBP, Within NormalDBP, Above NormalDBP, Below NormalDBP, Within Normal
BMS-663068 1200 mg Q12H1090010
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H1090010
BMS-663068 1200 mg Q12H + RTV 100 mg QAM00100010
BMS-663068 1200 mg QHS + RTV 100 mg QHS3070010
BMS-663068 600 mg Q12H + RTV 100 mg Q12H00100010

[back to top]

Number of Participants With Worst-case Abnormalities in Electrocardiogram (ECG) Parameters

A 12-lead ECG was recorded during the study using an ECG machine that automatically measures ECG parameters. Normal range for ECG parameters were: PR interval (upper: 200 milliseconds [ms]); QRS (lower: 50 ms; upper: 120 ms); Corrected QT interval by Bazett formula (QTcB) (change from Baseline - increases by > 30 ms); Corrected QT interval by Fredericia formula (QTcF) (change from Baseline - increases by > 30 ms). Number of participants with worst-case abnormalities are presented which was defined as: (1) Below Normal: at least one post-Baseline assessment was below normal range, and no post-Baseline assessment was above normal range. (2) Above Normal: at least one post-Baseline assessment was above normal range, and no post-Baseline assessment was below normal range. Within Normal: all post-Baseline assessments were within normal range. It was to be considered as 'Missing' when there were no post-Baseline assessments. (NCT01009814)
Timeframe: Up to 50 days

,,,,
InterventionParticipants (Count of Participants)
PR interval, Above NormalPR interval, Below NormalPR interval, Within NormalQTcB, Above NormalQTcB, Below NormalQTcB, Within NormalQTcF, Above NormalQTcF, Below NormalQTcF, Within Normal
BMS-663068 1200 mg Q12H109109208
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H0010208109
BMS-663068 1200 mg Q12H + RTV 100 mg QAM001000100010
BMS-663068 1200 mg QHS + RTV 100 mg QHS1091090010
BMS-663068 600 mg Q12H + RTV 100 mg Q12H00100010109

[back to top]

Trough Observed Plasma Concentration (Ctrough) of BMS-626529 Following Q12H Dosing

Blood samples were collected at indicated time points to access Ctrough of BMS-626529 following Q12H dosing. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 1, Days 5, 6, 7, 8, Day 8 morning dose (AM) and Day 8 evening dose (PM). (NCT01009814)
Timeframe: Days 1, 8: pre-morning dose, 1,2,3,4,5,6,8,12 hours; Day 8: 13,14,15,16,17,18,20 hours; Days 5,6,7: pre-morning dose

,,,
InterventionMicrograms per milliliter (μg/mL) (Geometric Mean)
Day 1, n=10,10,10,10Day 5, n=9,10,10,10Day 6, n=9,10,10,10Day 7, n=9,10,10,10Day 8, n=9,10,10,10Day 8, AM, n=9,10,10,10Day 8, PM, n=9,10,10,10
BMS-663068 1200 mg Q12H0.2440.5180.4510.5420.6280.4690.487
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H0.4991.040.8391.080.7430.9510.653
BMS-663068 1200 mg Q12H + RTV 100 mg QAM0.6400.7600.7370.9980.7200.5790.553
BMS-663068 600 mg Q12H + RTV 100 mg Q12H0.3490.4730.4360.4950.4600.4220.358

[back to top]

Number of Participants With Worst-case Abnormalities in Body Temperature, Respiratory Rate [RR], and Heart Rate [HR]

Vital signs (body temperature, RR, and HR) were recorded. Normal range were: For HR, lower limit: 55 beats per minute (bpm) and change <-15 bpm; upper limit: >100 bpm and change >30 bpm). For temperature, lower limit: 36.0 Celsius; upper limit: >37.5 Celsius or change >1.7 Celsius). For RR, lower limit: 8 breaths per minute; upper limit: >16 breaths per minute or change >10 breaths per minute. Number of participants with worst-case abnormalities are presented. Worst-case abnormality was defined as: (1) Below Normal: at least one post-Baseline assessment was below normal range, and no post-Baseline assessment was above normal range. (2) Above Normal: at least one post-Baseline assessment was above normal range, and no post-Baseline assessment was below normal range. (3) Within Normal: all post-Baseline assessments were within normal range. It was to be considered as 'Missing' when there were no post-Baseline assessments. (NCT01009814)
Timeframe: Up to 50 days

,,,,
InterventionParticipants (Count of Participants)
HR, Above NormalHR, Below NormalHR, Within NormalRR, Above NormalRR, Below NormalRR, Within NormalTemperature, Above NormalTemperature, Below NormalTemperature, Within Normal
BMS-663068 1200 mg Q12H0198020010
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H0193070010
BMS-663068 1200 mg Q12H + RTV 100 mg QAM0010307019
BMS-663068 1200 mg QHS + RTV 100 mg QHS028307028
BMS-663068 600 mg Q12H + RTV 100 mg Q12H019406019

[back to top]

Accumulation Index (AI) of BMS-626529 Following Q12H Dosing

Blood samples were collected at indicated time points to assess Accumulation Index of BMS-626529 following Q12H dosing. AI was calculated as ratio of AUC(tau) at steady-state to AUC(tau) after the first dose. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 8 morning dose (AM). (NCT01009814)
Timeframe: Day 8: pre-morning dose, 1,2,3,4,5,6,8,12 hours; Day 8: 13,14,15,16,17,18,20 hours

InterventionRatio of AUC (Geometric Mean)
BMS-663068 600 mg Q12H + RTV 100 mg Q12H1.53
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H1.63
BMS-663068 1200 mg Q12H + RTV 100 mg QAM1.34
BMS-663068 1200 mg Q12H1.42

[back to top]

Accumulation Index of BMS-626529 Following QHS Dosing

Blood samples were collected at indicated time points to assess Accumulation Index of BMS-626529 following QHS dosing. AI was calculated as ratio of AUC(tau) at steady-state to AUC(tau) after the first dose. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 8 evening dose (PM). (NCT01009814)
Timeframe: Day 8: pre-evening dose, 1,2,3,4,5,6,8 hours

InterventionRatio of AUC (Geometric Mean)
BMS-663068 1200 mg QHS + RTV 100 mg QHS0.912

[back to top]

Accumulation Index of Ritonavir Following Q12H Dosing

Blood samples were collected at indicated time points to assess Accumulation Index of ritonavir following Q12H dosing. AI was calculated as ratio of AUC(tau) at steady-state to AUC(tau) after the first dose. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 8 morning dose (AM). (NCT01009814)
Timeframe: Day 8: pre-morning dose, 1,2,3,4,5,6,8,12 hours; Day 8: 13,14,15,16,17,18,20 hours

InterventionRatio of AUC (Geometric Mean)
BMS-663068 600 mg Q12H + RTV 100 mg Q12H3.58
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H4.41
BMS-663068 1200 mg Q12H + RTV 100 mg QAM3.62

[back to top]

Accumulation Index of Ritonavir Following QHS Dosing

Blood samples were collected at indicated time points to assess Accumulation Index of ritonavir following QHS dosing. AI was calculated as ratio of AUC(tau) at steady-state to AUC(tau) after the first dose. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 8 evening dose (PM). (NCT01009814)
Timeframe: Day 8: pre-evening dose, 1,2,3,4,5,6,8 hours

InterventionRatio of AUC (Geometric Mean)
BMS-663068 1200 mg QHS + RTV 100 mg QHS2.19

[back to top]

Area Under the Concentration-time Curve Over a 24-hour Period (AUC [0-24]) of BMS-626529 Following Q12H Dosing

Blood samples were collected at indicated time points to assess AUC (0-24) of BMS-626529 following Q12H dosing. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 8. (NCT01009814)
Timeframe: Day 8: pre-morning dose, 1,2,3,4,5,6,8,12, 13,14,15,16,17,18,20 hours

InterventionHour*micrograms per milliliter (Geometric Mean)
BMS-663068 600 mg Q12H + RTV 100 mg Q12H26.8
BMS-663068 1200 mg Q12H + RTV 100 mg Q12H60.6
BMS-663068 1200 mg Q12H + RTV 100 mg QAM55.1
BMS-663068 1200 mg Q12H42.6

[back to top]

AUC (0-24) of BMS-626529 Following QHS Dosing

Blood samples were collected at indicated time points to assess AUC (0-24) of BMS-626529 following QHS dosing. Pharmacokinetic parameter values were derived by non-compartmental methods. Geometric mean and geometric coefficient of variation are presented for Day 8. (NCT01009814)
Timeframe: Day 8: pre-evening dose, 1,2,3,4,5,6,8 hours

InterventionHour*micrograms per milliliter (Geometric Mean)
BMS-663068 1200 mg QHS + RTV 100 mg QHS23.0

[back to top]

Proportion of Subjects With HIV-1 RNA <50 Copies/mL

(NCT01010399)
Timeframe: 24 weeks

Interventionparticipants (Number)
Boosted Lexiva With Lovaza28

[back to top]

Proportion of Subjects With Triglycerides <200 mg/dL

(NCT01010399)
Timeframe: 24 weeks

Interventionparticipants (Number)
Boosted Lexiva With Lovaza12

[back to top]

Area Under the Curve From Time 0 to Tau (AUC 0-τ)

Area under the curve from start to elimination. (NCT01057433)
Timeframe: 0, 1, 2, 3, 4, 5, 6, 8, and 12 hours after dosing on days 19 and 24

Interventionng•h/mL (Mean)
All Subjects113.92

[back to top] [back to top]

Antepartum Component: Probability of Overall and HIV-free Infant Survival Until 104 Weeks of Age, by Antepartum Arm (in Conjunction With Infants in the Postpartum Component)

For overall survival, failure was defined to be death. For HIV-free survival, failure was defined to be either death or developing HIV. The probability of living, or living without HIV infection, at 104 weeks was calculated by Kaplan-Meier estimation of the survival function. (NCT01061151)
Timeframe: Measured from birth through 104 weeks of age

InterventionProportional probability (Number)
Overall survival, period 2 groupHIV-free survival, period 2 group
Antepartum Arm C0.9420.921

[back to top]

Antepartum Component: Number of Mothers With Obstetrical Complications

"Complications included deaths, diagnoses, signs/symptoms, chemistry lab tests, or hematological lab tests, with grades of 3 (Severe) or worse. Obstetrical complications were those classified by the MedDra coding system as Pregnancy, puerperium and perinatal conditions, except if the condition was the death of the fetus: Abortions not specified as induced or spontaneous, Abortions spontaneous, or Stillbirth and foetal death." (NCT01061151)
Timeframe: Measured through the Week 1 postpartum study visit

InterventionParticipants (Count of Participants)
Period 2
Antepartum Arm C23

[back to top]

Antepartum Component: Number of Mothers With Grade 3 or Higher Toxicities and Selected Grade 2 Hematologic, Renal, and Hepatic Adverse Events

These events were graded using the Division of AIDS (DAIDS) AE Grading Table, Version 1.0, December 2004, Clarification August 2009, which is available on the RSC website (http://rsc.tech-res.com). (NCT01061151)
Timeframe: Measured through the Week 1 postpartum study visit

,
InterventionParticipants (Count of Participants)
Periods 1 and 2Period 2
Antepartum Arm A26159
Antepartum Arm B31861

[back to top]

Antepartum Component: Number of Mothers With Grade 3 or Higher Toxicities and Selected Grade 2 Hematologic, Renal, and Hepatic Adverse Events

These events were graded using the Division of AIDS (DAIDS) AE Grading Table, Version 1.0, December 2004, Clarification August 2009, which is available on the RSC website (http://rsc.tech-res.com). (NCT01061151)
Timeframe: Measured through the Week 1 postpartum study visit

InterventionParticipants (Count of Participants)
Period 2
Antepartum Arm C60

[back to top]

Antepartum Component: Number of Mothers With Adverse Pregnancy Outcomes (e.g.,Stillbirth, Preterm Delivery (< 37 Weeks), Low Birth Weight (< 2,500 Grams), and Congenital Anomalies)

Composite outcome (NCT01061151)
Timeframe: Measured at birth

,
InterventionParticipants (Count of Participants)
Periods 1 and 2Period 2
Antepartum Arm A38991
Antepartum Arm B563123

[back to top]

Antepartum Component: Number of Confirmed Infant HIV Infections

Defined as HIV nucleic acid test (NAT) positivity of the specimen drawn at either the birth (Day 0-5) or Week 1 (Day 6-14) visit, confirmed by HIV NAT positivity of a second specimen collected at a different time point (NCT01061151)
Timeframe: Measured at birth or Week 1 study visit

InterventionParticipants (Count of Participants)
Antepartum Arm A25
Antepartum Arm B7
Antepartum Arm C2

[back to top]

Antepartum Component: Number of Mothers With Adverse Pregnancy Outcomes (e.g.,Stillbirth, Preterm Delivery (< 37 Weeks), Low Birth Weight (< 2,500 Grams), and Congenital Anomalies)

Composite outcome (NCT01061151)
Timeframe: Measured at birth

InterventionParticipants (Count of Participants)
Period 2
Antepartum Arm C111

[back to top]

Postpartum Component: Proportion of Mother-Infant Pairs With no Death or HIV Diagnosis Through 24 Months Post-delivery

Defined as infant HIV NAT positivity of a specimen drawn at any post-randomization visit, confirmed by HIV NAT positivity of a second specimen drawn at a different time point, or infant death. Analyses (Kaplan-Meier probabilities) were conducted at the Mother-Infant (M-I) pair level, hence the worst outcome for multiple births was counted as a single event. (NCT01061151)
Timeframe: Measured through 24 months post-delivery

InterventionProbability (Number)
Postpartum Arm A (Maternal Prophylaxis)0.971
Postpartum Arm B (Infant Prophylaxis)0.977

[back to top]

Postpartum Component: Incidence of Grade 3 or Higher Adverse Events and Selected Grade 2 Hematologic, Renal, and Hepatic Adverse Events

These events were graded using the Division of AIDS (DAIDS) AE Grading Table, Version 1.0, December 2004, Clarification August 2009, which is available on the RSC website (http://rsc.tech-res.com). (NCT01061151)
Timeframe: Measured through site recommended duration of breastfeeding, complete cessation of breastfeeding or 18 months of age, whichever comes first

InterventionNew cases per 100 person-years (Number)
Postpartum Arm A (Maternal Prophylaxis)14.4
Postpartum Arm B (Infant Prophylaxis)14.1

[back to top]

Antepartum Component: Number of Infant HIV Infections

Detected by HIV NAT positivity (NCT01061151)
Timeframe: Measured at the birth (<= 3 days postpartum) visit

InterventionParticipants (Count of Participants)
Antepartum Arm A22
Antepartum Arm B4
Antepartum Arm C2

[back to top]

Maternal Health Component: Incidence of AIDS-defining Illness

"AIDS-defining illness refers to the WHO Clinical Stage 4 illnesses listed in Appendix IV. Stage 4 illnesses were reviewed and confirmed by an Endpoint review group." (NCT01061151)
Timeframe: From study entry until July 7, 2015, an average of 94 weeks of follow-up.

InterventionNew cases per 100 person-years (Number)
Maternal Health Arm A (Continue Triple ARVs)0.08
Maternal Health Arm B (Discontinue Triple ARVs)0.25

[back to top] [back to top]

Antepartum Component: Number of Mothers With Obstetrical Complications

"Complications included deaths, diagnoses, signs/symptoms, chemistry lab tests, or hematological lab tests, with grades of 3 (Severe) or worse. Obstetrical complications were those classified by the MedDra coding system as Pregnancy, puerperium and perinatal conditions, except if the condition was the death of the fetus: Abortions not specified as induced or spontaneous, Abortions spontaneous, or Stillbirth and foetal death." (NCT01061151)
Timeframe: Measured through the Week 1 postpartum study visit

,
InterventionParticipants (Count of Participants)
Periods 1 and 2Period 2
Antepartum Arm A8920
Antepartum Arm B7512

[back to top]

Postpartum Component: Adherence to the Maternal and/or Infant ARV Regimens, as Measured by Maternal Report and Hair Measures

"Adherence is by maternal report; adherence through hair analysis is not included here.~The protocol did not distinguish between outcomes essential to the primary publication and outcomes for subsequent publications of lesser priority. This outcome measure was listed as secondary in the protocol but the intention was as an exploratory outcome since adherence was not a focus of the study." (NCT01061151)
Timeframe: Week 6 visit (14 days - 9 weeks postpartum); Week 14 visit (10-19 weeks postpartum); Week 26 visit (20 to 31 weeks postpartum); Week 50 visit (44 to 55 weeks postpartum); and Week 74 visit (68 to 80 weeks postpartum).

InterventionParticipants (Count of Participants)
Week 6 visit72329524Week 6 visit72329525Week 14 visit72329524Week 14 visit72329525Week 26 visit72329524Week 26 visit72329525Week 50 visit72329524Week 50 visit72329525Week 74 visit72329524Week 74 visit72329525
Missed dose over 1 month agoNever missed a doseMissed dose 2-4 weeks agoMissed dose within last 2 weeks
Postpartum Arm A (Maternal Prophylaxis)1003
Postpartum Arm B (Infant Prophylaxis)1104
Postpartum Arm A (Maternal Prophylaxis)12
Postpartum Arm A (Maternal Prophylaxis)17
Postpartum Arm B (Infant Prophylaxis)4
Postpartum Arm A (Maternal Prophylaxis)140
Postpartum Arm B (Infant Prophylaxis)74
Postpartum Arm A (Maternal Prophylaxis)956
Postpartum Arm B (Infant Prophylaxis)1081
Postpartum Arm A (Maternal Prophylaxis)20
Postpartum Arm B (Infant Prophylaxis)0
Postpartum Arm A (Maternal Prophylaxis)35
Postpartum Arm A (Maternal Prophylaxis)112
Postpartum Arm B (Infant Prophylaxis)50
Postpartum Arm A (Maternal Prophylaxis)888
Postpartum Arm B (Infant Prophylaxis)1035
Postpartum Arm A (Maternal Prophylaxis)48
Postpartum Arm A (Maternal Prophylaxis)31
Postpartum Arm B (Infant Prophylaxis)8
Postpartum Arm A (Maternal Prophylaxis)103
Postpartum Arm B (Infant Prophylaxis)47
Postpartum Arm A (Maternal Prophylaxis)716
Postpartum Arm B (Infant Prophylaxis)841
Postpartum Arm A (Maternal Prophylaxis)37
Postpartum Arm A (Maternal Prophylaxis)34
Postpartum Arm B (Infant Prophylaxis)7
Postpartum Arm A (Maternal Prophylaxis)64
Postpartum Arm B (Infant Prophylaxis)30
Postpartum Arm A (Maternal Prophylaxis)311
Postpartum Arm B (Infant Prophylaxis)377
Postpartum Arm A (Maternal Prophylaxis)15
Postpartum Arm B (Infant Prophylaxis)2
Postpartum Arm B (Infant Prophylaxis)1
Postpartum Arm B (Infant Prophylaxis)9

[back to top]

Antepartum Component: Maternal HIV RNA Less Than 400 Copies/mL at Delivery

Analysis used the principle of intent to treat. (NCT01061151)
Timeframe: Measured at the time of delivery

InterventionParticipants (Count of Participants)Participants (Count of Participants)
Periods 1 and 272329519Periods 1 and 272329520Period 272329520Period 272329519Period 272329521
HIV RNA < 400 copies/mLHIV RNA >= 400 copies/mL
Antepartum Arm A415
Antepartum Arm B1092
Antepartum Arm A929
Antepartum Arm B275
Antepartum Arm A102
Antepartum Arm B259
Antepartum Arm C225
Antepartum Arm A210
Antepartum Arm B62
Antepartum Arm C79

[back to top]

Postpartum Component: Proportion of Infants Alive Through 12 and 24 Months Post-delivery

Analyses (Kaplan-Meier probabilities) conducted for all individual infants (rather than M-I pair) (NCT01061151)
Timeframe: Measured at 12 and 24 months post-delivery

,
InterventionProbability (Number)
12 months post delivery24 months post delivery
Postpartum Arm A (Maternal Prophylaxis)0.9880.978
Postpartum Arm B (Infant Prophylaxis)0.9890.987

[back to top]

Antepartum Component: Probability of Overall and HIV-free Infant Survival Until 104 Weeks of Age, by Antepartum Arm (in Conjunction With Infants in the Postpartum Component)

For overall survival, failure was defined to be death. For HIV-free survival, failure was defined to be either death or developing HIV. The probability of living, or living without HIV infection, at 104 weeks was calculated by Kaplan-Meier estimation of the survival function. (NCT01061151)
Timeframe: Measured from birth through 104 weeks of age

,
InterventionProportional probability (Number)
Overall survival, Periods 1 & 2 group (arms A & B only)Overall survival, period 2 groupHIV-free survival, Periods 1&2 group (arms A&B only)HIV-free survival, period 2 group
Antepartum Arm A0.9590.9510.9370.936
Antepartum Arm B0.9670.9820.9470.940

[back to top]

Postpartum Component: Incidence of Confirmed Infant HIV Infection

Defined as infant HIV NAT positivity of a specimen drawn at any post-randomization visit (i.e., any visit after the Week 1 [Day 6-14] visit), confirmed by HIV NAT positivity of a second specimen drawn at a different time point. Analyses were conducted at the Mother-Infant (M-I) pair level, hence the worst outcome for multiple births was counted as a single event. (NCT01061151)
Timeframe: Measured through site recommended duration of breastfeeding, complete cessation of breastfeeding or 18 months of age, whichever comes first

InterventionNew cases per 100 person-years (Number)
Postpartum Arm A (Maternal Prophylaxis)0.56
Postpartum Arm B (Infant Prophylaxis)0.55

[back to top]

Maternal Health Component: Incidence of Death

Number of women who died during the maternal health component; that is, who had been randomized to either continue or discontinue ART after risk of HIV vertical transmission through breastfeeding was over. (NCT01061151)
Timeframe: From study entry until July 7, 2015, an average of 94 weeks of follow-up.

InterventionNew cases per 100 person-years (Number)
Maternal Health Arm A (Continue Triple ARVs)0.24
Maternal Health Arm B (Discontinue Triple ARVs)0.43

[back to top]

Maternal Health Component: Incidence Rate of Progression to AIDS-defining Illness, Death, or a Serious Non-AIDS Cardiovascular, Hepatic, or Renal Event

"This outcome included AIDS-defining illnesses or cardiovascular, hepatic, or renal adverse events of particular concern which were evaluated as serious. Serious outcomes were both those defined as serious according to the International Conference on Harmonization (ICH) definition, or outcomes with grades equal to or worse than 3 (Severe). A Poisson model with time to first event as an offset and an over-dispersion parameter was used to estimate incidence rates. Cardiovascular events considered were hypertension, congestive heart failure, stroke, Transient Ischemia Event (TIA), pulmonary embolism, myocardial infarction (whether acute symptomatic or silent), coronary artery disease, deep vein thrombosis, peripheral vascular disease, or symptomatic HIV-associated cardiomyopathy. Hepatic events considered were cirrhosis and idiopathic sclerosing cholangitis. Renal events considered were renal insufficiency, acute or chronic." (NCT01061151)
Timeframe: From study entry until July 7, 2015, an average of 94 weeks of follow-up.

Interventionevents per 100 person-years (Number)
Maternal Health Arm A (Continue Triple ARVs)0.5
Maternal Health Arm B (Discontinue Triple ARVs)0.9

[back to top] [back to top]

Maternal Health Component: Toxicity: Incidence of Grade 3 or Greater Laboratory Results or Signs and Symptoms and Selected Grade 2 Hematologic, Renal, and Hepatic Laboratory Results

The maternal safety endpoints summarized include grade 2, 3 or 4 hematologies (hemoglobin (Hb), White Blood Cells (WBC), Absolute Neutrophil Count (ANC), platelet count), chemistries (Alanine Aminotransferase (ALT or SGPT), serum creatinine), and grade 3 or 4 signs and symptoms that occurred post-randomization. These events were graded using the Division of AIDS (DAIDS) AE Grading Table, Version 1.0, December 2004, Clarification August 2009, which is available on the RSC website (http://rsc.tech-res.com). (NCT01061151)
Timeframe: From study entry until July 7, 2015, an average of 94 weeks of follow-up.

InterventionNew cases per 100 person-years (Number)
Maternal Health Arm A (Continue Triple ARVs)15.3
Maternal Health Arm B (Discontinue Triple ARVs)13.9

[back to top]

Maternal Health Component: Incidence of Progression to AIDS-defining Illness or Death

AIDS-defining illness refers to the WHO Clinical Stage 4 illnesses in Appendix IV of the protocol. These events were reviewed and confirmed by an Endpoint review group. (NCT01061151)
Timeframe: From study entry until July 7, 2015, an average of 94 weeks of follow-up.

InterventionNew cases per 100 person-years (Number)
Maternal Health Arm A (Continue Triple ARVs)0.24
Maternal Health Arm B (Discontinue Triple ARVs)0.49

[back to top]

Maternal Health Component: Incidence of Tuberculosis

Incidence of tuberculosis. (NCT01061151)
Timeframe: From study entry until July 7, 2015, an average of 94 weeks of follow-up.

InterventionNew cases per 100 person-years (Number)
Maternal Health Arm A (Continue Triple ARVs)0.40
Maternal Health Arm B (Discontinue Triple ARVs)0.31

[back to top]

Maternal Health Component: Incidence Rate of Cardiovascular or Other Metabolic Events

"Cardiovascular or metabolic events of particular concern were included in this analysis. A Poisson model with time to first event as an offset and an over-dispersion parameter was used to estimate incidence rates.~Metabolic events considered were diabetes mellitus, lipodystrophy, or dyslipidemia. Cardiovascular events considered were hypertension, congestive heart failure, stroke, Transient Ischemia Event (TIA), pulmonary embolism, myocardial infarction (whether acute symptomatic or silent), coronary artery disease, deep vein thrombosis, peripheral vascular disease, or symptomatic HIV-associated cardiomyopathy." (NCT01061151)
Timeframe: From study entry until July 7, 2015, an average of 94 weeks of follow-up.

Interventionevents per 100 person-years (Number)
Maternal Health Arm A (Continue Triple ARVs)2.9
Maternal Health Arm B (Discontinue Triple ARVs)5.7

[back to top]

Maternal Health Component: Incidence Rate of Death or Any Condition of Particular Concern

Particular events were targeted as those of particular concern. This outcome considered all such events: death, events defining WHO stages II, III, or IV, targeted cardiovascular adverse events, other targeted adverse events, or cancers which were not AIDS-defining. A complete list can be found in Appendix IV of the Protocol. A Poisson model with time to first event as an offset and an over-dispersion parameter was used to estimate incidence rates. (NCT01061151)
Timeframe: From study entry until July 7, 2015, an average of 94 weeks of follow-up.

Interventionevents per 100 person-years (Number)
Maternal Health Arm A (Continue Triple ARVs)9.0
Maternal Health Arm B (Discontinue Triple ARVs)14.0

[back to top]

Maternal Health Component: Other Targeted Medical Conditions

Other (non-cardiologic) medical conditions of particular concern were included in this outcome. A Poisson model with time to first event as an offset and an over-dispersion parameter was used to estimate incidence rates. Events included were metabolic events, hepatic events, renal events, infections such as pulmonary tuberculosis, malaria, or other serious bacterial infections, and others. (NCT01061151)
Timeframe: From study entry until July 7, 2015, an average of 94 weeks of follow-up.

Interventionevents per 100 person-years (Number)
Maternal Health Arm A (Continue Triple ARVs)4.0
Maternal Health Arm B (Discontinue Triple ARVs)4.6

[back to top]

Number of Participants With HIV RNA < 50 and <400 Copies/ml.

(NCT01068873)
Timeframe: week 24

InterventionParticipants (Count of Participants)
Open Label Single Arm1

[back to top]

Number of Participants With Resistance-Associated Variants and Phenotypic Resistance to ABT-333 in Non-structural Viral Protein 5B (NS5B)

Baseline samples were analyzed for resistance-associated amino acid variants using population and clonal sequencing and were compared with the appropriate reference sequence (1a-H77 or 1b-Con1). Phenotypic resistance to ABT-333 at baseline was assessed by calculating the fold difference in the half maximal effective concentration (EC50) compared with the EC50 for the appropriate reference replicon (1a-H77 or 1b-Con1). Available samples at Day 4 with HCV RNA ≥ 1000 IU/mL were analyzed for the presence of resistance-associated variants using population and clonal sequencing and were compared with the baseline sequences to assess amino acid changes. Phenotypic resistance to ABT-333 at Day 4 was assessed by calculating the fold difference in the EC50 compared with the EC50 for the corresponding baseline sample. The number of participants with variants at resistance-associated amino acid positions and phenotypic resistance are presented. (NCT01074008)
Timeframe: Baseline and Day 4

,
Interventionparticipants (Number)
Baseline Variants in NS5BBaseline Resistance to ABT-333 >10-fold NS5BDay 4: Variants in NS5B (n=7, 8)Day 4: Resistance to ABT-333 >10-fold (n=7, 8)
ABT-333 (400 mg) Twice a Day (BID) + pegIFN/RBV1030
ABT-333 (800 mg) Twice Daily (BID) + pegIFN/RBV2130

[back to top]

Number of Participants With Resistance-Associated Variants and Phenotypic Resistance to ABT-072 in Non-structural Viral Protein 5B (NS5B)

Baseline samples were analyzed for resistance-associated amino acid variants using population and clonal sequencing and were compared with the appropriate reference sequence (1a-H77 or 1b-Con1). Phenotypic resistance to ABT-072 at baseline was assessed by calculating the fold difference in the half maximal effective concentration (EC50) compared with the EC50 for the appropriate reference replicon (1a-H77 or 1b-Con1). Available samples at Day 4 with HCV RNA ≥ 1000 IU/mL were analyzed for the presence of resistance-associated variants using population and clonal sequencing and were compared with the baseline sequences to assess amino acid changes. Phenotypic resistance to ABT-072 at Day 4 was assessed by calculating the fold difference in the EC50 compared with the EC50 for the corresponding baseline sample. The number of participants with variants at resistance-associated amino acid positions and phenotypic resistance are presented. (NCT01074008)
Timeframe: Baseline and Day 4

,,
Interventionparticipants (Number)
Baseline Variants in NS5BBaseline Resistance to ABT-072 >10-foldDay 4: Variants in NS5B (n=6, 7, 6)Day 4: Resistance to ABT-072 >10-fold (n=6, 7, 6)
ABT-072 (100 mg) Once Daily (QD) + pegIFN/RBV0010
ABT-072 (300 mg) Once Daily (QD) + pegIFN/RBV1120
ABT-072 (600 mg) Once Daily (QD) + pegIFN/RBV0011

[back to top]

Time to Maximum Plasma Concentration (Tmax) of Ritonavir

Blood samples were collected immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose). The samples were analyzed for the concentration of ritonavir using validated analytical methods. The time to maximum plasma concentration (Tmax; measured in hours) is the time it takes for a drug to achieve Cmax. The Tmax of ritonavir was estimated using non-compartmental methods and data are reported as the mean ± standard deviation. (NCT01074008)
Timeframe: Immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose)

InterventionHours (Mean)
ABT-450/r (50/100 mg) Once Daily (QD) + pegIFN/RBV4.8
ABT-450/r (100/100 mg) Once Daily (QD) + pegIFN/RBV5.3
ABT-450/r (200/100 mg) Once Daily (QD) + pegIFN/RBV4.5

[back to top]

Time to Maximum Plasma Concentration (Tmax) of ABT-450

Blood samples were collected immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose). The samples were analyzed for the concentration of ABT-450 using validated analytical methods. The time to maximum plasma concentration (Tmax; measured in hours) is the time it takes for a drug to achieve Cmax. The Tmax of ABT-450 was estimated using non-compartmental methods and data are reported as the mean ± standard deviation. (NCT01074008)
Timeframe: Immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose)

InterventionHours (Mean)
ABT-450/r (50/100 mg) Once Daily (QD) + pegIFN/RBV2.5
ABT-450/r (100/100 mg) Once Daily (QD) + pegIFN/RBV4.8
ABT-450/r (200/100 mg) Once Daily (QD) + pegIFN/RBV3.0

[back to top]

Time to Maximum Plasma Concentration (Tmax) of ABT-333

Blood samples were collected immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose). The samples were analyzed for the concentration of ABT-333 using validated analytical methods. The time to maximum plasma concentration (Tmax; measured in hours) is the time it takes for a drug to achieve Cmax. The Tmax of ABT-333 was estimated using non-compartmental methods and data are reported as the mean ± standard deviation. (NCT01074008)
Timeframe: Immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose)

InterventionHours (Mean)
ABT-333 (400 mg) Twice a Day (BID) + pegIFN/RBV4.5
ABT-333 (800 mg) Twice Daily (BID) + pegIFN/RBV7.0

[back to top]

Time to Maximum Plasma Concentration (Tmax) of ABT-072

Blood samples were collected immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose). The samples were analyzed for the concentration of ABT-072 using validated analytical methods. The time to maximum plasma concentration (Tmax; measured in hours) is the time it takes for a drug to achieve Cmax. The Tmax of ABT-072 was estimated using non-compartmental methods and data are reported as the mean ± standard deviation. (NCT01074008)
Timeframe: Immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose)

InterventionHours (Mean)
ABT-072 (100 mg) Once Daily (QD) + pegIFN/RBV4.5
ABT-072 (300 mg) Once Daily (QD) + pegIFN/RBV2.8
ABT-072 (600 mg) Once Daily (QD) + pegIFN/RBV3.1

[back to top]

Percentage of Participants With Rapid Virologic Response (RVR) at Week 4

Plasma hepatitis C virus ribonucleic acid (HCV RNA) levels were determined for each sample using a real-time reverse transcriptase-polymerase chain reaction (RT-PCR) assay that had a lower limit of detection of 10 IU/mL and a lower limit of quantification (LLOQ) of 25 IU/mL. Rapid virologic response was defined as HCV RNA level < LLOQ (< 25 IU/mL) at Week 4. Data are reported as the percentage of participants with RVR. (NCT01074008)
Timeframe: Week 4

Interventionpercentage of participants (Number)
ABT-450/r (50/100 mg) Once Daily (QD) + pegIFN/RBV87.5
ABT-450/r (100/100 mg) Once Daily (QD) + pegIFN/RBV75.0
ABT-450/r (200/100 mg) Once Daily (QD) + pegIFN/RBV100.0
ABT-072 (100 mg) Once Daily (QD) + pegIFN/RBV37.5
ABT-072 (300 mg) Once Daily (QD) + pegIFN/RBV12.5
ABT-072 (600 mg) Once Daily (QD) + pegIFN/RBV42.9
ABT-333 (400 mg) Twice a Day (BID) + pegIFN/RBV50.0
ABT-333 (800 mg) Twice Daily (BID) + pegIFN/RBV50.0
Placebo + pegIFN/RBV9.1

[back to top]

Percentage of Participants With Complete Early Virologic Response (cEVR) at Week 12

Plasma hepatitis C virus ribonucleic acid (HCV RNA) levels were determined for each sample using a real-time reverse transcriptase-polymerase chain reaction (RT-PCR) assay that had a lower limit of detection of 10 IU/mL and a lower limit of quantification (LLOQ) of 25 IU/mL. Complete EVR was defined as HCV RNA levels < LLOQ (< 25 IU/mL) at Week 12. Data are reported as the percentage of participants with cEVR. (NCT01074008)
Timeframe: Week 12

Interventionpercentage of participants (Number)
ABT-450/r (50/100 mg) Once Daily (QD) + pegIFN/RBV87.5
ABT-450/r (100/100 mg) Once Daily (QD) + pegIFN/RBV87.5
ABT-450/r (200/100 mg) Once Daily (QD) + pegIFN/RBV100.0
ABT-072 (100 mg) Once Daily (QD) + pegIFN/RBV75.0
ABT-072 (300 mg) Once Daily (QD) + pegIFN/RBV50.0
ABT-072 (600 mg) Once Daily (QD) + pegIFN/RBV85.7
ABT-333 (400 mg) Twice a Day (BID) + pegIFN/RBV87.5
ABT-333 (800 mg) Twice Daily (BID) + pegIFN/RBV62.5
Placebo + pegIFN/RBV18.2

[back to top]

Maximum Plasma Concentration (Cmax) of Ritonavir

Blood samples were collected immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose). The samples were analyzed for the concentration of ritonavir using validated analytical methods. The maximum plasma concentration (Cmax; measured in ng/mL) is the highest concentration that a drug achieves in the blood after administration in a dosing interval. The Cmax of ritonavir was estimated using non-compartmental methods and data are reported as the mean ± standard deviation. (NCT01074008)
Timeframe: Immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose)

Interventionng/mL (Mean)
ABT-450/r (50/100 mg) Once Daily (QD) + pegIFN/RBV563
ABT-450/r (100/100 mg) Once Daily (QD) + pegIFN/RBV851
ABT-450/r (200/100 mg) Once Daily (QD) + pegIFN/RBV1098

[back to top]

Maximum Plasma Concentration (Cmax) of ABT-450

Blood samples were collected immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose). The samples were analyzed for the concentration of ABT-450 using validated analytical methods. The maximum plasma concentration (Cmax; measured in ng/mL) is the highest concentration that a drug achieves in the blood after administration in a dosing interval. The Cmax of ABT-450 was estimated using non-compartmental methods and data are reported as the mean ± standard deviation. (NCT01074008)
Timeframe: Immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose)

Interventionng/mL (Mean)
ABT-450/r (50/100 mg) Once Daily (QD) + pegIFN/RBV34.4
ABT-450/r (100/100 mg) Once Daily (QD) + pegIFN/RBV165
ABT-450/r (200/100 mg) Once Daily (QD) + pegIFN/RBV2923

[back to top]

Maximum Plasma Concentration (Cmax) of ABT-333

Blood samples were collected immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose). The samples were analyzed for the concentration of ABT-333 using validated analytical methods. The maximum plasma concentration (Cmax; measured in ng/mL) is the highest concentration that a drug achieves in the blood after administration in a dosing interval. The Cmax of ABT-333 was estimated using non-compartmental methods and data are reported as the mean ± standard deviation. (NCT01074008)
Timeframe: Immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose)

Interventionng/mL (Mean)
ABT-333 (400 mg) Twice a Day (BID) + pegIFN/RBV800
ABT-333 (800 mg) Twice Daily (BID) + pegIFN/RBV1201

[back to top]

Maximum Plasma Concentration (Cmax) of ABT-072

Blood samples were collected immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose). The samples were analyzed for the concentration of ABT-072 using validated analytical methods. The maximum plasma concentration (Cmax; measured in ng/mL) is the highest concentration that a drug achieves in the blood after administration in a dosing interval. The Cmax of ABT-072 was estimated using non-compartmental methods and data are reported as the mean ± standard deviation. (NCT01074008)
Timeframe: Immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose)

Interventionng/mL (Mean)
ABT-072 (100 mg) Once Daily (QD) + pegIFN/RBV390
ABT-072 (300 mg) Once Daily (QD) + pegIFN/RBV1218
ABT-072 (600 mg) Once Daily (QD) + pegIFN/RBV1748

[back to top]

Maximal Change From Baseline in Hepatitis C Virus Ribonucleic Acid (HCV RNA) Levels During ABT-450/r, ABT-333, or ABT-072 Monotherapy Treatment

Plasma HCV RNA levels (reported as log10 IU/mL) were determined for each sample using a real-time reverse transcriptase-polymerase chain reaction (RT-PCR) assay that had a lower limit of detection of 10 IU/mL and a lower limit of quantification of 25 IU/mL. The baseline value was the HCV RNA level before the first dose of study drug on Day 1. The maximal change during monotherapy was the difference from baseline to the lowest log10 HCV RNA level anytime after the first dose of study drug on Day 1 through the last log10 HCV RNA level before the first dose of study drug on Day 4. Data are reported as the mean ± standard deviation. (NCT01074008)
Timeframe: Prior to dosing on Day 1 to before the morning dose on Day 4

Interventionlog10 IU/mL (Mean)
ABT-450/r (50/100 mg) Once Daily (QD) + pegIFN/RBV-4.07
ABT-450/r (100/100 mg) Once Daily (QD) + pegIFN/RBV-3.91
ABT-450/r (200/100 mg) Once Daily (QD) + pegIFN/RBV-4.11
ABT-072 (100 mg) Once Daily (QD) + pegIFN/RBV-1.14
ABT-072 (300 mg) Once Daily (QD) + pegIFN/RBV-1.07
ABT-072 (600 mg) Once Daily (QD) + pegIFN/RBV-1.57
ABT-333 (400 mg) Twice a Day (BID) + pegIFN/RBV-1.08
ABT-333 (800 mg) Twice Daily (BID) + pegIFN/RBV-0.95
Placebo + pegIFN/RBV-0.36

[back to top]

Change From Baseline in SF-36 Physical Component Summary (PCS)

The Physical Component Summary (PCS) of the SF-36 was used to measure the overall physical health status of a participant. The aggregated score of the SF-36 PCS score was standardized using a linear T-score transformation with a mean of 50 and a standard deviation of 10; a higher score indicated better physical function and well-being. Data presented are the summaries across all participants, for each treatment arm, regardless of dose. Data are presented as the group mean change from baseline ± standard deviation. (NCT01074008)
Timeframe: Baseline and Post-treatment Week 24

Interventionunits on a scale (Mean)
ABT-450/r (Regardless of Dose) + pegIFN/RBV3.01
ABT-072 (Regardless of Dose) + pegIFN/RBV-1.92
ABT-333 (Regardless of Dose) + pegIFN/RBV4.08
Placebo (Regardless of Dose) + pegIFN/RBV-2.84

[back to top]

Change From Baseline in SF-36 Mental Component Summary (MCS)

The Mental Component Summary (MCS) of the SF-36 was used to measure the overall mental health status of participants. The aggregated score of the SF-36 MPS was standardized using a linear T-score transformation with a mean of 50 and a standard deviation of 10; a higher score indicated better mental function and well-being. Data presented are the summaries across all participants, for each treatment arm, regardless of dose. Data are presented as the group mean change from baseline ± standard deviation. (NCT01074008)
Timeframe: Baseline and Post-treatment Week 24

Interventionunits on a scale (Mean)
ABT-450/r (Regardless of Dose) + pegIFN/RBV-2.77
ABT-072 (Regardless of Dose) + pegIFN/RBV-0.41
ABT-333 (Regardless of Dose) + pegIFN/RBV-4.37
Placebo (Regardless of Dose) + pegIFN/RBV-10.29

[back to top]

Change From Baseline in Hepatitis C Virus Patient-reported Outcomes (HCV-PRO) Total Score

The Hepatitis C Virus Patient-report Outcomes (HCV-PRO, formerly known as HCV Quality of Life) survey was used to assess disease-specific function and well-being on a scale from 0 to 100; a higher score indicated relatively good function and well-being of treated participants. Data presented are the summaries across all participants, for each treatment arm, regardless of dose. Data are reported as the group mean change from baseline ± standard deviation. (NCT01074008)
Timeframe: Baseline up to Post-treatment Week 24

Interventionunits on a scale (Mean)
ABT-450/r (Regardless of Dose) + pegIFN/RBV-0.78
ABT-072 (Regardless of Dose) + pegIFN/RBV-0.37
ABT-333 (Regardless of Dose) + pegIFN/RBV-3.25
Placebo (Regardless of Dose) + pegIFN/RBV-9.04

[back to top]

Change From Baseline in EQ-5D (3 Level) Health Index Score

The EQ-5D was a health state questionnaire used to measure five health dimensions (mobility, self-care, usual activities, pain/discomfort, and anxiety/depression). The combination of responses from all five dimensions were derived into an index score ranging from 0 to 1; a higher score indicated a more preferable health utility value from the societal perspectives. Data presented are the summaries across all participants, for each treatment arm, regardless of dose. Data are presented as the group mean change from baseline ± standard deviation. (NCT01074008)
Timeframe: Baseline and Post-treatment Week 24

Interventionunits on a scale (Mean)
ABT-450/r (Regardless of Dose) + pegIFN/RBV-0.06
ABT-072 (Regardless of Dose) + pegIFN/RBV-0.06
ABT-333 (Regardless of Dose) + pegIFN/RBV-0.02
Placebo (Regardless of Dose) + pegIFN/RBV-0.09

[back to top]

Change From Baseline in ED-5D Visual Analog Scale (ED-5D VAS) Score

The ED-5D VAS was a self-rating survey used to capture the current health status of a participant and ranged from 0 (the worst imaginable health state) to 100 (best imaginable health state). Data presented are the summaries across all participants, for each treatment arm, regardless of dose. Data are presented as the group mean change from baseline ± standard deviation. (NCT01074008)
Timeframe: Baseline and Post-treatment Week 24

Interventionunits on a scale (Mean)
ABT-450/r (Regardless of Dose) + pegIFN/RBV-5.91
ABT-072 (Regardless of Dose) + pegIFN/RBV-2.38
ABT-333 (Regardless of Dose) + pegIFN/RBV11.18
Placebo (Regardless of Dose) + pegIFN/RBV-9.00

[back to top]

Area Under the Plasma Concentration-time Curve From 0 to 24 Hours (AUC24) Post-dose of Ritonavir

Blood samples were collected immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose). The samples were analyzed for the concentration of ritonavir using validated analytical methods. The area under the plasma concentration-time curve (AUC; measured in ng*hr/mL) is a method of measurement to determine the total exposure of a drug in blood plasma. The AUC24 of ritonavir was estimated using non-compartmental methods and data are reported as the mean ± standard deviation. (NCT01074008)
Timeframe: Immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose)

Interventionng*hr/mL (Mean)
ABT-450/r (50/100 mg) Once Daily (QD) + pegIFN/RBV4518
ABT-450/r (100/100 mg) Once Daily (QD) + pegIFN/RBV7638
ABT-450/r (200/100 mg) Once Daily (QD) + pegIFN/RBV8663

[back to top]

Area Under the Plasma Concentration-time Curve From 0 to 12 Hours (AUC12) Post-dose of ABT-333

Blood samples were collected immediately prior to morning dose (time 0 hours) and at 2, 4, 8, and 12 hours after the morning dose on Day 1. The samples were analyzed for the concentration of ABT-333 using validated analytical methods. The area under the plasma concentration-time curve (AUC; measured in ng*hr/mL) is a method of measurement to determine the total exposure of a drug in blood plasma. The AUC12 of ABT-333 was estimated using non-compartmental methods and data are reported as the mean ± standard deviation. (NCT01074008)
Timeframe: Immediately prior to morning dose (time 0 hours) and at 2, 4, 8, and 12 hours after the morning dose on Day 1

Interventionng*hr/mL (Mean)
ABT-333 (400 mg) Twice a Day (BID) + pegIFN/RBV4315
ABT-333 (800 mg) Twice Daily (BID) + pegIFN/RBV6431

[back to top]

Area Under the Plasma Concentration-time Curve From 0 to 24 Hours (AUC24) Post-dose of ABT-450

Blood samples were collected immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose). The samples were analyzed for the concentration of ABT-450 using validated analytical methods. The area under the plasma concentration-time curve (AUC; measured in ng*hr/mL) is a method of measurement to determine the total exposure of a drug in blood plasma. The AUC24 of ABT-450 was estimated using non-compartmental methods and data are reported as the mean ± standard deviation. (NCT01074008)
Timeframe: Immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose)

Interventionng*hr/mL (Mean)
ABT-450/r (50/100 mg) Once Daily (QD) + pegIFN/RBV250
ABT-450/r (100/100 mg) Once Daily (QD) + pegIFN/RBV1122
ABT-450/r (200/100 mg) Once Daily (QD) + pegIFN/RBV17351

[back to top]

Area Under the Plasma Concentration-time Curve From 0 to 24 Hours (AUC24) Post-dose of ABT-072

Blood samples were collected immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose). The samples were analyzed for the concentration of ABT-072 using validated analytical methods. The area under the plasma concentration-time curve (AUC; measured in ng*hr/mL) is a method of measurement to determine the total exposure of a drug in blood plasma. The AUC24 of ABT-072 was estimated using non-compartmental methods and data are reported as the mean ± standard deviation. (NCT01074008)
Timeframe: Immediately prior to morning dose (time 0 hours); 2, 4, 8, 12, and 16 hours after the morning dose on Day 1; and prior to dose on Day 2 (24 hours after Day 1 dose)

Interventionng*hr/mL (Mean)
ABT-072 (100 mg) Once Daily (QD) + pegIFN/RBV3678
ABT-072 (300 mg) Once Daily (QD) + pegIFN/RBV9413
ABT-072 (600 mg) Once Daily (QD) + pegIFN/RBV14126

[back to top]

Percentage of Participants With Partial Early Virologic Response (EVR) at Week 12

Plasma hepatitis C virus ribonucleic acid (HCV RNA) levels were determined for each sample using a real-time reverse transcriptase-polymerase chain reaction (RT-PCR) assay that had a lower limit of detection of 10 IU/mL and a lower limit of quantification of 25 IU/mL. Partial early virologic response (EVR) was defined as HCV RNA levels that decreased > 2 log10 IU/mL at Week 12 as compared to baseline. The baseline value was the last measurement before the first dose on Day 1. Data are reported as the percentage of participants with partial EVR. (NCT01074008)
Timeframe: Baseline and Week 12

Interventionpercentage of participants (Number)
ABT-450/r (50/100 mg) Once Daily (QD) + pegIFN/RBV100.0
ABT-450/r (100/100 mg) Once Daily (QD) + pegIFN/RBV87.5
ABT-450/r (200/100 mg) Once Daily (QD) + pegIFN/RBV100.0
ABT-072 (100 mg) Once Daily (QD) + pegIFN/RBV100.0
ABT-072 (300 mg) Once Daily (QD) + pegIFN/RBV62.5
ABT-072 (600 mg) Once Daily (QD) + pegIFN/RBV100.0
ABT-333 (400 mg) Twice a Day (BID) + pegIFN/RBV100.0
ABT-333 (800 mg) Twice Daily (BID) + pegIFN/RBV100.0
Placebo + pegIFN/RBV36.4

[back to top]

Number of Participants With Resistance-Associated Variants and Phenotypic Resistance to ABT-450 in Non-structural Viral Protein 3 (NS3)

Baseline samples were analyzed for resistance-associated amino acid variants using population and clonal sequencing and were compared with the appropriate reference sequence (1a-H77 or 1b-Con1). Phenotypic resistance to ABT-450 at baseline was assessed by calculating the fold difference in the half maximal effective concentration (EC50) compared with the EC50 for the appropriate reference replicon (1a-H77 or 1b-Con1). Available samples at Day 4 with HCV RNA ≥ 1000 IU/mL were analyzed for resistance-associated variants using population and clonal sequencing and were compared with the baseline sequences to assess amino acid changes. Phenotypic resistance to ABT-450 at Day 4 was assessed by calculating the fold difference in the EC50 compared with the EC50 for the corresponding baseline sample. The number of participants with variants at resistance-associated amino acid positions and phenotypic resistance are presented. (NCT01074008)
Timeframe: Baseline and Day 4

,,
Interventionparticipants (Number)
Baseline Variants in NS3Baseline Resistance to ABT-450 >10-foldDay 4 Variants in NS3 (n=3, 2, 3)Day 4: Resistance to ABT-450 >10-fold (n=3, 2, 3)
ABT-450/r (100/100 mg) Once Daily (QD) + pegIFN/RBV0021
ABT-450/r (200/100 mg) Once Daily (QD) + pegIFN/RBV0010
ABT-450/r (50/100 mg) Once Daily (QD) + pegIFN/RBV0021

[back to top]

Evolution of CD4 Count

The evolution of participants' CD4-positive (CD4+) T-lymphocyte counts after starting the lopinavir/ritonavir-containing regimen was to be assessed by measuring the number of CD4+ cells at baseline and each subsequent study visit. CD4+ count results are reported as the number of CD4+ cells per cubic millimeter (cmm). Study visits were to occur at approximately 3, 6, 12, and 18 months after starting treatment. The exact dates of each visit depended on the physician's judgment, so data are reported for Visits 1 through 4 rather than by month. (NCT01074931)
Timeframe: Month 3, 6, 12, 18

,
Interventioncells per cmm (Mean)
CD4+ count at BaselineCD4+ count at Visit 1CD4+ count at Visit 2CD4+ count at Visit 3CD4+ count at Visit 4
Lopinavir/Ritonavir: Treatment-experienced249.03285.96317.10378.56413.77
Lopinavir/Ritonavir: Treatment-naive101.00136.67200.57200.00239.00

[back to top]

Evolution of the HIV Viral Response

The protocol recommended that HIV viral load tests be performed at baseline and each study visit. Test results indicate the number of HIV-1 ribonucleic acid (RNA) copies per milliliter (mL). The number of participants who underwent testing and had detectable levels (greater than 50 copies/mL) or undetectable levels (less than 50 copies/mL) are presented by subgroup. Study visits were to occur at approximately 3, 6, 12, and 18 months after starting treatment. The exact dates of each visit depended on the physician's judgment, so data are reported for Visits 1 through 4 rather than by month. (NCT01074931)
Timeframe: Month 3, 6, 12, 18

,
Interventionparticipant (Number)
Viral load detectable at BaselineViral load detectable at Visit 1Viral load detectable at Visit 2Viral load detectable at Visit 3Viral load detectable at Visit 4Viral load undetectable at BaselineViral load undetectable at Visit 1Viral load undetectable at Visit 2Viral load undetectable at Visit 3Viral load undetectable at Visit 4
Lopinavir/Ritonavir: Treatment-experienced51312551522394725
Lopinavir/Ritonavir: Treatment-naive0000010010

[back to top]

Number of Participants Who Missed Doses, Interrupt or Discontinue Regimen, and Experience Changes in Dosage or of Combination Regimen

Visits were to occur at approximately 3, 6, 12, and 18 months after starting treatment. The frequency with which each participant forgot to take their medication since the last visit and discontinuations of treatment and the reasons were documented at each visit and are summarized. The number of participants changing from lopinavir/ritonavir soft gel capsule to tablet are also presented. The exact dates of each visit depended on the physician's judgment, so data are reported for Visits 1 through 4 rather than by month. Note: participants may have had multiple missed doses or therapy changes. (NCT01074931)
Timeframe: Month 3, 6, 12, 18

InterventionParticipants (Number)
Total Missed Dosesa) Missed doses reported at Visit 1b) Missed doses reported at Visit 2c) Missed doses reported at Visit 3d) Missed doses reported at Visit 4Discontinued lopinavir/ritonavir therapya) Discontinued due to serious adverse eventb) Discontinued due to economic reasonsInterrupted lopinavir/ritonavir therapya) Interrupted due to adverse eventb) Required treatment with prohibited medicationc) Away because of workTherapy change: Switch from capsule to tableta) Switch from capsule to tablet at Visit 1b) Switch from capsule to tablet at Visit 2c) Switch from capsule to tablet at Visit 3d) Switch from capsule to tablet at Visit 4
Lopinavir/Ritonavir Group499131710651421151351240

[back to top]

Evolution of the Tolerance Issues

At each study visit, treating physicians evaluated participants and used their clinical judgment to determine if they were tolerating the lopinavir/ritonavir-containing regimen. Study visits were to occur at approximately 3, 6, 12, and 18 months after starting treatment. The exact dates of each visit depended on the physician's judgment, so data are reported for Visits 1 through 4 rather than by month. (NCT01074931)
Timeframe: Month 3, 6, 12, 18

InterventionParticipants (Number)
Lopinavir/ritonavir not tolerated at Visit 1Lopinavir/ritonavir not tolerated at Visit 2Lopinavir/ritonavir not tolerated at Visit 3Lopinavir/ritonavir not tolerated at Visit 4
Lopinavir/Ritonavir Group2000

[back to top]

Adverse Events Observed and Development of Lipodystrophy Lesion and Their Locations

The types of adverse events reported are summarized. The presence of lipodystrophy (abnormal body fat distribution) and its location was to be recorded. However, due to an oversight, there was not a place to record the location of lipodystrophy on the case report form. Doctors used clinical judgment to rate lipodystrophy in treatment-experienced participants. Study visits were to occur at approximately 3, 6, 12, and 18 months after starting treatment. The exact dates of each visit depended on the physician's judgment, so data are reported for Visits 1 through 4 rather than by month. (NCT01074931)
Timeframe: Month 3, 6, 12, 18

InterventionParticipants (Number)
Non-serious adverse eventsSerious adverse eventsLipodystrophy: Visit 1 evaluationLipodystrophy: Visit 2 evaluationc) WorsenedLipodystrophy: Visit 3 evaluationb) UnchangedLipodystrophy: Visit 4 evaluationa) Improved
Lopinavir/Ritonavir Group189024651888782878790849750

[back to top]

The Duration on Treatment Until Development of an Adverse Event Leading to Treatment Discontinuation or Until Escape From Treatment

As so few participants withdrew from lopinavir/ritonavir treatment, durations of lopinavir/ritonavir therapy required for 25 percent, 50 percent and 75 percent of participants could not be established. The numbers of participants in each subgroup who discontinued from treatment due to an adverse event are presented. (NCT01074931)
Timeframe: Month 3, 6, 12, 18

InterventionParticipants (Number)
Lopinavir/Ritonavir Group: Treatment-naive1
Lopinavir/Ritonavir Group: Treatment-experienced5

[back to top]

Mean Number of Human Immunodeficiency Virus (HIV) Ribonucleic Acid (RNA) Copies Per Milliliter (mL) Using a Logarithmic (Base 10) Transformation at Each Visit

Number of HIV RNA copies per mL is presented by the mean per visit for patients that were naive to previous antiretroviral treatment and those that were not. HIV-RNA data reported as < 400 copies/mL were considered 399 copies/mL in calculations. The mean and standard deviation of HIV-RNA levels were thus calculated after logarithmic (base 10) transformation (log10 399 is 2.6). Only observed cases were included in analyses; no data were imputed. n = xx, xx is the number of treatment-naive, treatment-experienced participants who had CD4+ T-cell counts available for analysis at each study visit. (NCT01076972)
Timeframe: Baseline (Month 0), every 3 months thereafter up to Month 12 and every year thereafter up to Year 8 (Month 96) during the course of the survey period

,
Interventioncopies/mL (Mean)
Baseline (Month 0 [n = 416, 418])Month 3 (n = 315, 280)Month 6 (n = 288, 253)Month 9 (n = 224, 238)Month 12 (Year 1 [n = 203, 230])Year 2 (n = 145, 190)Year 3 (n = 107, 147)Year 4 (n = 70, 99)Year 5 (n = 39, 72)Year 6 (n = 25, 41)Year 7 (n = 3, 17)Year 8 (n = 0, 3)
Lopinavir/Ritonavir: Treatment-experienced3.52.82.82.82.82.72.72.72.82.62.82.6
Lopinavir/Ritonavir: Treatment-Naive4.92.72.72.62.72.72.62.62.72.72.6NA

[back to top]

Cluster of Differentiation 4 Lymphocyte Count (CD4)

The evolution of patients' CD4-positive (CD4+) T-lymphocyte counts after starting treatment with Kaletra was assessed by measuring the number of CD4+ cells at baseline and each subsequent study visit. CD4+ counts are reported as the number of CD4+ cells per cubic millimeter (cmm) and presented by the mean at each visit. Only observed cases were included in analyses; no data were imputed. n = xx, xx is the number of patients naive to previous antiretroviral treatment and those that were not who had CD4+ T-cell counts available for analysis at each study visit. (NCT01076972)
Timeframe: Baseline (Month 0), every 3 months thereafter up to Month 12 and every year thereafter up to Year 8 (Month 96) during the course of the survey period

,
Interventioncells per cubic millimeter (Mean)
Baseline (Month 0 [n = 416, 420])Month 3 (n = 315, 283)Month 6 (n = 288, 252)Month 9 (n = 223, 238)Month 12 (Year 1 [n = 201, 233])Year 2 (n = 146, 190)Year 3 (n = 106, 150)Year 4 (n = 69, 99)Year 5 (n = 40, 73)Year 6 (n = 25, 42)Year 7 (n = 3, 17)Year 8 (n = 0, 3)
Lopinavir/Ritonavir: Treatment-Experienced290.6342.9366.4385.0410.4437.4481.5526.6496.2569.7597.7493.7
Lopinavir/Ritonavir: Treatment-Naive125.0257.2275.9311.1329.9419.4455.6475.2535.1577.1602.0NA

[back to top]

Number of Patients Included in Each Center for Disease Control and Prevention (CDC) Classification Category for HIV-infected Adults and Adolescents

Number of patients in each CDC category at Baseline (last assessment within 30 days prior to first dose of Kaletra) and after treatment. CDC categories defined as: Category A (asymptomatic acute HIV infection), Category B (symptomatic HIV infection; not Categories A and C), Category C (acquired immunodeficiency syndrome [AIDS] indicator status), Class P-0 (children not confirmed for HIV infection), Class P-1 (children with asymptomatic HIV infection), or Class P-2 (children with symptomatic HIV infection). (NCT01076972)
Timeframe: Baseline (Month 0) and following last treatment dose during the course of the survey period

,,,,
Interventionparticipants (Number)
Category A after lopinavir/ritonavir treatmentCategory B after lopinavir/ritonavir treatmentCategory C after lopinavir/ritonavir treatmentCategory P-0 after lopinavir/ritonavir treatmentCategory P-1 after lopinavir/ritonavir treatmentCategory P-2 after lopinavir/ritonavir treatmentCategory unknown after treatment
Lopinavir/Ritonavir: Baseline Category A20626000164
Lopinavir/Ritonavir: Baseline Category B032200037
Lopinavir/Ritonavir: Baseline Category C00191000132
Lopinavir/Ritonavir: Baseline Category P-20000001
Lopinavir/Ritonavir: Baseline Category Unknown1032286010199

[back to top]

Total Number of Patients With Adverse Drug Reactions

"Number of patients with adverse drug reactions, defined as adverse events for which the causal relationship with Kaletra was something other than not related by the investigator (i.e., probable, possible, or unclear), that occurred in ≥ 5% of patients. Adverse drug reactions are reported by preferred term and inclusive of all those reported at each visit. Although a patient may experience a particular preferred term more than once, each patient was counted only once for each preferred term." (NCT01076972)
Timeframe: During the course of the survey period up to Year 8

Interventionparticipants (Number)
Any adverse drug reactionHypertriglyceridaemiaHyperlipidaemiaDiarrhoeaNauseaBlood triglycerides increased
Lopinavir/Ritonavir Group649672111307299

[back to top]

Number of Patients With Adverse Drug Reactions (ADRs)

"The number of patients (mothers and infants) with adverse drug reactions, defined as adverse events for which the causal relationship with Kaletra was something other than not related by the investigator (i.e., probable, possible, or unclear). ADRs are reported by preferred term and inclusive of all those reported at any visit. Although a patient may experience a particular preferred term more than once, each patient was counted only once for each preferred term." (NCT01076985)
Timeframe: During pregnancy and for one year after birth

,
Interventionparticipants (Number)
AnemiaHypercholesterolemiaHyperlipidemiaDiarrheaNauseaRashAbortion missedPremature laborThreatened laborNeutropeniaHypoglycemiaCardio-respiratory arrestApneaNeonatal respiratory distress syndromeTransient tachypnea of the newbornSmall for dates babyPyrexia
Infants50000100011111111
Lopinavir/Ritonavir21112111100000000

[back to top]

Percentage of Patients With HIV-1 RNA 50 to <200 Copies/ml

All 3 protocols recommended that HIV viral load tests be performed at Baseline and each study visit. Study visits were to occur at approximately Weeks 4, 12, and 24, followed by 12-week intervals up to Week 144 in therapy-naive participants and up to Week 240 in the pre-treated and non-B subtype groups. The percentage of participants with HIV-1 RNA levels of 50 to less than 200 copies/mL at each time point is presented by subgroup. (NCT01083810)
Timeframe: Baseline, Week 4, 12, 24, followed by 12-week intervals up to 144/240 weeks

,,
InterventionPercentage of participants (Number)
BaselineWeek 4Week 12Week 24Week 36Week 48Week 60Week 72Week 84Week 96Week 108Week 120Week 132Week 144Week 156Week 168Week 180Week 192Week 204Week 216Week 228Week 240
Non-B0142518105636104400008000250
Pre-treated5151520912101713983130005561000
Therapy-naive117322218101359614530NANANANANANANANA

[back to top]

Change in Absolute CD4 Cell Count [CD4+ Cells/µL]

The evolution of participants' CD4-positive (CD4+) T-lymphocyte counts after starting the lopinavir/ritonavir-containing regimen was to be assessed by measuring the number of CD4+ cells at baseline and each subsequent study visit. Study visits were to occur at approximately Weeks 4, 12, 24, followed by 12-week intervals up to Week 144 in therapy-naive participants and up to Week 240 in the pre-treated and non-B subtype groups. CD4+ cell count results are reported as the change from Baseline in the absolute number of CD4+ cells per microliter. (NCT01083810)
Timeframe: Baseline, Week 4, 12, 24, followed by 12-week intervals up to 144/240 weeks

,,
InterventionCD4+ cells/µL (Mean)
BaselineWeek 4Week 12Week 24Week 36Week 48Week 60Week 72Week 84Week 96Week 108Week 120Week 132Week 144Week 156Week 168Week 180Week 192Week 204Week 216Week 228Week 240
Non-B013192128148204222259226239255275325320404426422474420395399542
Pre-treated06898106144114144179181192223217213251220190174192131210208179
Therapy-naive0140146193211240266248295296329316313292NANANANANANANANA

[back to top]

Percentage of Patients With HIV-1 RNA <50 Copies/ml

All 3 protocols recommended that HIV viral load tests be performed at Baseline and each study visit. Study visits were to occur at approximately Weeks 4, 12, and 24, followed by 12-week intervals up to Week 144 in therapy-naive participants and up to Week 240 in the pre-treated and non-B subtype groups. The percentage of participants with HIV-1 ribonucleic acid (RNA) less than 50 copies/mL at each time point is presented by subgroup. (NCT01083810)
Timeframe: Baseline, Week 4, 12, 24, followed by 12-week intervals up to 144/240 weeks

,,
InterventionPercentage of participants (Number)
BaselineWeek 4Week 12Week 24Week 36Week 48Week 60Week 72Week 84Week 96Week 108Week 120Week 132Week 144Week 156Week 168Week 180Week 192Week 204Week 216Week 228Week 240
Non-B054274768079838877878895100100918310010010075100
Pre-treated3263858606765626658666352654852534541504438
Therapy-naive07446975837889879081858788NANANANANANANANA

[back to top]

Percentage of Patients With HIV-1 RNA 200 to <500 Copies/ml

All 3 protocols recommended that HIV viral load tests be performed at Baseline and each study visit. Study visits were to occur at approximately Weeks 4, 12, and 24, followed by 12-week intervals up to Week 144 in therapy-naive participants and up to Week 240 in the pre-treated and non-B subtype groups. The percentage of participants with HIV-1 RNA levels of 200 to less than 500 copies/mL at each time point is presented by subgroup. (NCT01083810)
Timeframe: Baseline, Week 4, 12, 24, followed by 12-week intervals up to 144/240 weeks

,,
InterventionPercentage of participants (Number)
BaselineWeek 4Week 12Week 24Week 36Week 48Week 60Week 72Week 84Week 96Week 108Week 120Week 132Week 144Week 156Week 168Week 180Week 192Week 204Week 216Week 228Week 240
Non-B0191746139837000000800000
Pre-treated3121569066570901010100100066
Therapy-naive0181231120200230NANANANANANANANA

[back to top]

Percentage of Patients With HIV-1 RNA >500 Copies/ml

All 3 protocols recommended that HIV viral load tests be performed at Baseline and each study visit. Study visits were to occur at approximately Weeks 4, 12, and 24, followed by 12-week intervals up to Week 144 in therapy-naive participants and up to Week 240 in the pre-treated and non-B subtype groups. The percentage of participants with more than 500 HIV-1 RNA copies/mL at each time point is presented by subgroup. (NCT01083810)
Timeframe: Baseline, Week 4, 12, 24, followed by 12-week intervals up to 144/240 weeks

,,
InterventionPercentage of participants (Number)
BaselineWeek 4Week 12Week 24Week 36Week 48Week 60Week 72Week 84Week 96Week 108Week 120Week 132Week 144Week 156Week 168Week 180Week 192Week 204Week 216Week 228Week 240
Non-B10063154836637985009000000
Pre-treated89463217232118151626262636264338424053405056
Therapy-naive995913666752457813NANANANANANANANA

[back to top]

Number of Patients With Virus That Develop Mutations Conferring Resistance to Lopinavir/Ritonavir, NRTIs or NNRTIs

Standard genotypic resistance assays were developed for HIV-1 viral load levels greater than 500 to 1000 copies per milliliter (mL). All 3 protocols recommended this testing be done at Baseline prior to lopinavir/ritonavir therapy and (if possible) in cases of virologic failure. The exact timing varied and depended on whether there was an adequate viral load and physician clinical judgment. Participants with resistance to lopinavir/ritonavir, nucleoside reverse transcriptase inhibitors (NRTI) or non-nucleoside reverse transcriptase inhibitors (NNRTI) at Baseline and follow-up are reported. (NCT01083810)
Timeframe: Baseline and at any timepoint where testing is possible

,,
InterventionParticipants (Number)
Genotypic resistance testing performed at BaselineComplete resistance testing results at Baseline>Resistance to lopinavir/ritonavir at Baseline>Partial resistance to NRTI at Baseline>Partial resistance to NNRTI at BaselineUnderwent resistance testing at follow-up>Resistance to lopinavir/ritonavir*>>Resistance to NRTIs>>Resistance to NNRTIs*No baseline results avail for this participant
Non-B55550002000NA
Pre-treated686820021101
Therapy-naive1371220502000NA

[back to top]

Grade 3-5 Toxicity as Assessed by NCI CTC v3.0

Number of participants with adverse events grades 3-5. For a detailed list of adverse events see the adverse event module. (NCT01095094)
Timeframe: at 6 months from start of treatment

Interventionparticipants (Number)
Arm I7

[back to top]

Progression-free Survival

Number of patients that remained disease free at 6 months from start of treatment. (NCT01095094)
Timeframe: At 6 months

Interventionparticipants (Number)
Arm I4

[back to top]

Time to Maximum Observed Concentration (Tmax) of Atazanavir and Ritonavir

(NCT01099579)
Timeframe: At Week 2 at Hour 0 predose and at Hours 1.5, 2.5, 4, 6, 8, 12, and 24 postdose

,,
InterventionHours (Median)
Tmax AtazanavirTmax Ritonavir
Atazanavir Powder, 150 mg/Ritonavir Oral Solution, 80 mg1.581.8
Atazanavir Powder, 200 mg/Ritonavir Oral Solution, 80 mg1.972.9
Atazanavir Powder, 250 mg/Ritonavir Oral Solution, 80 mg4.04.0

[back to top]

Maximum Observed Concentration (Cmax) and Minimum Observed Concentration (Cmin) of Atazanavir and Ritonavir

(NCT01099579)
Timeframe: At Week 2 at Hour 0 predose and at Hours 1.5, 2.5, 4, 6, 8, 12, and 24 postdose

,,
Interventionng/mL (Geometric Mean)
Atazanavir CmaxAtazanavir CminRitonavir Cmax (n=19, 18, 15)Ritonavir Cmin (n=18, 16, 15)
Atazanavir Powder, 150 mg/Ritonavir Oral Solution, 80 mg4131336291941.8
Atazanavir Powder, 200 mg/Ritonavir Oral Solution, 80 mg51975722634143
Atazanavir Powder, 250 mg/Ritonavir Oral Solution, 80 mg6172698183851.0

[back to top]

Mean Change From Baseline in HIV RNA Levels at Week 48 by Prior Antiretroviral (ARV) Treatment Status

(NCT01099579)
Timeframe: From Baseline to Week 48

InterventionLog10 c/mL (Mean)
ARV-experienced-2.53
ARV-naive-2.81

[back to top]

Electrocardiogram Changes From Baseline in PR Interval, QTC Bazett, and QTC Fridericia at Week 48

Electrocardiogram parameters were measured at baseline for QTC Bazett, QTC Fridericia, and PR interval. The mean change from baseline at week 48 is reported by arm in milliseconds. (NCT01099579)
Timeframe: From Baseline to Week 48

,,
InterventionMilliseconds (Mean)
PR IntervalQTC BazettQTC Fridericia
Atazanavir Powder, 150 mg/Ritonavir Oral Solution, 80 mg4.91.77.9
Atazanavir Powder, 200 mg/Ritonavir Oral Solution, 80 mg12.0-3.213.2
Atazanavir Powder, 250 mg/Ritonavir Oral Solution, 80 mg6.2-4.24.8

[back to top]

Percentage of Participants With HIV RNA Levels <50 c/mL and <400 c/mL at Week 48 by Treatment/Weight

The definition of virologic success included HIV RNA levels <50 c/mL or 400 c/mL at the Week 48 analysis window. . (NCT01099579)
Timeframe: At Week 48

,,
InterventionPercentage of participants (Number)
HIV RNA levels <50 c/mLHIV RNA levels <400 c/mL
Atazanavir Powder, 150 mg/Ritonavir Oral Solution, 80 mg47.666.7
Atazanavir Powder, 200 mg/Ritonavir Oral Solution, 80 mg68.473.7
Atazanavir Powder, 250 mg/Ritonavir Oral Solution, 80 mg71.485.7

[back to top]

Percentage of Participants With HIV RNA Levels <50 c/mL and <400 c/mL at Week 48 by Prior Antiretroviral (ARV) Treatment Status

The definition of virologic success included HIV RNA levels <50 c/mL or <400 c/mL at the Week 48 analysis. (NCT01099579)
Timeframe: From Day 1 to Week 48

,
InterventionPercentage of participants (Number)
HIV RNA levels <50 c/mLHIV RNA levels <400 c/mL
ARV-experienced56.365.6
ARV-naive68.286.4

[back to top]

Number of Participants With Laboratory Test Results With Worst Toxicity of Grade 3-4

ALT=alanine aminotransferase; SGPT=serum glutamic-pyruvic transaminase; AST=aspartate aminotransferase; SGOT=serum glutamic-oxaloacetic transaminase; ULN=upper limit of normal. Grading by the National Institute of Health Division of AIDs and World Health Organization criteria. Hemoglobin (g/dL): Grade (Gr)1=9.5-11.0; Gr 2=8.0-9.4; Gr 3=6.5-7.9; Gr 4=<6.5. Neutrophils, absolute (/mm^3): Gr 1=>=1000-<1500; Gr 2= >=750-<1000; Gr 3=>=500-<750; Gr 4=<500. ALT/SGPT (*ULN): Gr 1=1.25-2.5; Gr 2=2.6-5; Gr 3=5.1-10; Gr 4=>10. AST/SGOT (*ULN): Gr 1=1.25-2.5; Gr 2=2.6-5; Gr 3=5.1-10; Gr 4=>10. Alkaline phosphatase(*ULN): Gr 1=1.25-2.5; Gr 2=2.6-5: Gr 3=5.1-10; Gr 4=>10. Total bilirubin (*ULN): Gr 1=1.1-1; Gr 2=1.6-2.5; Gr 3=2.6-5; Gr 4=>5. Amylase (*ULN): Gr 1=1.10-39; Gr 2=1.40-2; Gr 3=2.10-5.0; Gr 4=>5.0. Lipase (*ULN): Gr 1=1.10-1.39: Gr 2=1.40-2; Gr 3=2.10-5.0; Gr 4=>5.0. Uric acid (mg/dL): Gr 1=7.5-10.0; Gr 2=10.1-12.0; Gr 3=12.1-15.0; Gr 4=>15. (NCT01099579)
Timeframe: After Day 1 to Week 48

,,
InterventionParticipants (Number)
Hemoglobin (n=20, 17, 15)Neutrophils, absolute (n=20, 17, 15)ALT/SGPT (n=20, 18, 15)AST/SGOT (n=20, 18, 15)Alkaline phosphatase (n=20, 18, 15)Total bilirubin (n=20, 18, 15)Amylase (n=20, 18, 15)Lipase (n=20, 18, 15)Uric acid n=20, 18, 15)
Atazanavir Powder, 150 mg/Ritonavir Oral Solution, 80 mg235102800
Atazanavir Powder, 200 mg/Ritonavir Oral Solution, 80 mg320010510
Atazanavir Powder, 250 mg/Ritonavir Oral Solution, 80 mg001003111

[back to top]

Number of Participants Who Acquired Phenotypic Resistance to Atazanavir or Atazanovir/Ritonavir

Criteria for resistance testing= meeting at least 1 of the following: <1 log10 drop from baseline in HIV RNA level by Week 16 and confirmed by a second HIV RNA level; an HIV RNA level >200 copies/mL after Week 24, confirmed by a second HIV RNA level; repeated HIV RNA levels ≥50 copies/mL after Week 48; an HIV RNA level ≥400 copies/mL confirmed by a second HIV RNA level of ≥400 copies/mL at any time in a participant who had previously achieved a plasma HIV RNA level <50 copies/mL; or discontinued due to lack of efficacy. Virologic failure was defined as an incomplete virologic response to therapy or as a viral rebound after the achievement of virologic suppression. The phenotypic resistance to a drug is defined as a fold change (ie, ratio of the 50% inhibitory concentration [IC50] of the clinical isolate to the IC50 of the reference strain) greater than the cut-off for reduced susceptibility. (NCT01099579)
Timeframe: After Day 1 to Week 48

,
InterventionParticipants (Number)
AtazanavirRitonavir
ARV-experienced00
ARV-naive00

[back to top]

CD4 Cell Count Changes From Baseline at Week 48 by Prior Antiretroviral (ARV) Treatment Status

(NCT01099579)
Timeframe: From Baseline to Week 48

InterventionCells/mm^3 (Mean)
ARV-experienced437.9
ARV-naive352.1

[back to top]

CD4 Cell Count Changes From Baseline at Week 48 by Treatment/Weight

(NCT01099579)
Timeframe: From Baseline to Week 48

InterventionCells/mm^3 (Mean)
Atazanavir Powder, 150 mg/Ritonavir Oral Solution, 80 mg550.1
Atazanavir Powder, 200 mg/Ritonavir Oral Solution, 80 mg225.3
Atazanavir Powder, 250 mg/Ritonavir Oral Solution, 80 mg373.8

[back to top]

Mean CD4 Percent Changes From Baseline at Week 48 by Antiretroviral (ARV) Treatment Status

(NCT01099579)
Timeframe: From Baseline to Week 48

InterventionPercentage of lymphocytes (Mean)
ARV-experienced4.3
ARV-naive9.8

[back to top]

Mean CD4 Percent Changes From Baseline at Week 48 by Treatment/Weight

(NCT01099579)
Timeframe: From Baseline to Week 48

InterventionPercentage of lymphocytes (Mean)
Atazanavir Powder, 150 mg/Ritonavir Oral Solution, 80 mg6.1
Atazanavir Powder, 200 mg/Ritonavir Oral Solution, 80 mg7.3
Atazanavir Powder, 250 mg/Ritonavir Oral Solution, 80 mg8.8

[back to top]

Mean Change From Baseline in HIV RNA Levels at Week 48 by Treatment/Weight

Participants who received at least 1 dose of atazanavir (ATV) and had an HIV RNA measurement on ATV powder at did not switch to the capsule formulation before Week 48 (NCT01099579)
Timeframe: From Baseline to Week 48

InterventionLog10 c/mL (Mean)
Atazanavir Powder, 150 mg/Ritonavir Oral Solution, 80 mg-2.61
Atazanavir Powder, 200 mg/Ritonavir Oral Solution, 80 mg-2.93
Atazanavir Powder, 250 mg/Ritonavir Oral Solution, 80 mg-2.40

[back to top]

Number of Participants With Centers for Disease Control (CDC) Class C AIDS Events

CDC Class C events are AIDS-defining events that include recurrent bacterial pneumonia (>=2 episodes in 12 months); candidiasis of the bronchi, trachea, lungs, or esophagus; invasive cervical carcinoma; disseminated or extrapulmonary coccidioidomycosis; extrapulmonary cryptococcosis; chronic intestinal cryptosporidiosis (>1 month); cytomegalovirus disease; HIV-related encephalopathy; herpes simplex: chronic ulcers, or bronchitis, pneumonitis, or esophagitis; disseminated or extrapulmonary histoplasmosis; chronic intestinal isosporiasis; Kaposi sarcoma; immunoblastic or primary brain Burkitt lymphoma; mycobacterium avium complex, kansasii, or tuberculosis; mycobacterium, other species; Pneumocystis carinii pneumonia; progressive multifocal leukoencephalopathy; Salmonella septicemia; recurrent toxoplasmosis of brain; HIV wasting syndrome (involuntary weight loss >10% of baseline body weight) with chronic diarrhea or chronic weakness and documented fever for ≥1 month. (NCT01099579)
Timeframe: From Day 1 to Week 48

InterventionParticipants (Number)
Atazanavir Powder, 150 mg/Ritonavir Oral Solution, 80 mg1
Atazanavir Powder, 200 mg/Ritonavir Oral Solution, 80 mg1
Atazanavir Powder, 250 mg/Ritonavir Oral Solution, 80 mg0

[back to top]

Apparent Total Body Clearance (CLT/F) of Atazanavir and Ritonavir

Calculated as dose divided by AUC(TAU). AUC(TAU)=area under the concentration-time curve in 1 dosing interval from time 0 to 24 hours post observed dose. (NCT01099579)
Timeframe: At Week 2

,,
InterventionL/h (Geometric Mean)
CLT/F AtazanavirCLT/F Ritonavir (n=19, 18, 15
Atazanavir Powder, 150 mg/Ritonavir Oral Solution, 80 mg4.614.59
Atazanavir Powder, 200 mg/Ritonavir Oral Solution, 80 mg3.983.90
Atazanavir Powder, 250 mg/Ritonavir Oral Solution, 80 mg4.075.87

[back to top]

Number of Participants With Death as Outcome, Serious Adverse Events (SAEs), Adverse Events (AEs) Leading to Discontinuation

AE=any new unfavorable symptom, sign, or disease or worsening of a preexisting condition that may not have a causal relationship with treatment. SAE=a medical event that at any dose results in death, persistent or significant disability/incapacity, or drug dependency/abuse; is life-threatening, an important medical event, or a congenital anomaly/birth defect; or requires or prolongs hospitalization. (NCT01099579)
Timeframe: From Day 1 to Week 48

,,
InterventionParticipants (Number)
DeathsSAEsAEs leading to discontinuation
Atazanavir Powder, 150 mg/Ritonavir Oral Solution, 80 mg054
Atazanavir Powder, 200 mg/Ritonavir Oral Solution, 80 mg021
Atazanavir Powder, 250 mg/Ritonavir Oral Solution, 80 mg040

[back to top]

Apparent Total Body Clearance Per Body Weight (CLT/F) Per Kilogram of Atazanavir and Ritonavir

Calculated as CLT/F divided by body weight (NCT01099579)
Timeframe: At Week 2

,,
InterventionL/h per kilogram (Geometric Mean)
CLT/F per kilogram AtazanavirCLT/F per kilogram Ritonavir (n=19, 18, 15)
Atazanavir Powder, 150 mg/Ritonavir Oral Solution, 80 mg0.650.65
Atazanavir Powder, 200 mg/Ritonavir Oral Solution, 80 mg0.320.32
Atazanavir Powder, 250 mg/Ritonavir Oral Solution, 80 mg0.240.35

[back to top]

Area Under the Concentration Curve (in 1 Dosing Interval From Time 0 to 24 Hours Post Observed Dose) (AUC[TAU])of Atazanavir and Ritonavir

(NCT01099579)
Timeframe: At Week 2 at Hour 0 predose and at Hours 1.5, 2.5, 4, 6, 8, 12, and 24 postdose

,,
Interventionng*h/mL (Geometric Mean)
AUC(TAU) AtazanavirAUC(TAU) Ritonavir (n=19, 18, 15)
Atazanavir Powder, 150 mg/Ritonavir Oral Solution, 80 mg3250317439
Atazanavir Powder, 200 mg/Ritonavir Oral Solution, 80 mg5030520510
Atazanavir Powder, 250 mg/Ritonavir Oral Solution, 80 mg6148513640

[back to top]

The Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 48

(NCT01106586)
Timeframe: Week 48

Interventionpercentage of participants (Number)
Stribild91.5
ATV/r + FTC/TDF88.5

[back to top]

The Percentage of Participants With Virologic Success Using the Food and Drug Administration (FDA)-Defined Snapshot Analysis as Determined by the Achievement of HIV-1 Ribonucleic Acid (RNA) < 50 Copies/mL at Week 48

(NCT01106586)
Timeframe: Week 48

Interventionpercentage of participants (Number)
Stribild89.5
ATV/r + FTC/TDF86.8

[back to top]

The Percentage of Participants Achieving and Maintaining Confirmed HIV-1 RNA < 50 Copies/mL at Week 48 Using the FDA-defined Time to Loss of Virologic Response (TLOVR) Algorithm

(NCT01106586)
Timeframe: Week 48

Interventionpercentage of participants (Number)
Stribild86.1
ATV/r + FTC/TDF84.8

[back to top]

The Percentage of Participants With Virologic Success Using the FDA-Defined Snapshot Analysis as Determined by the Achievement of HIV-1 RNA < 50 Copies/mL at Week 96

(NCT01106586)
Timeframe: Week 96

Interventionpercentage of participants (Number)
Stribild83.3
ATV/r + FTC/TDF82.3

[back to top]

The Percentage of Participants With Virologic Success Using the FDA-Defined Snapshot Analysis as Determined by the Achievement of HIV-1 RNA < 50 Copies/mL at Week 192

(NCT01106586)
Timeframe: Week 192

Interventionpercentage of participants (Number)
Stribild78.4
ATV/r + FTC/TDF73.1

[back to top]

The Percentage of Participants With Virologic Success Using the FDA-Defined Snapshot Analysis as Determined by the Achievement of HIV-1 RNA < 50 Copies/mL at Week 144

(NCT01106586)
Timeframe: Week 144

Interventionpercentage of participants (Number)
Stribild77.6
ATV/r + FTC/TDF74.6

[back to top]

The Change From Baseline in Cluster Determinant 4 (CD4) Cell Count at Weeks 48, 96, 144, and 192

Change = value of the relevant time point minus the baseline value (NCT01106586)
Timeframe: Baseline; Weeks 48, 96, 144, and 192

,
Interventioncells/µL (Mean)
Change at Wk 48 (Stribild, n=334; ATV/r, n=321)Change at Wk 96 (Stribild, n=317; ATV/r, n=315)Change at Wk 144 (Stribild, n=297; ATV/r, n=286)Change at Wk 192 (Stribild, n=69; ATV/r, n=72)
ATV/r + FTC/TDF211261293340
Stribild207256280338

[back to top]

Viral Failure

Probability of viral failure defined as >= 2 HIV RNA measurements >1000 copies/ml using survival analysis by 48 weeks post-randomization. (NCT01146873)
Timeframe: 48 weeks

Interventionprobability of viral failure (Mean)
Group 1: Lopinavir/Ritonavir (LPV/r)0.020
Group 2: Efavirenz (EFV)0.027

[back to top]

Viral Rebound

Probability of viral rebound defined as >=1 HIV RNA measurements >50 copies/ml using survival analysis by 48 weeks post-randomization. (NCT01146873)
Timeframe: 48 weeks

Interventionprobability of viral rebound (Mean)
Group 1: Lopinavir/Ritonavir (LPV/r)0.284
Group 2: Efavirenz (EFV)0.176

[back to top]

CD4 Cell Percentage at 48 Weeks After Randomization

CD4 Cell Percentage at 48 Weeks After Randomization (NCT01146873)
Timeframe: 48 weeks

Interventionpercentage of cells (Mean)
Group 1: Lopinavir/Ritonavir (LPV/r)34.7
Group 2: Efavirenz (EFV)37.5

[back to top]

Highest Grade ALT After Randomization

Highest grade ALT after randomization. Grading was determined based on the Division of AIDS (2004) Toxicity Tables to grade adverse reactions. Grading scale: 0 (none), 1 (mild), 2 (moderate), 3 (severe), 4 (potentially life-threatening). (NCT01146873)
Timeframe: through 48 weeks post randomization

,
Interventionnumber of participants (Number)
Grade 0Grade 1Grade 2Grade 3Grade 4
Group 1: Lopinavir/Ritonavir (LPV/r)1398010
Group 2: Efavirenz (EFV)120161031

[back to top]

Percentage of Participants With Elevated Total Cholesterol, Elevated LDL, Abnormal HDL, or Abnormal Triglycerides at 40 Weeks After Randomization

Percentage of participants with elevated total cholesterol, elevated LDL, abnormal HDL, or abnormal triglycerides at 40 weeks after randomization (NCT01146873)
Timeframe: 40 weeks

,
Interventionpercentage of participants (Number)
Elevated total cholesterolElevated LDLAbnormal HDLAbnormal triglycerides
Group 1: Lopinavir/Ritonavir (LPV/r)24.818.64.822.8
Group 2: Efavirenz (EFV)13.39.84.210.5

[back to top]

Viral Load

The decision to perform HIV-1 ribonucleic acid (RNA) tests to monitor participants' viral load was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 108 are presented. (NCT01153269)
Timeframe: Week 108

Interventionlog10 copies/mL (Mean)
HIV-infected Participants With Hepatitis Co-infection1.6

[back to top]

Viral Load

The decision to perform HIV-1 ribonucleic acid (RNA) tests to monitor participants' viral load was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 12 are presented. (NCT01153269)
Timeframe: Week 12

Interventionlog10 copies/mL (Mean)
HIV-infected Participants With Hepatitis Co-infection2.0

[back to top]

Viral Load

The decision to perform HIV-1 ribonucleic acid (RNA) tests to monitor participants' viral load was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 120 are presented. (NCT01153269)
Timeframe: Week 120

Interventionlog10 copies/mL (Mean)
HIV-infected Participants With Hepatitis Co-infection1.6

[back to top]

Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters

The decision to perform aspartate aminotransferase (AST) and alanine aminotransferase (ALT) laboratory tests to monitor participants' liver function was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Baseline are presented. (NCT01153269)
Timeframe: Baseline

InterventionU/liter (Mean)
HIV-infected Participants With Hepatitis Co-infection: AST64
HIV-infected Participants With Hepatitis Co-infection: ALT69

[back to top]

Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters

The decision to perform aspartate aminotransferase (AST) and alanine aminotransferase (ALT) laboratory tests to monitor participants' liver function was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 72 are presented. (NCT01153269)
Timeframe: Week 72

InterventionU/liter (Mean)
HIV-infected Participants With Hepatitis Co-infection: AST51
HIV-infected Participants With Hepatitis Co-infection: ALT50

[back to top]

Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters

The decision to perform aspartate aminotransferase (AST) and alanine aminotransferase (ALT) laboratory tests to monitor participants' liver function was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 84 are presented. (NCT01153269)
Timeframe: Week 84

InterventionU/liter (Mean)
HIV-infected Participants With Hepatitis Co-infection: AST55
HIV-infected Participants With Hepatitis Co-infection: ALT49

[back to top]

Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters

The decision to perform aspartate aminotransferase (AST) and alanine aminotransferase (ALT) laboratory tests to monitor participants' liver function was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 96 are presented. (NCT01153269)
Timeframe: Week 96

InterventionU/liter (Mean)
HIV-infected Participants With Hepatitis Co-infection: AST63
HIV-infected Participants With Hepatitis Co-infection: ALT63

[back to top]

CD4 Cell Count

The decision to perform laboratory tests to determine participants' CD4-positive (CD4+) T-lymphocyte counts was left to the treating physician's clinical judgment. The mean and standard deviation for those tested at Baseline are presented. (NCT01153269)
Timeframe: Baseline

InterventionCD4+ cells/μL (Mean)
HIV-infected Participants With Hepatitis Co-infection288

[back to top]

CD4 Cell Count

The decision to perform laboratory tests to determine participants' CD4-positive (CD4+) T-lymphocyte counts was left to the treating physician's clinical judgment. The mean and standard deviation for those tested at Week 108 are presented. (NCT01153269)
Timeframe: Week 108

InterventionCD4+ cells/μL (Mean)
HIV-infected Participants With Hepatitis Co-infection581

[back to top]

CD4 Cell Count

The decision to perform laboratory tests to determine participants' CD4-positive (CD4+) T-lymphocyte counts was left to the treating physician's clinical judgment. The mean and standard deviation for those tested at Week 12 are presented. (NCT01153269)
Timeframe: Week 12

InterventionCD4+ cells/μL (Mean)
HIV-infected Participants With Hepatitis Co-infection378

[back to top]

CD4 Cell Count

The decision to perform laboratory tests to determine participants' CD4-positive (CD4+) T-lymphocyte counts was left to the treating physician's clinical judgment. The mean and standard deviation for those tested at Week 120 are presented. (NCT01153269)
Timeframe: Week 120

InterventionCD4+ cells/μL (Mean)
HIV-infected Participants With Hepatitis Co-infection565

[back to top]

Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters

The decision to perform aspartate aminotransferase (AST) and alanine aminotransferase (ALT) laboratory tests to monitor participants' liver function was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 108 are presented. (NCT01153269)
Timeframe: Week 108

InterventionU/liter (Mean)
HIV-infected Participants With Hepatitis Co-infection: AST47
HIV-infected Participants With Hepatitis Co-infection: ALT43

[back to top]

Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters

The decision to perform aspartate aminotransferase (AST) and alanine aminotransferase (ALT) laboratory tests to monitor participants' liver function was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 12 are presented. (NCT01153269)
Timeframe: Week 12

InterventionU/liter (Mean)
HIV-infected Participants With Hepatitis Co-infection: AST69
HIV-infected Participants With Hepatitis Co-infection: ALT85

[back to top]

Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters

The decision to perform aspartate aminotransferase (AST) and alanine aminotransferase (ALT) laboratory tests to monitor participants' liver function was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 120 are presented. (NCT01153269)
Timeframe: Week 120

InterventionU/liter (Mean)
HIV-infected Participants With Hepatitis Co-infection: AST56
HIV-infected Participants With Hepatitis Co-infection: ALT65

[back to top]

Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters

The decision to perform aspartate aminotransferase (AST) and alanine aminotransferase (ALT) laboratory tests to monitor participants' liver function was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 144 are presented. (NCT01153269)
Timeframe: Week 144

InterventionU/liter (Mean)
HIV-infected Participants With Hepatitis Co-infection: AST52
HIV-infected Participants With Hepatitis Co-infection: ALT53

[back to top]

Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters

The decision to perform aspartate aminotransferase (AST) and alanine aminotransferase (ALT) laboratory tests to monitor participants' liver function was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 24 are presented. (NCT01153269)
Timeframe: Week 24

InterventionU/liter (Mean)
HIV-infected Participants With Hepatitis Co-infection: AST78
HIV-infected Participants With Hepatitis Co-infection: ALT97

[back to top]

Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters

The decision to perform aspartate aminotransferase (AST) and alanine aminotransferase (ALT) laboratory tests to monitor participants' liver function was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 36 are presented. (NCT01153269)
Timeframe: Week 36

InterventionU/liter (Mean)
HIV-infected Participants With Hepatitis Co-infection: AST54
HIV-infected Participants With Hepatitis Co-infection: ALT75

[back to top]

Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters

The decision to perform aspartate aminotransferase (AST) and alanine aminotransferase (ALT) laboratory tests to monitor participants' liver function was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 4 are presented. (NCT01153269)
Timeframe: Week 4

InterventionU/liter (Mean)
HIV-infected Participants With Hepatitis Co-infection: AST85
HIV-infected Participants With Hepatitis Co-infection: ALT85

[back to top]

Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters

The decision to perform aspartate aminotransferase (AST) and alanine aminotransferase (ALT) laboratory tests to monitor participants' liver function was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 48 are presented. (NCT01153269)
Timeframe: Week 48

InterventionU/liter (Mean)
HIV-infected Participants With Hepatitis Co-infection: AST60
HIV-infected Participants With Hepatitis Co-infection: ALT79

[back to top]

Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters

The decision to perform aspartate aminotransferase (AST) and alanine aminotransferase (ALT) laboratory tests to monitor participants' liver function was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 60 are presented. (NCT01153269)
Timeframe: Week 60

InterventionU/liter (Mean)
HIV-infected Participants With Hepatitis Co-infection: AST55
HIV-infected Participants With Hepatitis Co-infection: ALT59

[back to top]

Viral Load

The decision to perform HIV-1 ribonucleic acid (RNA) tests to monitor participants' viral load was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 132 are presented. (NCT01153269)
Timeframe: Week 132

Interventionlog10 copies/mL (Mean)
HIV-infected Participants With Hepatitis Co-infection1.6

[back to top]

Viral Load

The decision to perform HIV-1 ribonucleic acid (RNA) tests to monitor participants' viral load was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 144 are presented. (NCT01153269)
Timeframe: Week 144

Interventionlog10 copies/mL (Mean)
HIV-infected Participants With Hepatitis Co-infection1.5

[back to top]

Viral Load

The decision to perform HIV-1 ribonucleic acid (RNA) tests to monitor participants' viral load was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 24 are presented. (NCT01153269)
Timeframe: Week 24

Interventionlog10 copies/mL (Mean)
HIV-infected Participants With Hepatitis Co-infection1.7

[back to top]

Viral Load

The decision to perform HIV-1 ribonucleic acid (RNA) tests to monitor participants' viral load was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 36 are presented. (NCT01153269)
Timeframe: Week 36

Interventionlog10 copies/mL (Mean)
HIV-infected Participants With Hepatitis Co-infection1.6

[back to top]

Viral Load

The decision to perform HIV-1 ribonucleic acid (RNA) tests to monitor participants' viral load was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 4 are presented. (NCT01153269)
Timeframe: Week 4

Interventionlog10 copies/mL (Mean)
HIV-infected Participants With Hepatitis Co-infection2.5

[back to top]

Viral Load

The decision to perform HIV-1 ribonucleic acid (RNA) tests to monitor participants' viral load was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 48 are presented. (NCT01153269)
Timeframe: Week 48

Interventionlog10 copies/mL (Mean)
HIV-infected Participants With Hepatitis Co-infection1.7

[back to top]

Viral Load

The decision to perform HIV-1 ribonucleic acid (RNA) tests to monitor participants' viral load was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 60 are presented. (NCT01153269)
Timeframe: Week 60

Interventionlog10 copies/mL (Mean)
HIV-infected Participants With Hepatitis Co-infection1.7

[back to top]

Viral Load

The decision to perform HIV-1 ribonucleic acid (RNA) tests to monitor participants' viral load was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 72 are presented. (NCT01153269)
Timeframe: Week 72

Interventionlog10 copies/mL (Mean)
HIV-infected Participants With Hepatitis Co-infection1.6

[back to top]

Viral Load

The decision to perform HIV-1 ribonucleic acid (RNA) tests to monitor participants' viral load was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 84 are presented. (NCT01153269)
Timeframe: Week 84

Interventionlog10 copies/mL (Mean)
HIV-infected Participants With Hepatitis Co-infection1.5

[back to top]

Viral Load

The decision to perform HIV-1 ribonucleic acid (RNA) tests to monitor participants' viral load was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 96 are presented. (NCT01153269)
Timeframe: Week 96

Interventionlog10 copies/mL (Mean)
HIV-infected Participants With Hepatitis Co-infection1.6

[back to top]

CD4 Cell Count

The decision to perform laboratory tests to determine participants' CD4-positive (CD4+) T-lymphocyte counts was left to the treating physician's clinical judgment. The mean and standard deviation for those tested at Week 132 are presented. (NCT01153269)
Timeframe: Week 132

InterventionCD4+ cells/μL (Mean)
HIV-infected Participants With Hepatitis Co-infection502

[back to top]

CD4 Cell Count

The decision to perform laboratory tests to determine participants' CD4-positive (CD4+) T-lymphocyte counts was left to the treating physician's clinical judgment. The mean and standard deviation for those tested at Week 144 are presented. (NCT01153269)
Timeframe: Week 144

InterventionCD4+ cells/μL (Mean)
HIV-infected Participants With Hepatitis Co-infection525

[back to top]

CD4 Cell Count

The decision to perform laboratory tests to determine participants' CD4-positive (CD4+) T-lymphocyte counts was left to the treating physician's clinical judgment. The mean and standard deviation for those tested at Week 24 are presented. (NCT01153269)
Timeframe: Week 24

InterventionCD4+ cells/μL (Mean)
HIV-infected Participants With Hepatitis Co-infection383

[back to top]

Aspartate Aminotransferase (AST) / Alanine Aminotransferase (ALT) Parameters

The decision to perform aspartate aminotransferase (AST) and alanine aminotransferase (ALT) laboratory tests to monitor participants' liver function was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Week 132 are presented. (NCT01153269)
Timeframe: Week 132

InterventionU/liter (Mean)
HIV-infected Participants With Hepatitis Co-infection: AST49
HIV-infected Participants With Hepatitis Co-infection: ALT51

[back to top]

CD4 Cell Count

The decision to perform laboratory tests to determine participants' CD4-positive (CD4+) T-lymphocyte counts was left to the treating physician's clinical judgment. The mean and standard deviation for those tested at Week 36 are presented. (NCT01153269)
Timeframe: Week 36

InterventionCD4+ cells/μL (Mean)
HIV-infected Participants With Hepatitis Co-infection408

[back to top]

CD4 Cell Count

The decision to perform laboratory tests to determine participants' CD4-positive (CD4+) T-lymphocyte counts was left to the treating physician's clinical judgment. The mean and standard deviation for those tested at Week 4 are presented. (NCT01153269)
Timeframe: Week 4

InterventionCD4+ cells/μL (Mean)
HIV-infected Participants With Hepatitis Co-infection350

[back to top]

CD4 Cell Count

The decision to perform laboratory tests to determine participants' CD4-positive (CD4+) T-lymphocyte counts was left to the treating physician's clinical judgment. The mean and standard deviation for those tested at Week 48 are presented. (NCT01153269)
Timeframe: Week 48

InterventionCD4+ cells/μL (Mean)
HIV-infected Participants With Hepatitis Co-infection402

[back to top]

CD4 Cell Count

The decision to perform laboratory tests to determine participants' CD4-positive (CD4+) T-lymphocyte counts was left to the treating physician's clinical judgment. The mean and standard deviation for those tested at Week 60 are presented. (NCT01153269)
Timeframe: Week 60

InterventionCD4+ cells/μL (Mean)
HIV-infected Participants With Hepatitis Co-infection424

[back to top]

CD4 Cell Count

The decision to perform laboratory tests to determine participants' CD4-positive (CD4+) T-lymphocyte counts was left to the treating physician's clinical judgment. The mean and standard deviation for those tested at Week 72 are presented. (NCT01153269)
Timeframe: Week 72

InterventionCD4+ cells/μL (Mean)
HIV-infected Participants With Hepatitis Co-infection431

[back to top]

CD4 Cell Count

The decision to perform laboratory tests to determine participants' CD4-positive (CD4+) T-lymphocyte counts was left to the treating physician's clinical judgment. The mean and standard deviation for those tested at Week 84 are presented. (NCT01153269)
Timeframe: Week 84

InterventionCD4+ cells/μL (Mean)
HIV-infected Participants With Hepatitis Co-infection405

[back to top]

CD4 Cell Count

The decision to perform laboratory tests to determine participants' CD4-positive (CD4+) T-lymphocyte counts was left to the treating physician's clinical judgment. The mean and standard deviation for those tested at Week 96 are presented. (NCT01153269)
Timeframe: Week 96

InterventionCD4+ cells/μL (Mean)
HIV-infected Participants With Hepatitis Co-infection503

[back to top]

Viral Load

The decision to perform HIV-1 ribonucleic acid (RNA) tests to monitor participants' viral load was left to the treating physician's clinical judgment. The mean values and standard deviations for those tested at Baseline are presented. (NCT01153269)
Timeframe: Baseline

Interventionlog10 copies/mL (Mean)
HIV-infected Participants With Hepatitis Co-infection3.9

[back to top]

Change in Proviral HIV-1 DNA in Total CD4+ T-cells From Baseline to Week 48 in Participants Randomized to the Intensified Arm Versus the Control Arm Who Received Placebo in Addition to Standard HAART.

The level of HIV Provirus in CD4 T cells obtained from peripheral blood at 48 weeks compared to baseline. A quantitative HIV PCR assay was done. The mean/median values from the standard HAART group is compared to the intensive HAART treatment regimen. (NCT01154673)
Timeframe: Baseline to Week 48

InterventionHIV DNA copies/ million CD4 cells (Median)
Intensive HAART279
Placebo Arm244

[back to top]

Pharmacokinetics Analysis: Cmax

"Cmax for pyronaridine, artesunate, DHA:~Cmax - maximum peak observed concentration~Blood samples for pyronaridine analysis were taken at Day 8 predose, Day 9 predose, Day 10 predose and 0.5, 1, 1.5, 2, 3, 4, 6, 8 and 12 hours postdose, and Days 11,12, 13, 15, 22, 29, 36 and 43.~Blood samples for artesunate / DHA analysis were taken at Day 10 predose, 0.25, 0.5, 0.75, 1, 1.5, 2, 3, 4, 6, 8 and 12 hours postdose." (NCT01156389)
Timeframe: Until Day 50 for Arm A and until Day 43 for Arm B

,
Interventionng/ml (Mean)
Pyronaridine Cmax (adjusted to a common dose of 9.72 mg/kg)Artesunate Cmax (adjusted to common dose of 3.25 mg/kg)DHA Cmax (adjusted for common 3.25 mg/kg artesunate dose)
Arm A Ritonavir Plus Pyramax480.2128.2611.4
Arm B Pyramax407.5108.7779.2

[back to top]

Summary of Treatment Emergent Adverse Events

Including all the treatment emergent adverse events, the number of subjects discontinued due to adverse events and the number of subjects with serious adverse events. (NCT01156389)
Timeframe: Throughout the study

,
InterventionParticipants (Count of Participants)
Mild treatment emergent adverse eventsModerate treatment emergent adverse eventsSevere treatment emergent adverse eventsSubjects discontinued due to adverse eventsSubjects with serious adverse events
Arm A Ritonavir Plus Pyramax126040
Arm B Pyramax153010

[back to top]

Pharmacokinetics Analysis: Half-life, Tmax

"Half-life and Tmax for pyronaridine, artesunate, DHA:~Tmax - time of the maximum observed concentration Half life - apparent plasma terminal elimination half-life, computed as ln (2)/Kel~Blood samples for pyronaridine analysis were taken at Day 8 predose, Day 9 predose, Day 10 predose and 0.5, 1, 1.5, 2, 3, 4, 6, 8 and 12 hours postdose, and Days 11,12, 13, 15, 22, 29, 36 and 43.~Blood samples for artesunate / DHA analysis were taken at Day 10 predose, 0.25, 0.5, 0.75, 1, 1.5, 2, 3, 4, 6, 8 and 12 hours postdose." (NCT01156389)
Timeframe: Until Day 50 for Arm A and until Day 43 for Arm B

,
Interventionhours (Mean)
Pyronaridine half- life (adjusted to a common dose of 9.72 mg/kg)Pyronaridine Tmax (adjusted to a common dose of 9.72 mg/kg)Artesunate half-life (adjusted to common dose of 3.25 mg/kg)Artesunate Tmax (adjusted to common dose of 3.25 mg/kg)DHA half-life (adjusted for common 3.25 mg/kg artesunate dose)DHA Tmax (adjusted for common 3.25 mg/kg artesunate dose)
Arm A Ritonavir Plus Pyramax3842.320.4251.222.271.90
Arm B Pyramax321.61.440.4650.842.351.44

[back to top]

Pharmacokinetics Analysis: AUC0-tau, AUC0-∞

"AUC0-tau, AUC0-∞ for pyronaridine, artesunate, DHA:~AUC0-tau - area under the concentration-time curve from Hour 0 to the scheduled time of the next dose AUC0-∞ - area under the concentration-time curve from Hour 0 through the last quantifiable concentration time~Blood samples for pyronaridine analysis were taken at Day 8 predose, Day 9 predose, Day 10 predose and 0.5, 1, 1.5, 2, 3, 4, 6, 8 and 12 hours postdose, and Days 11,12, 13, 15, 22, 29, 36 and 43.~Blood samples for artesunate / DHA analysis were taken at Day 10 predose, 0.25, 0.5, 0.75, 1, 1.5, 2, 3, 4, 6, 8 and 12 hours postdose." (NCT01156389)
Timeframe: Until Day 50 for Arm A and until Day 43 for Arm B

,
Interventionhours*ng/ml (Mean)
Pyronaridine AUC0-tau (adjusted to a common dose of 9.72 mg/kg)Pyronaridine AUC0-∞ (adjusted to a common dose of 9.72 mg/kg)Artesunate AUC0-tau (adjusted to common dose of 3.25 mg/kg)Artesunate AUC0-∞(adjusted to common dose of 3.25 mg/kg)DHA AUC0-tau (adjusted for common 3.25 mg/kg artesunate dose)DHA AUC0-∞ (adjusted for common 3.25 mg/kg artesunate dose)
Arm A Ritonavir Plus Pyramax2291244185.4185.413081310
Arm B Pyramax222131815014919781982

[back to top]

Adherence

Adherence, defined as proportion of doses taken (note: proportion could be greater than 1.0 for reasons such as tablets having to be taken twice due to first one being spit out or imprecise measurement of liquid doses) (NCT01172535)
Timeframe: Measured at week 4, week 12, and study completion (week 24)

InterventionProportion of expected doses taken (Median)
Week 41.00
Week 120.99
Week 241.00

[back to top]

Minimum Concentration of Lopinavir/Ritonavir (Cmin)

Minimum concentration of lopinavir/ritonavir, as determined by analysis of 12-hour pharmacokinetic sampling (NCT01172535)
Timeframe: Measured at 4 weeks of treatment prior to the observed dose and at 2, 4, 6, 8, and 12 hours post-dose

Interventionmcg/mL (Geometric Mean)
Lopinavir/Ritonavir2.47

[back to top]

Clearance of Lopinavir/Ritonavir (CL/F)

Clearance of lopinavir/ritonavir, as determined by analysis of 12-hour pharmacokinetic sampling (NCT01172535)
Timeframe: Measured at 4 weeks of treatment prior to the observed dose and at 2, 4, 6, 8, and 12 hours post-dose

InterventionL/h/kg (Geometric Mean)
Lopinavir/Ritonavir0.15

[back to top]

Lopinovir/Ritonavir Area Under the Concentration-time Curve (AUC0-24)

Area under the curve over 24 hours (AUC0-24), as determined by a non-compartmental analysis of 12-hour pharmacokinetic sampling for lopinavir/ritonavir (NCT01172535)
Timeframe: Measured at 4 weeks of treatment prior to the observed dose and at 2, 4, 6, 8, and 12 hours post-dose

Interventionmcg*hr/mL (Geometric Mean)
Lopinavir/Ritonavir196

[back to top]

Maximum Concentration of Lopinavir/Ritonavir (Cmax)

Maximum concentration of lopinavir/ritonavir, as determined by analysis of 12-hour pharmacokinetic sampling (NCT01172535)
Timeframe: Measured at 4 weeks of treatment prior to the observed dose and at 2, 4, 6, 8, and 12 hours post-dose

Interventionmcg/mL (Geometric Mean)
Lopinavir/Ritonavir11.25

[back to top]

Treatment Efficacy (HIV Viral Load)

Having HIV viral load <400 copies/mL at the week 24 visit (NCT01172535)
Timeframe: Measured at entry and study completion (week 24)

Interventionproportion of participants (Number)
Lopinavir/Ritonavir0.72

[back to top]

Treatment Efficacy (CD4%)

Having CD4%≥25 at the week 24 visit. (NCT01172535)
Timeframe: Measured at entry and study completion (week 24)

Interventionproportion of participants (Number)
Lopinavir/Ritonavir0.71

[back to top]

Proportion of Participants Tolerating LPV/r

Participants were considered to have tolerated medication if they did not stop treatment before the 24 week PK visit for any reason other than completing treatment or death not related to treatment. (NCT01172535)
Timeframe: Measured at study completion (week 24)

Interventionproportion of participants (Number)
Lopinavir/Ritonavir0.93

[back to top]

Proportion of Participants With an AUC of Less Than 10% of Adults

Proportion of participants with an AUC less that 10% of adults (AUC0-24 <104 mcg*hr/mL) (NCT01172535)
Timeframe: Measured at 4 weeks of treatment prior to the observed dose and at 2, 4, 6, 8, and 12 hours post-dose

Interventionproportion of participants (Number)
Lopinavir/Ritonavir0.15

[back to top]

Number of Participants Experiencing Adverse Events of Grade 3 or 4

Adverse events were graded by the Division of AIDS Table for Grading the Severity of Adult and Pediatric Adverse Events, Version 1.0, dated December, 2004, Clarification August 2009, which is available on the RSC web site (http://rsc.tech-res.com/safetyandpharmacovigilance/). Grade 1 = mild, Grade 2 = moderate, Grade 3 = severe, Grade 4 = potentially life-threatening, Grade 5 = death (NCT01172535)
Timeframe: Measured at study visits through end of study (weeks 2, 4, 12, 24)

Interventionparticipants (Number)
Lopinavir/Ritonavir32

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 20

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 20

Interventionlog10 Copies/mL (Median)
ETR + DRV/Rtv-2.27

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 8

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 8

Interventionlog10 Copies/mL (Median)
ETR + DRV/Rtv-2.04

[back to top]

Change From Baseline in Cluster of Differentiation 4 (CD4+) and Cluster of Differentiation 8 (CD8+) Cell Counts at Week 48

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 48

Interventioncells/uL (Mean)
CD4CD8
ETR + DRV/Rtv168-69

[back to top]

Time to Reach First Confirmed Virologic Response

CVR is defined as confirmed plasma Viral Load of less than 50 human immunodeficiency virus - type 1 (HIV-1) ribonucleic acid (RNA) copies/mL. (NCT01199939)
Timeframe: Baseline (Day 1) to Week 48

InterventionDays (Mean)
ETR + DRV/Rtv82.98

[back to top]

Number of Participants With Virologic Failure

Virologic Failure is defined as participant who is a rebounder or a non-responder. Rebounder participant is defined as a participant who is still in the study at Week 12 and first achieves 2 consecutive virologic responses (<50 copies/mL) followed by 2 consecutive non-responses or a discontinued participant (any reason) for which the last observed time point shows a non-response. Non responder participant is defined as a participant who is still in the study at Week 12 and never achieves 2 consecutive responses. (NCT01199939)
Timeframe: Baseline (Day 1) to Week 48

InterventionParticipants (Number)
ETR + DRV/Rtv7

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 12

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 12

Interventionlog10 Copies/mL (Median)
ETR + DRV/Rtv-2.18

[back to top]

Number of Participants With Confirmed Virologic Response (CVR) at Week 48

CVR is defined as confirmed plasma Viral Load of less than 50 human immunodeficiency virus - type 1 (HIV-1) ribonucleic acid (RNA) copies/mL. (NCT01199939)
Timeframe: Week 48

InterventionParticipants (Number)
ETR + DRV/Rtv40

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 16

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 16

Interventionlog10 Copies/mL (Median)
ETR + DRV/Rtv-2.29

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 48

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 48

Interventionlog10 Copies/mL (Median)
ETR + DRV/Rtv-2.29

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 42

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 42

Interventionlog10 Copies/mL (Median)
ETR + DRV/Rtv-2.29

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 4

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 4

Interventionlog10 Copies/mL (Median)
ETR + DRV/Rtv-1.77

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 36

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 36

Interventionlog10 Copies/mL (Median)
ETR + DRV/Rtv-2.24

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 30

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 30

Interventionlog10 Copies/mL (Median)
ETR + DRV/Rtv-2.28

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 24

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 24

Interventionlog10 Copies/mL (Median)
ETR + DRV/Rtv-2.17

[back to top]

Percentage of Participants With Sustained Virologic Response 24 Weeks (SVR24) Post-Treatment

Sustained Virologic Response 24 (SVR24) is defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) less than the lower limit of quantification (LLOQ; < 25 IU/mL) 24 weeks after the last dose of study drug. (NCT01221298)
Timeframe: Post-treatment Day 1 to Post-treatment Week 24

Interventionpercentage of participants (Number)
ABT-450/r and ABT-072, Plus Ribavirin (RBV)90.9

[back to top]

Percentage of Participants With Sustained Virologic Response 12 Weeks (SVR12) Post-treatment

Sustained Virologic Response 12 (SVR12) is defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) less than the lower limit of quantification (< LLOQ; < 25 IU/mL) 12 weeks after the last dose of study drug. (NCT01221298)
Timeframe: Post-treatment Day 1 to Post-treatment Week 12

Interventionpercentage of participants (Number)
ABT-450/r and ABT-072, Plus Ribavirin (RBV)90.9

[back to top]

Percentage of Participants With Hepatitis C Virus (HCV) Ribonucleic Acid (RNA) Suppressed Below the Lower Limit of Quantitation (LLOQ) From Week 4 Through Week 12

Analysis of the percentage of participants with hepatitis C virus ribonucleic acid less than the lower limit of quantitation (< 25 IU/mL). (NCT01221298)
Timeframe: Week 4 through Week 12

Interventionpercentage of participants (Number)
ABT-450/r and ABT-072, Plus Ribavirin (RBV)100

[back to top]

Percentage of Participants With Hepatitis C Virus (HCV) Ribonucleic Acid (RNA) Below the Lower Limit of Quantitation (LLOQ) at Week 4

Analysis of percentage of participants with hepatitis C virus ribonucleic acid less than the lower limit of quantitation (< 25 IU/mL). (NCT01221298)
Timeframe: Week 4

Interventionpercentage of participants (Number)
ABT-450/r and ABT-072, Plus Ribavirin (RBV)100

[back to top]

Time to Virologic Relapse Through 24 Weeks Post-treatment

Time to confirmed hepatitis C virus (HCV) ribonucleic acid (RNA) ≥ lower limit of quantitation (LLOQ) (2 consecutive measurements ≥ LLOQ) at any point in the post-treatment period among participants with HCV RNA < LLOQ at the end of treatment. (NCT01221298)
Timeframe: Post-treatment Day 1 to Post-treatment Week 24

InterventionDays (Mean)
ABT-450/r and ABT-072, Plus Ribavirin (RBV)84

[back to top]

Percentage of Participants With Hepatitis C Virus (HCV) Ribonucleic Acid (RNA) < 1000 International Units Per Milliliter (IU/mL)

Analysis of participants with HCV RNA levels below 1000 IU/mL at Week 2. (NCT01221298)
Timeframe: Week 2

Interventionpercentage of participants (Number)
ABT-450/r and ABT-072, Plus Ribavirin (RBV)100

[back to top]

Time to Failure to Suppress or Rebound During Treatment

The time to failure to suppress was defined as first day a participant met any virologic stopping criteria during treatment. The virologic stopping criteria also includes failure to achieve a 2 log10 IU/mL decrease in HCV RNA by Week 1, failure to achieve HCV RNA lower limit of detection (LLOD) for participants who previously achieved HCV RNA < LLOD. (NCT01221298)
Timeframe: Day 1 through Week 12

InterventionDays (Mean)
ABT-450/r and ABT-072, Plus Ribavirin (RBV)NA

[back to top]

Number of Participants With Death as Outcome, Serious Adverse Events (SAEs), Adverse Events (AEs) Leading to Discontinuation, AES, and AEs of Clinical Interest

An AE is any new unfavorable symptom, sign, or disease or worsening of a preexisting condition that does not necessarily have a causal relationship with treatment. An SAE is any unfavorable medical event that at any dose results in death, persistent or significant disability/incapacity, or drug dependency or abuse; is life-threatening, an important medical event, or a congenital anomaly/birth defect; or requires or prolongs hospitalization. (NCT01232127)
Timeframe: Days 1 through 25 (end of study), continuously, and at study discharge for those who discontinued prematurely.

,,
InterventionParticipants (Number)
DeathsSAEsAEs leading to discontinuationAEsAEs of clinical interest: NauseaAEs of clinical interest: Diarrhea
Atazanavir/Ritonavir (300/100) + TDF + ≥NRTI000600
Atazanavir/Ritonavir (400/100) + TDF + ≥NRTI + Famotidine (20)000723
Atazanavir/Ritonavir (400/100) + TDF + ≥NRTI + Famotidine (40)000500

[back to top]

Time of Maximum Observed Plasma Concentration (Tmax) for Atazanavir and Ritonavir

(NCT01232127)
Timeframe: Days 10, 11, 17, 18, 24, and 25 (end of study) and at study discharge for those who discontinued prematurely.

,,
InterventionHours (Median)
Atazanavir TmaxRitonavir Tmax
Atazanavir/Ritonavir (300/100) + TDF + ≥NRTI3.04.0
Atazanavir/Ritonavir (400/100) + TDF + ≥NRTI + Famotidine (20)3.04.00
Atazanavir/Ritonavir (400/100) + TDF + ≥NRTI + Famotidine (40)3.04.0

[back to top]

Maximum Observed Plasma Concentration (Cmax) and Trough Observed Plasma Concentration (Ctrough) for Atazanavir and Ritonavir

(NCT01232127)
Timeframe: Days 10, 11, 17, 18, 24, and 25 (end of study) and at study discharge for those who discontinued prematurely.

,,
Interventionng/mL (Geometric Mean)
Atazanavir CmaxAtazanavir CtroughRitonavir CmaxRitonavir Ctrough
Atazanavir/Ritonavir (300/100) + TDF + ≥NRTI3512496114145.8
Atazanavir/Ritonavir (400/100) + TDF + ≥NRTI + Famotidine (20)4131602114849.2
Atazanavir/Ritonavir (400/100) + TDF + ≥NRTI + Famotidine (40)3322494109647.3

[back to top]

Number of Participants With Abnormalities in Electrocardiogram (ECG) Findings

ECG findings include heart rate, ECG intervals (including PR, QRS, QT, and corrections to QT using both Bazett's and Fridericia's formulae), and Investigator-identified ECG abnormalities. (NCT01232127)
Timeframe: Days 1 and 25 (end of study) and at study discharge for those who discontinued prematurely.

InterventionParticipants (Number)
Nonspecific ST/T wave abnormalityShort PR interval
All Treated11

[back to top]

Area Under the Plasma Concentration-time Curve in 1 Dosing Interval (Time 0 to 24 Hours Postdose) (AUC[TAU]) for Atazanavir and Ritonavir

(NCT01232127)
Timeframe: Days 10, 11, 17, 18, 24, and 25 (end of study) and at study discharge for those who discontinued prematurely.

,,
Interventionng*h/mL (Geometric Mean)
Atazanavir AUCRitonavir AUC
Atazanavir/Ritonavir (300/100) + TDF + ≥NRTI325627317
Atazanavir/Ritonavir (400/100) + TDF + ≥NRTI + Famotidine (20)378947430
Atazanavir/Ritonavir (400/100) + TDF + ≥NRTI + Famotidine (40)314817052

[back to top]

Number of Participants With Abnormalities in Laboratory Test Results

PreRX=pretreatment; ULN=upper limit of normal. Neutrophils, (absolute), low (10*3 c/uL): <0.85*PreRx, if PreRx <1.5; <1.5 if PreRx ≥1.5. Alanine aminotransferase, high (U/L): >1.25*PreRx if PreRx >ULN; >1.25*ULN if PreRx ≤ULN. Bilirubin, direct (mg/dL), high: >1.1*ULN if PreRx ≤ULN;> 1.1*ULN if PreRx is missing; >1.25*PreRx if PreRx >ULN. Bilirubin, total (mg/dL), high: >1.1*ULN if PreRx ≤ULN;> 1.1*ULN if PreRx is missing; >1.25*PreRx if PreRx >ULN. (NCT01232127)
Timeframe: Days 11, 18, and 25 (end of study) and at study discharge for those who discontinued prematurely.

,,
InterventionParticipants (Number)
WBC differential count (low)Neutrophils (absolute) (low)Alanine aminotransferase (high)Aspartate aminotransferase (high)Bilirubin, direct (high)Bilirubin, total (high)
Atazanavir/Ritonavir (300/100) + TDF + ≥NRTI100013
Atazanavir/Ritonavir (400/100) + TDF + ≥NRTI + Famotidine (20)011007
Atazanavir/Ritonavir (400/100) + TDF + ≥NRTI + Famotidine (40)000105

[back to top]

Number of Participants With Abnormalities in Vital Signs

Vital signs include temperature, respiratory rate, seated blood pressure, and heart rate. (NCT01232127)
Timeframe: Days 1, 11, 18, and 25 (end of study) and at study discharge for those who discontinued prematurely.

InterventionParticipants (Number)
Isolated decrease in heart rateSporadic respiration rate >16 bpm
All Treated211

[back to top]

Number of Participants With Adverse Events Leading to Study Drug Discontinuation

Adverse events (AEs) were defined as any untoward medical occurrence (any unfavorable and unintended sign [including an abnormal laboratory finding], symptom, or disease) that occurred in a participant or clinical investigation participant administered a pharmaceutical product and which does not necessarily have a causal relationship with this treatment. Adult participants who received either DRV/rtv 800/100 mg once daily or DRV/rtv 600/100 mg twice daily per parent study were included in single arm and combined analysis was performed as planned in protocol because the 2 doses were extensively evaluated in their respective parent studies. Main purpose of this study was to provide continued access and not to compare the 2 doses. (NCT01281813)
Timeframe: Up to 9 years 11 months

InterventionParticipants (Count of Participants)
Continued Treatment With DRV in Combination With Rtv:Children Less Than (<) 12 Years0
Continued Treatment With DRV in Combination With Rtv: Adolescents (12-17 Years)0
Continued Treatment With DRV in Combination With Rtv:Adults (Greater Than or Equal to [>=] 18 Years)9

[back to top] [back to top]

Number of Participants With Serious Adverse Events

Adverse events (AEs): any untoward medical occurrence (any unfavorable and unintended sign [including an abnormal laboratory finding], symptom, or disease) that occurred in a participant administered a pharmaceutical product and which did not necessarily have causal relationship with study treatment. Serious adverse events (SAEs): any untoward medical occurrence at any dose resulted: death; was life threatening; requires inpatient hospitalization/prolongation of existing hospitalization; resulted in persistent/significant disability/incapacity or congenital anomaly/birth defect. Adult participants who received either DRV/rtv 800/100 mg once daily or DRV/rtv 600/100 mg twice daily per parent study were included in single arm and combined analysis was performed as planned in protocol because the 2 doses were extensively evaluated in their respective parent studies. Main purpose of this study was to provide continued access and not to compare the 2 doses. (NCT01281813)
Timeframe: Up to 9 years 11 months

InterventionParticipants (Count of Participants)
Continued Treatment With DRV in Combination With Rtv:Children Less Than (<) 12 Years0
Continued Treatment With DRV in Combination With Rtv: Adolescents (12-17 Years)1
Continued Treatment With DRV in Combination With Rtv:Adults (Greater Than or Equal to [>=] 18 Years)26

[back to top]

Percentage of Participants With Sustained Virologic Response 24 Weeks (SVR24) Post-Treatment

Sustained virologic response 24 (SVR24) is defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) less than the lower limit of quantification (LLOQ; < 25 IU/mL) 24 weeks after the last dose of study drug. (NCT01306617)
Timeframe: Post-treatment Day 1 to Post-treatment Week 24

Interventionpercentage of participants (Number)
ABT-450/r (250/100 mg) and ABT-333 Plus RBV in Treatment-naïve94.7
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Treatment-naïve85.7
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Non-responders47.1

[back to top]

Pharmacokinetics (C Trough) of ABT 450 in HCV Infected Participants

Trough concentration (C trough) is the concentration 24 hours after once daily (QD) dose and 12 hours after twice daily (BID) dose. (NCT01306617)
Timeframe: Day 1 to Week 12

Interventionnanograms (ng) per milliliter (mL) (Geometric Mean)
ABT-450/r (250/100 mg) and ABT-333 Plus RBV in Treatment-naïve53.21
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Treatment-naïve16.81
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Non-responders16.15

[back to top]

Pharmacokinetics (C Trough) of ABT-333 in HCV Infected Participants

Trough concentration (C trough) is the concentration 24 hours after once daily (QD) dose and 12 hours after twice daily (BID) dose. (NCT01306617)
Timeframe: Day 1 to Week 12

Interventionnanograms (ng) per milliliter (mL) (Geometric Mean)
ABT-450/r (250/100 mg) and ABT-333 Plus RBV in Treatment-naïve148.44
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Treatment-naïve162.57
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Non-responders166.15

[back to top]

Pharmacokinetics (C Trough) of Ribavirin in HCV Infected Participants

Trough concentration (C trough) is the concentration 24 hours after once daily (QD) dose and 12 hours after twice daily (BID) dose. (NCT01306617)
Timeframe: Day 1 to Week 12

Interventionnanograms (ng) per milliliter (mL) (Geometric Mean)
ABT-450/r (250/100 mg) and ABT-333 Plus RBV in Treatment-naïve2480
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Treatment-naïve2280
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Non-responders2000

[back to top]

Pharmacokinetics (C Trough) of Ritonavir in HCV Infected Participants

Trough concentration (C trough) is the concentration 24 hours after once daily (QD) dose and 12 hours after twice daily (BID) dose. (NCT01306617)
Timeframe: Day 1 to Week 12

Interventionnanograms (ng) per milliliter (mL) (Geometric Mean)
ABT-450/r (250/100 mg) and ABT-333 Plus RBV in Treatment-naïve43.92
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Treatment-naïve42.35
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Non-responders44.21

[back to top]

Time to Failure to Suppress or Rebound During Treatment

Time to failure to achieve a 2 log10 IU/mL HCV RNA decrease at Week 1, failure to achieve HCV RNA < Lower Limit of Detection (LLOD) at Week 6, or a confirmed increase of at least 0.5 log10 IU/mL above nadir (local minimum value) or confirmed HCV RNA > lower limit of quantitation (LLOQ) for participants who previously achieved HCV RNA < LLOQ. (NCT01306617)
Timeframe: Day 1 through Week 12

Interventiondays (Mean)
ABT-450/r (250/100 mg) and ABT-333 Plus RBV in Treatment-naïve43.0
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Treatment-naïveNA
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Non-responders62.6

[back to top]

Time to Virologic Relapse Post-treatment

Time to the first of 2 consecutive measurements of confirmed HCV RNA ≥ lower limit of quantitation (LLOQ) at any point in the post-treatment period among participants with HCV RNA < LLOQ at the end of treatment. (NCT01306617)
Timeframe: Post-treatment Day 1 to post-treatment week 48

Interventiondays (Mean)
ABT-450/r (250/100 mg) and ABT-333 Plus RBV in Treatment-naïveNA
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Treatment-naïveNA
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Non-responders15.8

[back to top]

Resistance-Associated Variants and Phenotypic Resistance

Baseline samples were analyzed for resistance-associated amino acid variants using population sequencing. Phenotypic resistance to ABT-450 or ABT-333 at baseline was assessed by calculating the fold difference in the half maximal effective concentration (EC50) compared with the EC50 for the appropriate reference replicon (1a-H77 or 1b-Con1). Participants not achieving SVR12 were analyzed for resistance-associated variants at the time of failure using population sequencing and were compared with the baseline and appropriate reference sequences to assess amino acid changes. Phenotypic resistance to ABT-450 or ABT-333 at the time of failure was assessed by calculating the fold difference in the EC50 compared with the EC50 for the corresponding baseline sample. The number of participants with variants at resistance-associated amino acid positions and phenotypic resistance at baseline and at the time of failure are presented. (NCT01306617)
Timeframe: Day 1 to post-treatment week 48

,,
Interventionparticipants (Number)
Baseline Variants in NS3; n=49Baseline Resistance to ABT-450 >10-fold; n=40Baseline Variants in NS5B; n=49Baseline Resistance to ABT-333 >10-fold; n=49Post-treatment resistant variants in NS3; n=11Post-treatment resistance to ABT-450 >10-fold; n=9Post-treatment resistant variants in NS5B; n=11Post-treatment resistance to ABT-333 >10-fold;n=11
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Non-responders11008688
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Treatment-naïve00210000
ABT-450/r (250/100 mg) and ABT-333 Plus RBV in Treatment-naïve00200000

[back to top]

Percentage of Participants With HCV RNA < 1000 International Units Per Milliliter (IU/mL)

Analysis of participants with HCV RNA levels below 1000 IU/mL at Week 2. (NCT01306617)
Timeframe: Week 2

Interventionpercentage of participants (Number)
ABT-450/r (250/100 mg) and ABT-333 Plus RBV in Treatment-naïve100
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Treatment-naïve92.9
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Non-responders100

[back to top]

Percentage of Participants With HCV RNA Below the Lower Limit of Quantitation (LLOQ; <25 IU/mL) at Week 4

Analysis of percentage of participants with hepatitis C virus ribonucleic acid less than the lower limit of quantitation (< 25 IU/mL). (NCT01306617)
Timeframe: Week 4

Interventionpercentage of participants (Number)
ABT-450/r (250/100 mg) and ABT-333 Plus RBV in Treatment-naïve100
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Treatment-naïve92.9
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Non-responders88.2

[back to top]

Percentage of Participants With Hepatitis C Virus (HCV) Ribonucleic Acid (RNA) Suppressed Below the Lower Limit of Detection (LLOD) From Week 4 Through Week 12

Analysis of the percentage of participants with hepatitis C virus ribonucleic acid less than the lower limit of detection (< 15 IU/mL). (NCT01306617)
Timeframe: Week 4 through Week 12

Interventionpercentage of participants (Number)
ABT-450/r (250/100 mg) and ABT-333 Plus RBV in Treatment-naïve89.5
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Treatment-naïve78.6
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Non-responders58.8

[back to top]

Percentage of Participants With Sustained Virologic Response 12 Weeks (SVR12) Post-treatment

Sustained virologic response 12 (SVR12) is defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) less than the lower limit of quantification (LLOQ; < 25 IU/mL) 12 weeks after the last dose of study drug. (NCT01306617)
Timeframe: Post-treatment Day 1 to Post-treatment Week 12

Interventionpercentage of participants (Number)
ABT-450/r (250/100 mg) and ABT-333 Plus RBV in Treatment-naïve94.7
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Treatment-naïve92.9
ABT-450/r (150/100 mg) and ABT-333 Plus RBV in Non-responders47.1

[back to top]

Number of Participants With Genotypable/Phenotypable Isolates, Emergent Genotypic Substitutions in Patients With Genotypable Isolates, and Phenotypic Resistance in Patients With Phenotypable Isolates at Week 24

Viral genotypic and phenotypic resistance profiles were assessed for virologic rebound (HIV-1 RNA level ≥40 c/mL). Only patients with HIV-1 RNA levels ≥500 c/mL met the criteria for resistance testing. Genotypic substitutions at baseline were summarized for virologic rebound. The genotypic resistance profile presented patients with genotypable isolates, those with protease inhibitor substitutions from genotypable isolates, those with integrase substitutions from genotypable isolates, and those with selected reverse transcriptase substitutions from genotypable isolates using the most current version of the International AIDS Society-USA list and Stanford HIV Drug Resistance Database. Newly emergent genotypic substitutions were summarized analogously for virologic rebound without baseline phenotypic resistance to atazanavir, ritonavir, or raltegravir, using all on-treatment isolates. pts=patients (NCT01332227)
Timeframe: Day 1 to Week 24

,
InterventionParticipants (Number)
Genotypable (GI)/phenotypable isolates (PI)Emergent genotypic substitutions in GI pts (n=4,0)Phenotypic resistance in PI pts (n=4,0)
Atazanavir/Ritonavir + Raltegravir441
Atazanavir/Ritonavir + Tenofovir/Emtricitabine000

[back to top]

Number of Participants With Genotypable/Phenotypable Isolates, Emergent Genotypic Substitutions in Patients With Genotypable Isolates, and Phenotypic Resistance in Patients With Phenotypable Isolates at Week 48

Viral genotypic and phenotypic resistance profiles were assessed for virologic rebound (HIV-1 RNA level ≥40 c/mL). Only patients with HIV-1 RNA levels ≥500 c/mL met the criteria for resistance testing. Genotypic substitutions at baseline were summarized for virologic rebound. The genotypic resistance profile presented patients with genotypable isolates, those with protease inhibitor substitutions from genotypable isolates, those with integrase substitutions from genotypable isolates, and those with selected reverse transcriptase substitutions from genotypable isolates using the most current version of the International AIDS Society-USA list and Stanford HIV Drug Resistance Database. Newly emergent genotypic substitutions were summarized analogously for virologic rebound without baseline phenotypic resistance to atazanavir, ritonavir, or raltegravir, using all on-treatment isolates. pts=patients (NCT01332227)
Timeframe: Day 1 to Week 48

,
InterventionParticipants (Number)
Genotypable (GI)/phenotypable isolates (PI)Emergent genotypic substitutions in GI pts (n=5,0)Phenotypic resistance in PI pts (n=5,0)
Atazanavir/Ritonavir + Raltegravir551
Atazanavir/Ritonavir + Tenofovir/Emtricitabine000

[back to top] [back to top]

Number of Participants With Virologic Rebound at Weeks 24 and 48

Viral genotypic and phenotypic resistance profiles were assessed for virologic rebound (HIV-1 RNA level ≥40 c/mL). Only patients with HIV-1 RNA levels ≥500 c/mL met the criteria for resistance testing. Genotypic substitutions at baseline were summarized for virologic rebound. The genotypic resistance profile presented patients with genotypable isolates, those with protease inhibitor substitutions from genotypable isolates, those with integrase substitutions from genotypable isolates, and those with selected reverse transcriptase substitutions from genotypable isolates using the most current version of the International AIDS Society-USA list and Stanford HIV Drug Resistance Database. Newly emergent genotypic substitutions were summarized analogously for virologic rebound without baseline phenotypic resistance to atazanavir, ritonavir, or raltegravir, using all on-treatment isolates. (NCT01332227)
Timeframe: Day 1 to Weeks 28 and 48

,
InterventionParticipants (Number)
Week 24: Virologic reboundWeek 48: Virologic rebound
Atazanavir/Ritonavir + Raltegravir79
Atazanavir/Ritonavir + Tenofovir/Emtricitabine11

[back to top]

Mean Changes in Fasting Lipid Levels From Baseline to Week 48

LD=low-density lipoprotein; HDL=high-density lipoprotein. (NCT01332227)
Timeframe: From Baseline to Week 48

,
Interventionmg/dL (Mean)
Fasting total cholesterolFasting LDL cholesterolFasting HDL cholesterolFasting non-HDL cholesterolFasting triglycerides
Atazanavir/Ritonavir + Raltegravir11.77.72.79.014.7
Atazanavir/Ritonavir + Tenofovir/Emtricitabine-10.2-5.4-0.3-9.8-17.6

[back to top]

Percentage of Participants With HIV-1 RNA Level <40 c/mL at Week 24

HIV-1 RNA level was measured with the Abbott m2000rt® polymerase chain reaction assay. Response rates were assessed using an intent-to-treat algorithm, with numerator representing patients meeting the response criteria, and denominator representing all randomized patients. Randomized patients not meeting the criteria for treatment failure (eg, discontinuation of study therapy or virologic rebound at or before Week 24) were considered responders. Virologic rebound was defined as 2 consecutive on-treatment HIV-1 RNA levels ≥40 c/mL or the last on-treatment HIV-1 RNA level ≥40 c/mL followed by discontinuation. Patients who experienced treatment failure or had missing Week 24 HIV-1 RNA levels were considered failures. RNA=ribonucleic acid; HIV=human immunodeficiency virus. (NCT01332227)
Timeframe: From Day 1 to Week 24

InterventionPercentage of participants (Number)
Atazanavir/Ritonavir + Raltegravir80.6
Atazanavir/Ritonavir + Tenofovir/Emtricitabine94.6

[back to top]

Percentage of Participants With HIV-1 RNA Level <40 c/mL at Week 48

Percentages of patients with HIV-1 RNA levels <40 c/mL were summarized at each scheduled visit. Longitudinal plots were created to display proportion versus visit week through Weeks 24 and 48 with error bars representing 95% confidence intervals. (NCT01332227)
Timeframe: From Day 1 to Week 48

InterventionPercentage of participants (Number)
Atazanavir/Ritonavir + Raltegravir69.4
Atazanavir/Ritonavir + Tenofovir/Emtricitabine86.5

[back to top]

Number of Participants Who Died and With Adverse Events (AEs) Leading to Discontinuation, Hyperbilirubinemia, Jaundice, First-degree Arterioventricular Block, Tachycardia, and Rash on ATV Powder

AE=any new unfavorable symptom, sign, or disease or worsening of a preexisting condition that may not have a causal relationship with treatment. (NCT01335698)
Timeframe: Day one to week 300 (approximately 22-Jan-2018)

,,,,
InterventionParticipants (Number)
DeathsAEs leading to discontinuationHyperbilirubinemia related adverse eventsJaundiceAtrioventricular block, first degreeTachycardiaRash
Atazanavir, 150 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)0320003
Atazanavir, 200 mg + Ritonavir, 80 mg (Weight: 10 to <15 kg)0293014
Atazanavir, 200 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)0200000
Atazanavir, 250 mg + Ritonavir, 80 mg (Weight: 15 to <25 kg)0273105
Atazanavir, 300 mg + Ritonavir, 100 mg (Weight: 25 to <35 kg)0100001

[back to top]

Minimum Plasma Concentration (Cmin)

To describe the PK profile of ATV powder formulation with RTV in pediatric subjects weighing 25 - < 35 kg and/or 6 to < 11 years of age and for the new 5 - < 10 kg cohort (200 mg ATV and 80 mg RTV) in terms of ATV Cmin (NCT01335698)
Timeframe: Baseline to Week 2

Interventionng/mL (Mean)
Atazanavir, 150 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)718.90
Atazanavir, 250 mg + Ritonavir, 80 mg (Weight: 15 to <25 kg)857.06
Atazanavir, 300 mg + Ritonavir, 100 mg (Weight: 25 to <35 kg)1030.64

[back to top]

Number of Participants Who Experienced a SAE on ATV Powder

SAE= any of the the following: is life-threatening (defined as an event in which the subject was at risk of death at the time of the event; it does not refer to an event which hypothetically might have caused death if it were more severe), requires inpatient hospitalization or causes prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect, is an important medical event (defined as a medical event(s) that may not be immediately life threatening or result in death or hospitalization but, based upon appropriate medical and scientific judgment, may jeopardize the subject or may require intervention [eg, medical, surgical] to prevent one of the other serious outcomes listed in the definition above.) Examples of such events include, but are not limited to, intensive treatment in an emergency room or at home for allergic bronchospasm; blood dyscrasias or convulsions that do not result in hospitalization (NCT01335698)
Timeframe: Day one to week 300 (approximately 22-Jan-2018)

Interventionparticipants (Number)
Atazanavir, 150 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)6
Atazanavir, 200 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)3
Atazanavir, 200 mg + Ritonavir, 80 mg (Weight: 10 to <15 kg)8
Atazanavir, 250 mg + Ritonavir, 80 mg (Weight: 15 to <25 kg)8
Atazanavir, 300 mg + Ritonavir, 100 mg (Weight: 25 to <35 kg)0

[back to top]

CD4 Cell Count Changes From Baseline on ATV Powder

CD4 cell count change from baseline using observed values (NCT01335698)
Timeframe: Baseline to Weeks 24 and 48

InterventionCells/mm^3 (Mean)
Week 24
Atazanavir, 200 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)67.8

[back to top]

CD4 Cell Count Changes From Baseline on ATV Powder

CD4 cell count change from baseline using observed values (NCT01335698)
Timeframe: Baseline to Weeks 24 and 48

,,,
InterventionCells/mm^3 (Mean)
Week 24Week 48
Atazanavir, 150 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)218.7-409.8
Atazanavir, 200 mg + Ritonavir, 80 mg (Weight: 10 to <15 kg)247.1400.0
Atazanavir, 250 mg + Ritonavir, 80 mg (Weight: 15 to <25 kg)246.1335.4
Atazanavir, 300 mg + Ritonavir, 100 mg (Weight: 25 to <35 kg)145.8213.0

[back to top]

Mean Change From Baseline in CD4 Percent on ATV Powder

Change in CD4 percent using observed values (NCT01335698)
Timeframe: Baseline to Weeks 24 and 48

InterventionPercent Change (Mean)
Week 24
Atazanavir, 200 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)4.0

[back to top]

Mean Change From Baseline in CD4 Percent on ATV Powder

Change in CD4 percent using observed values (NCT01335698)
Timeframe: Baseline to Weeks 24 and 48

,,,
InterventionPercent Change (Mean)
Week 24Week 48
Atazanavir, 150 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)5.12.8
Atazanavir, 200 mg + Ritonavir, 80 mg (Weight: 10 to <15 kg)5.48.9
Atazanavir, 250 mg + Ritonavir, 80 mg (Weight: 15 to <25 kg)6.37.5
Atazanavir, 300 mg + Ritonavir, 100 mg (Weight: 25 to <35 kg)-0.31.0

[back to top]

Mean Change From Baseline in HIV RNA on ATV Powder

Human immunodeficiency virus ribonucleic acid (HIV RNA) change from baseline using observed values (NCT01335698)
Timeframe: Baseline to Weeks 24 and 48

,,,,
InterventionLog copies per millileter (Mean)
Week 24Week 48
Atazanavir, 150 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)-2.10-2.31
Atazanavir, 200 mg + Ritonavir, 80 mg (Weight: 10 to <15 kg)-2.69-2.91
Atazanavir, 200 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)-3.07-4.06
Atazanavir, 250 mg + Ritonavir, 80 mg (Weight: 15 to <25 kg)-2.66-2.70
Atazanavir, 300 mg + Ritonavir, 100 mg (Weight: 25 to <35 kg)-2.24-3.97

[back to top]

Number of Participants With A Center of Disease Control and Prevention (CDC) Class C AIDS Event on ATV Powder

The CDC disease staging system assesses the severity of HIV disease by CD4 cell counts and by the presence of specific HIV-related conditions. CD4 counts are classified as 1: ≥500 cells/µL, 2: 200-499 cells/µL, and 3: <200 cells/µL. Children with HIV infection are also classified in each of several categories. Category N: Not symptomatic. Category A: Mildly symptomatic. Category B: Moderately symptomatic. Category C: Severely symptomatic. (NCT01335698)
Timeframe: Day one to week 300 (approximately 22-Jan-2018)

,,,,
InterventionParticipants (Number)
Pulmonary tuberculosisLymph node tuberculosisTuberculosisOropharyngeal Candidiasis
Atazanavir, 150 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)2001
Atazanavir, 200 mg + Ritonavir, 80 mg (Weight: 10 to <15 kg)1000
Atazanavir, 200 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)0000
Atazanavir, 250 mg + Ritonavir, 80 mg (Weight: 15 to <25 kg)0110
Atazanavir, 300 mg + Ritonavir, 100 mg (Weight: 25 to <35 kg)0000

[back to top]

Number of Participants With Emergent Genotypic Substitutions on ATV Powder Through Week 48

Newly emergent substitutions are on-treatment substitutions that were not detected at baseline.Viral rebound in the resistance analysis was defined as: Less than a 1 log10 drop from baseline in plasma HIV RNA level by Week 16, confirmed by a second plasma HIV RNA level redrawn within 2 and 4 weeks from original sample. Or, a plasma HIV RNA level >200 c/mL after Week 24, confirmed by a second plasma HIV RNA level redrawn within 2 and 4 weeks from original sample. Or, repeated plasma HIV RNA level ≥50 c/mL after Week 48. Viral rebound was defined as a plasma HIV RNA level ≥400 c/mL at any time in a patient who had previously achieved a plasma HIV RNA level <50 c/mL. Or, a plasma HIV RNA level ≥50 c/mL and <1,000 c/mL followed by a return to virologic suppression was considered a viral blip and not a viral rebound. NRTI=nucleoside reverse transcriptase inhibitor (NCT01335698)
Timeframe: Baseline through Week 48

,,,,
InterventionParticipants (Number)
Any PI substitutionsAny IAS-USA PI substitutionsAny select RT substitutionsNRTI
Atazanavir, 150 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)4121
Atazanavir, 200 mg + Ritonavir, 80 mg (Weight: 10 to <15 kg)4222
Atazanavir, 200 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)0011
Atazanavir, 250 mg + Ritonavir, 80 mg (Weight: 15 to <25 kg)3011
Atazanavir, 300 mg + Ritonavir, 100 mg (Weight: 25 to <35 kg)0000

[back to top]

Number of Participants With HIV RNA <50 Copies/mL and <400 Copies/mL in the Week 24 Atazanavir Powder Cohort and the Eligible Week 48 Atazanavir Powder Cohort

Virologic success includes patients with HIV RNA <50 copies/mL. Two cohorts were assessed: The Atazanavir Powder Cohort=patients who received treatment and did not switch to capsule before analysis Week 24 or before their HIV RNA Week 24 assessment, and the Eligible Week 48 Atazanavir Powder Cohort=patients who initiated study treatment at least 48 weeks before last person last visit and did not switch to capsule before analysis Week 48 or before their HIV RNA Week 48 assessment. (NCT01335698)
Timeframe: Day 1 of treatment to weeks 24 and 48

,,,,
InterventionParticipants (Number)
Week 24: HIV RNA<50 copies/mLWeek 24: HIV RNA<400 copies/mLWeek 48: HIV RNA<50 copies/mLWeek 48:HIV RNA<400 copies/mL
Atazanavir, 150 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)10151114
Atazanavir, 200 mg + Ritonavir, 80 mg (Weight: 10 to <15 kg)1015614
Atazanavir, 200 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)2501
Atazanavir, 250 mg + Ritonavir, 80 mg (Weight: 15 to <25 kg)19241822
Atazanavir, 300 mg + Ritonavir, 100 mg (Weight: 25 to <35 kg)5611

[back to top]

Number of Participants With Laboratory Test Results Meeting the Criteria for Grade 3-4 Abnormality on ATV Powder

Criteria of the Division of AIDS for grading the severity of adult and pediatric adverse events as follows: Grade (Gr) 1=mild; Gr 2=moderate; Gr 3=severe; Gr 4=potentially life-threatening. Neutrophils (absolute) (adult and infants >7 days): Gr 1=1.000-1300/mm^3; Gr 2=750-999 mm^3; Gr 3=500-749 mm^3; Gr 4= <500 mm^3. Alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase: Gr 1=1.25-2.5*upper limit of normal (ULN); Gr 2=2.6-5.0*ULN; Gr 3=5.1-10.0*ULN; Gr 4= >10.0*ULN. Bilirubin, total (adults and infants >14 days): Gr 1=1.1-1.5*ULN; Gr 2=1.6-2.5*ULN; Gr 3=2.6-5.0*ULN; Gr 4= >5.0*ULN. Lipase: Gr 1=1.1-1.5*ULN; Gr 2=1.6-3.0*ULN; Gr 3=3.1-5.0*ULN; Gr 4= >5.0*ULN. Bicarbonate, serum low: Gr 1=16.0 mEq/L-5.0*ULN. (NCT01335698)
Timeframe: Day one to week 300 (approximately 22-Jan-2018)

,,,,
InterventionParticipants (Number)
Neutrophils (absolute)Alanine aminotransferaseAspartate aminotransferaseAlkaline phosphataseTotal bilirubinAmylaseLipaseBicarbonateAlbuminCalcium, HighChloride, LowTotal Cholesterol, FastingLDL Cholesterol, FastingGlucose, Fasting, Low
Atazanavir, 150 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)042141532100000
Atazanavir, 200 mg + Ritonavir, 80 mg (Weight: 10 to <15 kg)411110600000111
Atazanavir, 200 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)01021720100000
Atazanavir, 250 mg + Ritonavir, 80 mg (Weight: 15 to <25 kg)62005620111000
Atazanavir, 300 mg + Ritonavir, 100 mg (Weight: 25 to <35 kg)00002110000000

[back to top]

Area Under the Concentration-Time Curve [AUC(TAU)]

To describe the PK profile of ATV powder formulation with RTV in pediatric subjects weighing 25 - < 35 kg and/or 6 to < 11 years of age and for the new 5 - < 10 kg cohort (200 mg ATV and 80 mg RTV) in terms of ATV AUC (NCT01335698)
Timeframe: Baseline to Week 2

Interventionng.h./mL (Mean)
Atazanavir, 150 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)49387.12
Atazanavir, 250 mg + Ritonavir, 80 mg (Weight: 15 to <25 kg)59671.80
Atazanavir, 300 mg + Ritonavir, 100 mg (Weight: 25 to <35 kg)56356.00

[back to top]

Maximum Observed Plasma Concentration (Cmax)

To describe the PK profile of ATV powder formulation with RTV in pediatric subjects weighing 25 - < 35 kg and/or 6 to < 11 years of age and for the new 5 - < 10 kg cohort (200 mg ATV and 80 mg RTV) in terms of ATV Cmax (NCT01335698)
Timeframe: Baseline to Week 2

Interventionng/mL (Mean)
Atazanavir, 150 mg + Ritonavir, 80 mg (Weight: 5 to <10 kg)5776.70
Atazanavir, 250 mg + Ritonavir, 80 mg (Weight: 15 to <25 kg)5644.12
Atazanavir, 300 mg + Ritonavir, 100 mg (Weight: 25 to <35 kg)4893.75

[back to top]

Changes in Bone Mineral Density (Using DEXA Scan and Serum Markers) From Baseline and at Week 48 - AP Lumbar Spine (L1 - L4) BMD

Bone mineral density was evaluated by DEXA scan in a subset of participants who consented to these evaluations. The effects on BMD were addressed by providing LSMs of change from baseline bone mineral density of the lumbar spine (L1-L4) as measured by the DEXA scan. (NCT01345630)
Timeframe: Week 48

Interventiong/cm^2 (Least Squares Mean)
MVC+DRV/r-0.020
FTC/TDF+DRV/r-0.025

[back to top]

Change in Bone Turnover Markers From Baseline and at Week 48 - Blood Osteocalcin

Bone turnover marker, osteocalcin, was collected in the subset of participants participating in the DEXA scan sub-study. (NCT01345630)
Timeframe: Week 48

Interventionng/mL (Mean)
MVC+DRV/r5.61
FTC/TDF+DRV/r6.77

[back to top]

Change in Bone Turnover Markers From Baseline and at Week 48 - Type 1 Collagen Peptide (CTX-1)

Bone turnover marker, C-telopeptide of type 1 collagen (CTx), was collected in the subset of participants participating in the DEXA scan sub-study. (NCT01345630)
Timeframe: Week 48

Interventionpg/mL (Mean)
MVC+DRV/r121.13
FTC/TDF+DRV/r223.52

[back to top]

Frequency of Adverse Events (AE).

Number of participants with treatment-emergent non serious AEs (NCT01345630)
Timeframe: Week 96

Interventionparticipants (Number)
MVC+DRV/r360
FTC/TDF+DRV/r365

[back to top]

Changes in Trunk to Limb Fat Distribution Using DEXA Scan From Baseline and at Week 48

A sub-study was conducted in which the participants underwent whole-body DEXA scans to evaluate peripheral fat tissue estimates for left and right arms and legs and truncal fat mass and truncal lean mass. Truncal abdominal fat were estimated from the DEXA scan field set on the torso. The effects on estimates of fat mass and lean mass were addressed by providing LSMs of change from baseline. (NCT01345630)
Timeframe: Week 48

Interventionratio (Least Squares Mean)
MVC+DRV/r0.017
FTC/TDF+DRV/r-0.014

[back to top]

Changes in Peripheral Fat Distribution Using Dual Energy X-ray Absorptiometry [DEXA] Scan From Baseline and at Week 48.

A sub-study was conducted in which the participants underwent whole-body DEXA scans to evaluate peripheral fat tissue estimates for left and right arms and legs and truncal fat mass and truncal lean mass. Truncal abdominal fat were estimated from the DEXA scan field set on the torso. The effects on estimates of fat mass and lean mass were addressed by providing LSMs of change from baseline. (NCT01345630)
Timeframe: Week 48

Interventiongram (Least Squares Mean)
MVC+DRV/r-181.6
FTC/TDF+DRV/r-257.5

[back to top] [back to top]

Changes in Bone Mineral Density (Using DEXA Scan and Serum Markers) From Baseline and at Week 48 - Femoral Neck BMD

Bone mineral density was evaluated by DEXA scan in a subset of participants who consented to these evaluations. The effects on BMD were addressed by providing LSMs of change from Baseline bone mineral density femoral neck as measured by the DEXA scan. (NCT01345630)
Timeframe: Week 48

Interventiong/cm^2 (Least Squares Mean)
MVC+DRV/r-0.021
FTC/TDF+DRV/r-0.029

[back to top]

Changes in Bone Mineral Density (Using DEXA Scan and Serum Markers) From Baseline and at Week 48 - Total Hip BMD

Bone mineral density was evaluated by DEXA scan in a subset of participants who consented to these evaluations. The effects on BMD were addressed by providing LSMs of change from baseline bone mineral density of the lumbar spine (L1-L4), left total hip and femoral neck as measured by the DEXA scan. (NCT01345630)
Timeframe: Week 48

Interventiong/cm^2 (Least Squares Mean)
MVC+DRV/r-0.014
FTC/TDF+DRV/r-0.028

[back to top]

Percent Change From Baseline in Immune Cell Function at Week 48: Lymphocyte Activation Marker CD4 (%)

The differences in the magnitude of changes in CD4+ from Baseline through Week 48 for maraviroc versus emtricitabine/tenofovir were compared. (NCT01345630)
Timeframe: Baseline, Week 48

,
InterventionPercentage of lymphocytes (Mean)
Baseline (n=396, 401)Week 48 (n=394, 396)Change from Baseline at Week 48 (n=394, 396)
FTC/TDF+DRV/r24.533.79.2
MVC+DRV/r24.231.37.0

[back to top]

Virologic Outcomes at Week 48 Using Protocol-Defined Treatment Failure (PDTF).

Per the protocol participants who meet the following criteria were regarded as PDTFs requiring a confirmatory plasma HIV-1 RNA determination: • Decrease in plasma HIV-1 RNA <1 log10 from baseline after Week 4 unless plasma HIV-1 RNA is <50 copies/mL, or • Plasma HIV-1 RNA >1.0 log10 above the nadir value after Week 4 where the nadir is the lowest plasma HIV-1 RNA concentration, or • Plasma HIV-1 RNA ≥50 copies/mL at any time after Week 24, or • Plasma HIV-1 RNA ≥50 copies/mL after suppression to <50 copies/mL on two consecutive visits, or • Decrease in plasma HIV-1 RNA ≤2 log10 from baseline on or after Week 12 unless plasma HIV-1 RNA is <400 copies/mL. Decrease in plasma HIV-1 RNA ≤2 log10 from baseline on or after Week 12 unless plasma HIV-1 RNA is <50 copies/mL (before August 30 2012) or <400 copies/mL (after August 30 2012). (NCT01345630)
Timeframe: Week 48

,
InterventionNumber of participants (Number)
Confirmed PDTFEvaluable PDTF
FTC/TDF+DRV/r133
MVC+DRV/r4017

[back to top]

Tropism Change Between Screening or Baseline and PDTF

For participants meeting the PDTF criteria, tropism was assessed using the original randomized and alternate assays (ie, both genotype testing and ESTA). Data reported here corresponds to the timepoint at or after PDTF. (NCT01345630)
Timeframe: Week 48

,,,
Interventionparticipants (Number)
R5 (Randomized Assay)NON R5 (Randomized Assay)NR (Randomized Assay)R5 (Alternate Assay)NON R5 (Alternate Assay)NR (Alternate Assay)
FTC/TDF+DRV/r - Baseline201300
FTC/TDF+DRV/r - Failure102210
MVC+DRV/r - Baseline14121025
MVC+DRV/r - Failure13131034

[back to top]

Severity of Abnormal Laboratory Values

Number of participants who had clinically significant laboratory abnormalities of Grade 3 and Grade 4 according to DAIDS. Abnormality incidence of highest grade was reported for a labcode for each individual participant. (NCT01345630)
Timeframe: Week 96

,
Interventionparticipants (Number)
Alanine Aminotransferase (ALT) (n=396, 400)Alkaline Phosphatase (n=396, 400)Amylase (n=396, 400)Aspartate Aminotransferase (AST) (n=396, 400)Blood Urea Nitrogen (BUN) (n=396, 400)Calcium (n=396, 400)Creatine Kinase (n=396, 400)Hemoglobin (n=396, 400)LDL Cholesterol (n=396, 400)Lipase (n=116, 122)Lymphocytes (Abs) (n=396, 400)Phosphate (n=396, 400)Platelets (n=396, 400)Potassium (n=396, 400)Sodium (n=396, 400)Total Bilirubin (n=396, 400)Total Neutrophils (Abs) (n=396, 400)Triglycerides (n=396, 400)Uric Acid (n=396, 400)White Blood Cell Count (n=396, 400)Creatinine (n=396, 400)
FTC/TDF+DRV/r601375102222410212120126201
MVC+DRV/r915113718450325532364010

[back to top]

Percent Change From Baseline in Immune Cell Function at Week 48: Lymphocyte Activation Marker CD8 (%)

The differences in the magnitude of changes in CD8+ cell counts from Baseline through Week 48 for maraviroc versus emtricitabine/tenofovir were compared. (NCT01345630)
Timeframe: Baseline, Week 48

,
InterventionPercentage of lymphocytes (Mean)
Baseline (n=396, 401)Week 48 (n=394, 396)Change from Baseline at Week 48 (n=394, 396)
FTC/TDF+DRV/r55.843.0-12.6
MVC+DRV/r57.046.0-10.9

[back to top]

Number of Participants With Viral Resistance to Maraviroc (Maraviroc Treated Participants Only) in Participants Meeting PDTF Criteria.

For participants meeting the PDTF criteria, viral resistance to maraviroc for maraviroc treated participants was assessed in patients with R5 virus at failure. The resistance level is calculated by reference to a laboratory strain of virus that is analyzed in parallel with the clinical isolate to identify 50% inhibitory concentrations (IC50). The maximal percent inhibition is the percent inhibition that is achieved in a titration of the drug at high concentrations when the addition of more drug does not result in increased inhibition. Maximal percent inhibition is obtained in the same way as the titration for IC50, but the key measure is of the plateau height of percent inhibition, where increased concentration of maraviroc does not result in additional inhibition. This is consistent with the virus developing some ability to use maraviroc-bound CCR5 for entry. A significant change in IC50 is not required for this mechanism. (NCT01345630)
Timeframe: Week 48

,
Interventionparticipants (Number)
Not eligible for analysis (failed tropism test)Not eligible for analysis (non-R5 tropism)Eligible for analysis (R5 virus using ESTA)Results reportedMaximal percent inhibition <95%IC50 FC ≥3.0
FTC/TDF+DRV/r111100
MVC+DRV/r41121200

[back to top]

Number of Participants With Resistance to Nucleoside/Nucleotide Reverse Transcriptase Inhibitors (NRTI), Non-nucleoside Reverse Transcriptase Inhibitors (NNRTI), and Protease Inhibitors (PI) in Participants Meeting PDTF Criteria

For participants meeting the PDTF criteria, viral resistance (both genotypic and phenotypic) to NRTI, NNRTI, and PI's were assessed at Baseline and on-treatment. The assessment was performed using the overall (i.e. net) susceptibility score provided using the PhenoSense GT assay. The number of participants with successful assessments were 15/17 for the MVC+DRV/r arm and 3/3 for the FTC/TDF+DRV/r arm. (NCT01345630)
Timeframe: Week 48

,
Interventionparticipants (Number)
NRTI - All (Baseline, n=15, 3)NNRTI Delavirdine (Baseline, n=15, 3)NNRTI Nevirapine (Baseline, n=15, 3)NNRTI Efavirenz (Baseline, n=15, 3)PRI - All (Baseline, n=15, 3)NRTI - All (PDTF, n=15, 3)NNRTI Delavirdine (PDTF, n=15, 3)NNRTI Nevirapine (PDTF, n=15, 3)NNRTI Efavirenz (PDTF, n=15, 3)PRI - All (PDTF, n=15, 3)
FTC/TDF+DRV/r0000000000
MVC+DRV/r0111001110

[back to top]

Number of Participants With Abnormal Laboratory Values

Number of participants with laboratory abnormalities are reported (NCT01345630)
Timeframe: Week 96

,
Interventionparticipants (Number)
Normal BaselineAbnormal Baseline
FTC/TDF+DRV/r205101
MVC+DRV/r210111

[back to top]

Absolute Change in CD4+/CD8+ Ratio From Baseline to Week 48

The differences in the magnitude of changes in CD4+/CD8+ ratio from Baseline through Weeks 48 for maraviroc versus emtricitabine/tenofovir were compared. (NCT01345630)
Timeframe: Baseline, Week 48

,
InterventionRatio (Mean)
Baseline (n=396, 401)Week 48 (n=394, 396)Change from Baseline at Week 48 (n=394, 396)
FTC/TDF+DRV/r0.480.870.39
MVC+DRV/r0.470.750.28

[back to top]

Absolute Change From Baseline in Immune Cell Function at Week 48: Lymphocyte Marker Cluster of Differentiation 8 (CD8, Cell/mm^3)

The differences in the magnitude of changes in CD8+ cell counts from baseline through Week 48 for maraviroc versus emtricitabine/tenofovir were compared. (NCT01345630)
Timeframe: Baseline, Week 48

,
Interventioncell/mm^3 (Mean)
Baseline (n=396, 401)Week 48 (n=394, 396)Change from Baseline at Week 48 (n=394, 396)
FTC/TDF+DRV/r914.5751.1-157.9
MVC+DRV/r954.4900.0-49.9

[back to top]

Absolute Change From Baseline in Immune Cell Function at Week 48: Lymphocyte Marker Cluster of Differentiation 4 (CD4, Cell/mm^3)

The differences in the magnitude of changes in CD4+ at Baseline and at Week 48 for maraviroc versus emtricitabine/tenofovir were compared. (NCT01345630)
Timeframe: Baseline, Week 48

,
Interventioncell/mm^3 (Mean)
Baseline (n=396, 401)Week 48 (n=394, 396)Change from Baseline at Week 48 (n=394, 396)
FTC/TDF+DRV/r379.5574.6194.2
MVC+DRV/r382.0576.9194.9

[back to top]

The Relationship Between the Proportion of Participants With Plasma HIV-1 RNA <50 Copies/mL at the Week 48 and the Screening Tropism Test (Genotype Test or ESTA).

The relationship of the proportion of participants achieving HIV-1 RNA <50 copies/mL at Week 48 with the screening tropism test for the MVC containing regimen was analyzed. Virologic response for a participant at Week 48 was derived using the FDA's Snapshot MSDF algorithm. Difference in proportions of patients with plasma HIV-1 RNA <50 copies/mL at week 48 between the maraviroc and the emtricitabine/tenofovir treatment arms, with two-sided 95% confidence interval, among patients who are R5 by genotype (including some who were originally randomized to ESTA and are R5 by genotype upon retesting), were calculated via the Maximum Likelihood method. The estimate was adjusted for the screening plasma HIV RNA level (<100,000 vs. ≥100,000) copies/mL via the Mantel Haenszel (MH) method. (NCT01345630)
Timeframe: Week 48

Interventionproportion of participants (Number)
MVC+DRV/r0.8047
FTC/TDF+DRV/r0.8797

[back to top]

Percentage of Participants With Plasma HIV-1 RNA <50 Copies/mL.

"The proportion of participants who achieved HIV-1 RNA <50 copies/mL at week 48 was assessed according to Food and Drug Administration's (FDA's) Missing, Switch, Discontinuation'=Failure (MSDF) Snapshot algorithm. The algorithm used the plasma HIV-1 RNA in the Week 48 visit window, followed the virology-first principle and considers a participant who has a missing plasma HIV-1 RNA, or switches to prohibited ARV regimen or discontinues from the study or study drug for any reason, or dies, as a failure." (NCT01345630)
Timeframe: Week 48

InterventionPercentage of participants (Number)
MVC+DRV/r77.3
FTC/TDF+DRV/r86.8

[back to top]

Number of Participants With Treatment-emergent Serious Adverse Events

Total number of participants with treatment-emergent serious adverse events are reported (NCT01345630)
Timeframe: Week 96

Interventionparticipants (Number)
MVC+DRV/r41
FTC/TDF+DRV/r40

[back to top]

Number of Participants With Grade 3 or 4 AEs

Number of participants with grade 3 or 4 AEs are presented here. (NCT01345630)
Timeframe: Week 96

Interventionparticipants (Number)
MVC+DRV/r65
FTC/TDF+DRV/r71

[back to top]

Number of Participants Who Discontinued Due to AEs

Number of participants who discontinued due to AEs are reported here. Three participants (two from the MVC+DRV/r arm and one from the FTC/TDF+DRV/r arm) were not considered as discontinued due to AE because other reasons for discontinuation were prioritized for these participants. (NCT01345630)
Timeframe: Week 96

Interventionparticipants (Number)
MVC+DRV/r22
FTC/TDF+DRV/r23

[back to top]

Proportions of Subjects Experiencing Virologic Failure by Randomized Treatment Arm

For the purposes of the study, virologic failure is defined as either regimen change for any reason, or plasma viral load >400 copies/mL on 2 consecutive measurements >2 weeks apart. (NCT01351740)
Timeframe: at or before 48 weeks.

InterventionParticipants (Count of Participants)
Switch2
Continuation6

[back to top]

Proportions of Subjects in Each Randomized Treatment Arm With Atazanavir Trough Levels Below 150ng/mL

Therapeutic drug monitoring (TDM) to determine atazanavir trough plasma level will be performed once on all subjects at 4-8 weeks (NCT01351740)
Timeframe: 1 month (4-8 weeks)

InterventionParticipants (Count of Participants)
Switch14
Continuation3

[back to top]

Number of Participants With HIV-1 Drug Resistance Mutations in Protease, Reverse Transcriptase, and Integrase in Participants With Virologic Failure at Baseline and at Time of Virologic Failure

Mutations were defined as major IAS mutations in the IAS-USA July 2014 list. New mutations were those detected at virologic failure but not at baseline. (NCT01352715)
Timeframe: From study entry through to week 96

,
Interventionparticipants (Number)
No new IAS mutations1-2 new IAS mutations3 new IAS mutations
Arm A: LPV/r Plus RAL2991
Arm B: LPV/r Plus Best Available NRTIs32130

[back to top]

Changes in Fasting Total Cholesterol, High-density Lipoprotein (HDL) Cholesterol, Low-density Lipoprotein (LDL) Cholesterol, Triglycerides, and Glucose From Baseline

Fasting was for 8 hours and the metabolic panel was drawn locally. (NCT01352715)
Timeframe: Study entry and week 48

,
Interventionmg/dL (Mean)
total cholesterol changehigh-density lipoprotein (HDL) cholesterol changelow-density lipoprotein (LDL) cholesterol changetriglycerides changeglucose change
Arm A: LPV/r Plus RAL31417802
Arm B: LPV/r Plus Best Available NRTIs15210313

[back to top]

Percentage of Time Spent in Hospital

The percentage of total study time that participants were in hospital. (NCT01352715)
Timeframe: From study entry throughout follow-up (up to 96 weeks)

Interventionpercentage of time spent in hospital (Number)
Arm A: LPV/r Plus RAL0.08
Arm B: LPV/r Plus Best Available NRTIs0.12

[back to top]

Number of Participants With a Targeted Serious Non-AIDS-defining Event or Death

Serious non-AIDS diagnoses were based on ACTG Appendix 60 Diagnosis Codes (NCT01352715)
Timeframe: From study entry throughout follow-up (up to 96 weeks)

Interventionparticipants (Number)
Arm A: LPV/r Plus RAL7
Arm B: LPV/r Plus Best Available NRTIs7

[back to top]

Change in CD4+ Cell Count From Baseline to Week 48

Change in CD4+ cell count was calculated as CD4+ cell count at week 48 minus CD4+ cell count at study entry. (NCT01352715)
Timeframe: Study entry and week 48

Interventioncells/mm^3 (Mean)
Arm A: LPV/r Plus RAL199
Arm B: LPV/r Plus Best Available NRTIs190

[back to top]

Cumulative Probability of Virologic Failure by Week 48

The primary endpoint was time to virologic failure. Virologic failure was defined as confirmed viral load >400 copies/mL at or after week 24. The Kaplan-Meier estimate of the cumulative probability of virologic failure by week 48 was used. (NCT01352715)
Timeframe: From study entry to week 48

Interventioncumulative probability per 100 persons (Number)
Arm A: LPV/r Plus RAL10.3
Arm B: LPV/r Plus Best Available NRTIs12.4

[back to top]

Number of Participants Discontinuing Randomized Treatment for Toxicity

Discontinuation of randomized treatment for toxicity included participant decision to discontinue for low grade toxicity. Within class NRTI changes were not considered discontinuations. (NCT01352715)
Timeframe: From Start of Randomized Treatment to Off Randomized Treatment (up to 96 weeks)

Interventionparticipants (Number)
Arm A: LPV/r Plus RAL3
Arm B: LPV/r Plus Best Available NRTIs3

[back to top]

Number of Participants With a New AIDS-defining Events or Death

AIDS-defining events were those recognized by the Centers for Disease Control (CDC) and World Health Organization (WHO) (NCT01352715)
Timeframe: From study entry throughout follow-up (up to 96 weeks)

Interventionparticipants (Number)
Arm A: LPV/r Plus RAL15
Arm B: LPV/r Plus Best Available NRTIs17

[back to top]

Number of Participants With Grade 3 or Higher Adverse Event (AE) at Least One Grade Higher Than Baseline

The DAIDS Adverse Event (AE) Grading Table, Version 1.0, December 2004 (Clarification, August 2009) was used for grading of AEs. (NCT01352715)
Timeframe: From start of randomized treatment to off randomized treatment (up to 96 weeks)

Interventionparticipants (Number)
Arm A: LPV/r Plus RAL62
Arm B: LPV/r Plus Best Available NRTIs81

[back to top]

Cτ,ss of Darunavir

concentration of the analyte in plasma at steady-state after a uniform dosing interval τ=24h of darunavir (NCT01374802)
Timeframe: 0:00, 0:30, 1:00, 1:30, 2:00, 3:00, 4:00, 6:00, 8:00, 10:00,12:00 h after drug administration on day 8 (DRV/r) and day 16 (BI 201335+DRV/r)

Interventionng/mL (Geometric Mean)
Darunavir+Ritonavir1330
Faldaprevir+Darunavir+Ritonavir1170

[back to top]

Cmax,ss of Darunavir

maximum measured concentration of the analyte in plasma at steady-state (NCT01374802)
Timeframe: 0:00, 0:30, 1:00, 1:30, 2:00, 3:00, 4:00, 6:00, 8:00, 10:00,12:00 h after drug administration on day 8 (DRV/r) and day 16 (BI 201335+DRV/r)

Interventionng/mL (Geometric Mean)
Darunavir+Ritonavir4930
Faldaprevir+Darunavir+Ritonavir6330

[back to top]

Tmax,ss of Darunavir

time from last dosing to maximum concentration of the analyte in plasma at steady state (NCT01374802)
Timeframe: 0:00, 0:30, 1:00, 1:30, 2:00, 3:00, 4:00, 6:00, 8:00, 10:00,12:00 hours (h) after drug administration on day 8 (DRV/r) and day 16 (BI 201335+DRV/r)

Interventionh (Median)
Darunavir+Ritonavir1.50
Faldaprevir+Darunavir+Ritonavir2.01

[back to top]

AUCτ,ss of Darunavir

"area under the concentration-time curve of the analyte in plasma at steadystate over a uniform dosing interval τ of darunavir.~The measured values show inter-individual variabilities, whereas the statistical analyses show intra-individual variabilities" (NCT01374802)
Timeframe: 0:00, 0:30, 1:00, 1:30, 2:00, 3:00, 4:00, 6:00, 8:00, 10:00,12:00 hours (h) after drug administration on day 8 (DRV/r) and day 16 (BI 201335+DRV/r)

Interventionng*h/mL (Geometric Mean)
Darunavir+Ritonavir57200
Faldaprevir+Darunavir+Ritonavir66000

[back to top]

Change From Monotherapy Baseline in Cluster of Differentiation (CD)4+ and CD8+ T-cell Counts During Monotherapy

Blood was collected and CD4+ and CD8+ cell count assessment was done by flow cytometery and was carried out at Baseline (Day 1) to evaluate the immunological activity of multiple doses of FTR. Baseline is defined as the last non-missing value on or before the date of first dose of study treatment and the values are absolute values. Change from Baseline was calculated as value at indicated time point minus Baseline value. Only those participants with data available at the specified time points were analyzed. (NCT01384734)
Timeframe: Baseline and Day 8

,,,
Interventioncells per cubic millimeter (Mean)
CD4+CD8+
FTR 1200 mg QD/RAL/TDF63.467.6
FTR 400 mg BID/RAL/TDF58.4134.2
FTR 600 mg QD/RAL/TDF71.8188.0
FTR 800 mg BID/RAL/TDF134.8216.3

[back to top]

Change From Monotherapy Baseline in log10 HIV RNA of the Monotherapy Period

Change from monotherapy Baseline in log10 HIV RNA to assess the antiviral activity of temsavir following administration of selected doses of FTR administered orally to HIV-1-infected participants for 7 days. Baseline is defined as the last non-missing value on or before the date of first dose of study treatment. Change from Baseline was calculated as value at indicated time point minus Baseline value. ITT-E Monotherapy Population comprised of participants that were randomized and participated in the monotherapy sub-study and received at least one dose of FTR Monotherapy. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles). (NCT01384734)
Timeframe: Baseline and up to Day 8 of the monotherapy period

,,,
Interventionlog10 c/mL (Mean)
Day 2, n=7, 5, 10, 9Day 5, n=7, 4, 10, 10Day 6, n=7, 5, 10, 10Day 7, n=6, 5, 10, 10Day 8, n=6, 4, 9, 9
FTR 1200 mg QD/RAL/TDF0.126-0.767-1.053-1.198-1.470
FTR 400 mg BID/RAL/TDF0.220-0.340-0.530-0.556-0.691
FTR 600 mg QD/RAL/TDF0.126-0.593-0.822-1.086-1.218
FTR 800 mg BID/RAL/TDF0.149-0.811-1.082-1.443-1.372

[back to top]

Number of Participants With Newly-emergent Genotypic Substitutions at Week 24

Participants who administered antiretroviral (ARV) with virologic failure (VF) were assessed. Genotypic substitution included assessment of Reverse Transcriptase (RT) substitution, Protease Inhibitor (PI) substitution and Integrase RAL substitution as per International Acquired Immune Deficiency Syndrome (AIDS) Society-USA (IAS-USA) list. ITT-E Resistance Tested through Week 24 population included participants who met the criteria for Resistance testing, and the confirmatory value or value at discontinuation occurred at or before the end of the Week 24 Snapshot analysis window. The criteria for resistance tested was participant who had virologic failure or met the following criteria a) Participants who achieved viral suppression (plasma HIV-1 RNA < 50 c/mL) and have confirmed plasma HIV-1 RNA >= 400 c/mL at any time during the study. b) Participants who were discontinued before achieving viral suppression (plasma HIV-1 RNA < 50 c/mL) after Week 8 with last plasma HIV-1 RNA >=400 c/mL. (NCT01384734)
Timeframe: Up to Week 24

,,,,
InterventionParticipants (Count of Participants)
PI substitutionRT substitutionIntegrase substitution
ATV/r/RAL/TDF000
FTR 1200 mg QD/RAL/TDF011
FTR 400 mg BID/RAL/TDF000
FTR 600 mg QD/RAL/TDF001
FTR 800 mg BID/RAL/TDF000

[back to top]

Number of Participants With Newly-emergent Genotypic Substitutions at Week 48

Participants who administered ARV with VF were assessed. Genotypic substitution included assessment of RT substitution, PI substitution and Integrase RAL substitution as per IAS-USA list. ITT-E Resistance Tested through Week 48 population included participants who met the criteria for Resistance testing, and the confirmatory value or value at discontinuation occurred at or before the end of the Week 48 Snapshot analysis window. The criteria for resistance tested was participant who had virologic failure or the following criteria a) Participants who achieved viral suppression (plasma HIV-1 RNA < 50 c/mL) and have confirmed plasma HIV-1 RNA >= 400 c/mL at any time during the study. b) Participants who were discontinued before achieving viral suppression (plasma HIV-1 RNA < 50 c/mL) after Week 8 with last plasma HIV-1 RNA >=400 c/mL. (NCT01384734)
Timeframe: Up to Week 48

,,,,
InterventionParticipants (Count of Participants)
PI substitutionRT substitutionIntegrase substitution
ATV/r/RAL/TDF000
FTR 1200 mg QD/RAL/TDF122
FTR 400 mg BID/RAL/TDF101
FTR 600 mg QD/RAL/TDF001
FTR 800 mg BID/RAL/TDF001

[back to top]

Number of Participants With Newly-emergent Genotypic Substitutions at Week 96

Participants who administered ARV with VF were assessed. Genotypic substitution included assessment of RT substitution, PI substitution and Integrase RAL substitution as per IAS-USA list. ITT-E Resistance Tested through Week 96 population included participants who met the criteria for Resistance testing, and the confirmatory value or value at discontinuation occurred at or before the end of the Week 96 Snapshot analysis window. The criteria for resistance tested was participants with virologic failure or the following criteria a) Participants who achieved viral suppression (plasma HIV-1 RNA < 50 c/mL) and have confirmed plasma HIV-1 RNA >= 400 c/mL at any time during the study. b) Participants who were discontinued before achieving viral suppression (plasma HIV-1 RNA < 50 c/mL) after Week 8 with last plasma HIV-1 RNA >=400 c/mL. (NCT01384734)
Timeframe: Up to Week 96

,,,,
InterventionParticipants (Count of Participants)
PI substitutionRT substitutionIntegrase RAL substitution
ATV/r/RAL/TDF010
FTR 1200 mg QD/RAL/TDF223
FTR 400 mg BID/RAL/TDF322
FTR 600 mg QD/RAL/TDF221
FTR 800 mg BID/RAL/TDF112

[back to top]

Number of Participants With SAE and Discontinuation Due to AEs During Monotherapy Period

Any untoward event resulting in death, life threatening, requires hospitalization or prolongation of existing hospitalization, results in disability/incapacity, congenital anomaly/birth defect, any other situation according to medical or scientific judgment or suspected transmission of an infectious agent via the study drug were categorized as SAE. AEs leading to discontinuation of study therapy were also reported as safety assessment. (NCT01384734)
Timeframe: Up to Day 8 of the monotherapy period

,,,
InterventionParticipants (Count of Participants)
SAEAEs leading to discontinuation
FTR 1200 mg QD/RAL/TDF00
FTR 400 mg BID/RAL/TDF00
FTR 600 mg QD/RAL/TDF00
FTR 800 mg BID/RAL/TDF00

[back to top]

Number of Participants With SAE and Discontinuation Due to AEs During Primary Study

Any untoward event resulting in death, life threatening, requires hospitalization or prolongation of existing hospitalization, results in disability/incapacity, congenital anomaly/birth defect, any other situation according to medical or scientific judgment or suspected transmission of an infectious agent via the study drug were categorized as SAE. AEs leading to discontinuation of study therapy were also reported as safety assessment. Safety Population comprised of participants who received at least one dose of study treatment. Summaries of SAEs and AEs leading to discontinuation or withdrawal through Week X (where X = 48 or 96) included AEs with onset on or after the start of study treatment (i.e. study date of first study treatment intake) up to and including the end of the Week 48 and 96 visit snapshot window. (NCT01384734)
Timeframe: Weeks 48 and 96

,,,,
InterventionParticipants (Count of Participants)
SAE, Week 48SAE, Week 96AEs leading to discontinuation, Week 48AEs leading to discontinuation, Week 96
ATV/r/RAL/TDF5735
FTR 1200 mg QD/RAL/TDF2412
FTR 400 mg BID/RAL/TDF3511
FTR 600 mg QD/RAL/TDF4600
FTR 800 mg BID/RAL/TDF5722

[back to top]

Number of Participants With Serious Adverse Events (SAE) and Discontinuation Due to AEs up to Week 24

Any untoward event resulting in death, life threatening, requires hospitalization or prolongation of existing hospitalization, results in disability/incapacity, congenital anomaly/birth defect, any other situation according to medical or scientific judgment or suspected transmission of an infectious agent via the study drug were categorized as SAE. AEs leading to discontinuation of study therapy were also reported as safety assessment. Safety population included all participants who received at least one dose of study treatment. Summaries of SAEs and AEs leading to discontinuation or withdrawal through Week 24 included AEs with onset on or after the start of study treatment (i.e. study date of first study treatment intake) up to and including the end of the Week 24 visit snapshot window. (NCT01384734)
Timeframe: Up to Week 24

,,,,
InterventionParticipants (Count of Participants)
SAEAEs leading to discontinuation
ATV/r/RAL/TDF52
FTR 1200 mg QD/RAL/TDF21
FTR 400 mg BID/RAL/TDF31
FTR 600 mg QD/RAL/TDF40
FTR 800 mg BID/RAL/TDF42

[back to top]

Percentage of Participants With Plasma HIV-1 RNA < 50 c/mL at Primary Study

Percentage of participants with plasma HIV 1 RNA < 50 c/mL at Weeks 48 and 96 using the FDA snapshot algorithm was assessed to evaluate the antiviral activity. Treatment comparisons were not performed as this was an estimation study. Response rates were tabulated by treatment arm with exact Clopper-Pearson binomial 95 percentage CI. Virologic success or failure was determined by the last available HIV-1 RNA assessment while the participant was on-treatment within the snapshot window of the visit of interest. (NCT01384734)
Timeframe: Weeks 48 and 96

,,,,
InterventionPercentage of Participants (Number)
Week 48Week 96
ATV/r/RAL/TDF7157
FTR 1200 mg QD/RAL/TDF6858
FTR 400 mg BID/RAL/TDF8278
FTR 600 mg QD/RAL/TDF6963
FTR 800 mg BID/RAL/TDF6149

[back to top]

Percentage of Participants With Plasma HIV-1 RNA < 50 c/mL at Day 8 of the Monotherapy Period

Percentage of participants with plasma HIV 1 RNA < 50 c/mL at Baseline of combination therapy was assessed to evaluate the antiviral activity of four doses of FTR. Baseline of combination therapy was the Day 1 of the combination therapy. Virologic success or failure was determined using the non-missing viral load value at Baseline of combination therapy. The assessment closest to the window target Study Day was used for the analysis. Only those participants with data available at the specified time points were analyzed. (NCT01384734)
Timeframe: Up to Day 8 of the monotherapy period

InterventionPercentage of Participants (Number)
FTR 400 mg BID/RAL/TDF0
FTR 800 mg BID/RAL/TDF0
FTR 600 mg QD/RAL/TDF0
FTR 1200 mg QD/RAL/TDF11

[back to top]

Change From Baseline in IC50 Fold Change Among Participants With VF at Week 48

Virologic failure is defined clinically as confirmed plasma HIV-1 RNA >= 50 copies/mL at Week 24 or later or later or virologic rebound defined as confirmed HIV-1 RNA >=50 copies/mL at any time after prior confirmed suppression to <50 copies/mL OR confirmed >1 log10 copies/mL increase in HIV-1 RNA at any time above nadir level where nadir was >= 50 copies/mL. The phenotypic resistance to a drug is defined as a fold change (i.e, ratio of the IC50 of the clinical isolate to the IC50 of the reference strain) greater than the cut-off for reduced susceptibility. Maximum change from Baseline in Temsavir IC50 fold change based on all on-treatment values has been presented. Baseline is defined as the last non-missing value on or before the date of first dose of study treatment and those values are absolute values. Change from Baseline was calculated as value at indicated time point minus Baseline value. Only those participants available at the specified time points were analyzed. (NCT01384734)
Timeframe: Baseline and up to Week 48

InterventionIC50 Fold Change (Mean)
FTR 400 mg BID/RAL/TDF-2.624
FTR 800 mg BID/RAL/TDF586.776
FTR 600 mg QD/RAL/TDF81.729
FTR 1200 mg QD/RAL/TDF449.092

[back to top]

Change From Baseline in IC50 Fold Change Among Participants With VF at Week 96

Virologic failure is defined clinically as confirmed plasma HIV-1 RNA >= 50 copies/mL at Week 24 or later or virologic rebound defined as confirmed HIV-1 RNA >=50 copies/mL at any time after prior confirmed suppression to <50 copies/mL OR confirmed >1 log10 copies/mL increase in HIV-1 RNA at any time above nadir level where nadir was >= 50 copies/mL. The phenotypic resistance to a drug is defined as a fold change (i.e, ratio of the IC50 of the clinical isolate to the IC50 of the reference strain) greater than the cut-off for reduced susceptibility. Maximum change from Baseline in Temsavir IC50 fold change based on all on-treatment values has been presented. Baseline is defined as the last non-missing value on or before the date of first dose of study treatment and those values are absolute values. Change from Baseline was calculated as value at indicated time point minus Baseline value. Only those participants available at the specified time points were analyzed. (NCT01384734)
Timeframe: Baseline and up to Week 96

InterventionIC50 Fold Change (Mean)
FTR 400 mg BID/RAL/TDF25.480
FTR 800 mg BID/RAL/TDF419.901
FTR 600 mg QD/RAL/TDF46.351
FTR 1200 mg QD/RAL/TDF777.818

[back to top]

Maximum Change From Baseline in Inhibitory Concentration at 50% (IC50) Fold Change Among Participants With VF at Week 24

Virologic failure is defined clinically as confirmed plasma HIV-1 RNA >= 50 copies/mL at Week 24 or later or virologic rebound defined as confirmed HIV-1 RNA >=50 copies/mL at any time after prior confirmed suppression to <50 copies/mL OR confirmed >1 log10 copies/mL increase in HIV-1 RNA at any time above nadir level where nadir was >= 50 copies/mL . The phenotypic resistance to a drug is defined as a fold change (i.e, ratio of the IC50 of the clinical isolate to the IC50 of the reference strain) greater than the cut-off for reduced susceptibility. Maximum change from Baseline in Temsavir IC50 fold change based on all on-treatment values has been presented. Baseline is defined as the last non-missing value on or before the date of first dose of study treatment and those values are absolute values. Change from Baseline was calculated as value at indicated time point minus Baseline value. Only those participants available at the specified time points were analyzed. (NCT01384734)
Timeframe: Baseline and up to Week 24

InterventionIC50 Fold Change (Mean)
FTR 400 mg BID/RAL/TDF-2.350
FTR 800 mg BID/RAL/TDF1014.748
FTR 600 mg QD/RAL/TDF101.627
FTR 1200 mg QD/RAL/TDF39.030

[back to top]

Maximum Decrease From Monotherapy Baseline in log10 Plasma HIV-1 RNA

Maximum decrease from monotherapy Baseline in log10 plasma HIV-1 RNA during monotherapy to assess the antiviral activity of temsavir following administration of selected doses of FTR administered orally to HIV-1-infected participants for 7 days. Baseline is defined as the last non-missing value on or before the date of first dose of study treatment. Change from Baseline was calculated as value at indicated time point minus Baseline value. The data for monotherapy nadir has been presented where nadir represents the maximum decrease from Baseline. (NCT01384734)
Timeframe: Baseline and up to Day 8 of the monotherapy period

Interventionlog10 c/mL (Mean)
FTR 400 mg BID/RAL/TDF-0.770
FTR 800 mg BID/RAL/TDF-1.524
FTR 600 mg QD/RAL/TDF-1.250
FTR 1200 mg QD/RAL/TDF-1.399

[back to top]

Percentage of Participants With Plasma HIV-1 Ribonucleic Acid (RNA) < 50 Copies Per Milliliter (c/mL) at Week 24

Percentage of participants with plasma HIV 1 RNA < 50 c/mL at Week 24 using the Food and Drug Administration (FDA) snapshot algorithm was assessed to evaluate the antiviral activity. Treatment comparisons were not performed as this was an estimation study. Response rates were tabulated by treatment arm with exact Clopper-Pearson binomial 95 percentage confidence intervals (CI). Virologic success or failure was determined by the last available HIV-1 RNA assessment while the participant was on-treatment within the snapshot window of the visit of interest. Intent-To-Treat-Exposed (ITT-E) Population includes all randomized participants who received at least one dose of study treatment. (NCT01384734)
Timeframe: Week 24

InterventionPercentage of Participants (Number)
FTR 400 mg BID/RAL/TDF80
FTR 800 mg BID/RAL/TDF69
FTR 600 mg QD/RAL/TDF76
FTR 1200 mg QD/RAL/TDF72
ATV/r/RAL/TDF75

[back to top]

Change From Baseline in CD4+ T-cell Count

Blood was collected and CD4+ cell count assessment by flow cytometery was carried out at Baseline (Day 1), Weeks 24, 48 and 96 to evaluate the immunological activity of multiple doses of BMS-663068/GSK3684934. Baseline is defined as the last non-missing value on or before the date of first dose of study treatment and those values are absolute values. Change from Baseline was calculated as value at indicated time point minus Baseline value. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles). (NCT01384734)
Timeframe: Baseline and Weeks 24, 48 and 96

,,,,
InterventionCells per cubic millimeter (Mean)
Week 24, n=41, 38, 48, 42, 40Week 48, n=43, 34, 43, 41, 41Week 96, n=42, 28, 35, 28, 31
ATV/r/RAL/TDF119.4178.7250.1
FTR 1200 mg QD/RAL/TDF124.5155.4211.7
FTR 400 mg BID/RAL/TDF134.3199.1264.6
FTR 600 mg QD/RAL/TDF109.5140.5175.7
FTR 800 mg BID/RAL/TDF111.0158.7210.8

[back to top]

Change From Monotherapy Baseline in CD4+ and CD8+ T-cell Proportion During Monotherapy

Blood was collected and CD4+ and CD8+ proportion assessment was done by flow cytometery and was carried out at Baseline (Day 1) to evaluate the immunological activity of multiple doses of FTR. Baseline is defined as the last non-missing value on or before the date of first dose of study treatment and the values are absolute values. Change from Baseline was calculated as value at indicated time point minus Baseline value. Only those participants with data available at the specified time points were analyzed. (NCT01384734)
Timeframe: Baseline and Day 8

,,,
Interventioncells per cubic millimeter (Mean)
CD4+CD8+
FTR 1200 mg QD/RAL/TDF0.014-0.021
FTR 400 mg BID/RAL/TDF-0.005-0.003
FTR 600 mg QD/RAL/TDF0.008-0.009
FTR 800 mg BID/RAL/TDF0.023-0.040

[back to top]

Change From Baseline in Mean Framingham Risk Score at Week 24 and Week 48: Medican Change in Framingham Risk Score

The Framingham Risk Score is used to estimate the 10-year cardiovascular risk of a participant. It is calculated according to age, laboratory values of total cholesterol and HDL cholesterol, smoking status, and systolic blood pressure. The framingham risk score is calculated as: for males: 0 point (1 percentage) up to 17 points (30 percentages); whereas for females: 0 to 9 points (1 percentage) up to 25 points (30 percentage). Higher scores indicate high cardiovascular risk. (NCT01391013)
Timeframe: Baseline, Week 24, and Week 48

,
InterventionFramingham risk score (Median)
Week 24Week 48
Combination Therapy01
Monotherapy01

[back to top]

Change From Baseline in Cluster of Differentiation 4 (CD4) Count Over Week 48

(NCT01391013)
Timeframe: Screening (Week -4), Week 1 (Day 1), Week 4, Week 12, Week 24, Week 36, Week 48, and follow-up (Week 52)

,
InterventionCD4 cells (Median)
Week 24Week 48
Combination Therapy-1260
Monotherapy6100.1

[back to top]

Change From Baseline in Insulin Sensitivity at Week 24 and Week 48: Median Change in Homeostasis Model Assessment of Insulin Resistance (HOMA-IR)

The Homeostatic Model Assessment (HOMA) is a method used to quantify insulin resistance and beta-cell function. HOMA-IR is reflected in the diminished effect of insulin on hepatic glucose production. HOMA-IR is calculated as: (Glucose [mg/dL] X Insulin [pmol/L]) / (405 X 6.945). Higher scores indicate worse insulin resistance. (NCT01391013)
Timeframe: Baseline, Week 24, and Week 48

,
InterventionHOMA score (Median)
Week 24Week 48
Combination Therapy-0.3-0.5
Monotherapy-0.2-0.6

[back to top]

Change From Baseline to Week 48 in Circulating Endothelial Cells

(NCT01391013)
Timeframe: Baseline to Week 48

,
InterventionEndothelial cells (Median)
BaselineWeek 48
Combination Therapy14.664
Monotherapy5.0937

[back to top]

Change From Baseline to Week 24 in Brachial Artery Flow Mediated Vasodilatation (FMD): Median Change in FMD (%)

Brachial artery FMD is calculated as the percentage increase in brachial artery diameter with hyperemia (an increase in the quantity of blood flow to a body part) induced relative to the resting brachial artery diameter. Percentage of brachial artery diameter is measured as FMD diameter/basal diameter. (NCT01391013)
Timeframe: Baseline (Day 1 of Week 1) to Week 24

InterventionPercentage of brachial artery diameter (Median)
Monotherapy-4.8
Combination Therapy-0.6

[back to top]

Change From Baseline to Week 48 in Lumbar Z Score: Median Change in Lumbar Z Score

Z score is used to calculate bone mineral density (calcium and other types of minerals) in an area of the bone. Z score is the number of standard deviations above or below the mean for the participant's age, sex and ethnicity. This score is calculated from participant's age, gender and race and skeletal site. Z score has a mean of '0' and a standard deviation of '1'. Z score lower than its mean indicate low bone mineral density. (NCT01391013)
Timeframe: Baseline to Week 48

InterventionZ score (Median)
Monotherapy0.1
Combination Therapy0.0

[back to top]

Change From Baseline in Mean Triglycerides at Week 24 and Week 48: Median Change in Triglycerides

(NCT01391013)
Timeframe: Baseline, Week 24, and Week 48

,
Interventionmg/dL (Median)
Week 24Week 48
Combination Therapy-16
Monotherapy1524

[back to top]

Change From Baseline in Mean Low-density Lipoprotein (LDL) Cholesterol at Week 24 and Week 48: Median Change in LDL

(NCT01391013)
Timeframe: Baseline (Day1 of Week 1), Week 24, and Week 48

,
Interventionmg/dL (Median)
Week 24Week 48
Combination Therapy65
Monotherapy1714

[back to top]

Change From Baseline in Mean High-density Lipoprotein (HDL) Cholesterol at Week 24 and Week 48: Median Change in HDL

(NCT01391013)
Timeframe: Baseline, Week 24, and Week 48

,
Interventionmg/dL (Median)
Week 24Week 48
Combination Therapy-6-6
Monotherapy-1-4

[back to top]

Change From Baseline to Week 48 in Lumbar T Score: Median Change in Lumbar T Score

T score is used to calculate bone mineral density (calcium and other types of minerals) in an area of the bone. T score is the number of standard deviations above or below the mean for a healthy 30 year old adult of the same sex and ethnicity as a participant. This score is calculated from participant's age, gender and race and skeletal site. T score has a mean of '50' and a standard deviation of '10'. T score lower than its mean indicate low bone mineral density. (NCT01391013)
Timeframe: Baseline to Week 48

InterventionT score (Median)
Monotherapy0.1
Combination Therapy0.0

[back to top]

Change From Baseline to Week 48 in Precursors of Circulating Endothelial Cells

(NCT01391013)
Timeframe: Baseline to Week 48

,
InterventionEndothelial cells (Median)
BaselineWeek 48
Combination Therapy18108
Monotherapy16120

[back to top]

Number of Participants With a Human Immunodeficiency Virus- Ribonucleic Acid (HIV-RNA) Greater Than or Equal to 50 Copies/mL

(NCT01391013)
Timeframe: Screening (Week -4), Week 1 (Day 1), Week 4, Week 12, Week 24, Week 36, Week 48, and follow-up (Week 52)

,
InterventionParticipants (Number)
Screening (Week -4)Week 1 (Day 1)Week 4Week 12Week 24Week 36Week 48Follow up (Week 52)
Combination Therapy00011000
Monotherapy01000212

[back to top]

Change From Baseline to Week 48 in Leg Fat Content: Median Change in Leg Fat (Total)

Leg fat content will be analyzed by Dual Energy X-ray Absortiometry (DEXA scan). (NCT01391013)
Timeframe: Baseline to Week 48

InterventionPercentage of fat (Median)
Monotherapy-57
Combination Therapy-288

[back to top]

Change From Baseline to Week 48 in Femoral Neck Z Score: Median Change in Femoral Neck Z Score

Z score is used to calculate bone mineral density (calcium and other types of minerals) in an area of the bone. Z score is the number of standard deviations above or below the mean for the participant's age, sex and ethnicity. This score is calculated from participant's age, gender and race and skeletal site. Z score has a mean of '0' and a standard deviation of '1'. Z score lower than its mean indicate low bone mineral density. (NCT01391013)
Timeframe: Baseline to Week 48

InterventionZ score (Median)
Monotherapy0.2
Combination Therapy0.0

[back to top]

Change From Baseline to Week 48 in Femoral Neck T Score: Median Change in Femoral Neck T Score

T score is used to calculate bone mineral density (calcium and other types of minerals) in an area of the bone. T score is the number of standard deviations above or below the mean for a healthy 30 year old adult of the same sex and ethnicity as a participant. This score is calculated from participant's age, gender and race and skeletal site. T score has a mean of '50' and a standard deviation of '10'. T score lower than its mean indicate low bone mineral density. (NCT01391013)
Timeframe: Baseline to Week 48

InterventionT score (Median)
Monotherapy0.2
Combination Therapy-0.1

[back to top]

Change From Baseline to Week 48 in Brachial Artery FMD: Median Change in FMD (%)

Brachial artery FMD is calculated as the percentage increase in brachial artery diameter with hyperemia induced relative to the resting brachial artery diameter. Percentage of brachial artery diameter is measured as FMD diameter/basal diameter. (NCT01391013)
Timeframe: Baseline to Week 48

InterventionPercentage of brachial artery diameter (Median)
Monotherapy-4.4
Combination Therapy-3

[back to top]

Change From Baseline to Week 48 in Visceral Fat Content in Abdomen: Median Change in Visceral Abdominal Tissue (VAT)

Visceral fat content in abdomen will be analyzed with median change in VAT by an abdomen Computerized Tomography. (NCT01391013)
Timeframe: Baseline to Week 48

Interventioncm square (Median)
Monotherapy-4
Combination Therapy-4

[back to top]

Change in Levels of D-dimer From Baseline

Change in levels of D-dimer from baseline (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionng/ml (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-82
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-61

[back to top]

Change in CD4 Count From Baseline to Week 24

Change in CD4 count from baseline (week 0) to week 24 (NCT01400412)
Timeframe: Week 0, week 24

Interventioncells/mm^3 (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)165
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)127

[back to top]

Percent Change in Lumbar Spine Bone Mineral Density (BMD)

The percent change in bone mineral density (BMD) at lumbar spine (as measured by DXA scan) from baseline (week 0) to week 48. (NCT01400412)
Timeframe: Week 0, week 48

Interventionpercentage change (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-0.88
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-2.35

[back to top]

Percent Change in Expression of RANKL+ on CD8+ T Cells From Baseline to Week 48

percentage change is defined as [ (week 48 - week 0) / week 0 ] * 100% (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionpercentage change (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-20.7
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-17.0

[back to top]

Change in CD4 Count From Baseline to Week 48

Change in CD4 count from baseline (week 0) to week 48 (NCT01400412)
Timeframe: Week 0, week 48

Interventioncells/mm^3 (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)234
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)188

[back to top]

Number of Participants Who Experienced Bone Fractures

Number of participants who experienced bone fractures during the study (NCT01400412)
Timeframe: From study treatment initiation to week 48

Interventionparticipants (Number)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)2
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)2

[back to top]

Percent Change in Expression of CD57+ on CD8+ T Cells From Baseline to Week 48

percentage change is define as [ (week 48 - week 0) / week 0 ] * 100% (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionpercentage change (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-3.5
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-4.6

[back to top]

Cumulative Probability of Virologic Failure by Week 48

"Confirmed virologic failure is defined as confirmed plasma HIV-1 RNA levels > 1000 copies/mL at or after week 16 and before week 24, or confirmed HIV-1 RNA levels> 200 copies/mL at or after week 24. Participants who discontinued the study with an unconfirmed virologic failure (HIV-1 RNA > 1000 copies at 16 weeks or HIV-1 RNA level > 200 copies/mL at or after week 24) are considered as virologic failures at the study visit week of the unconfirmed value. Time to virologic failure is defined as the time from study entry to the planned visit week of the initial failure.~Product-limit estimates for the survival function were used to estimate the cumulative probability of virologic failure over time and its corresponding 95% confidence interval for each treatment group." (NCT01400412)
Timeframe: From study treatment initiation to week 48

Interventioncumulative probability per 100 persons (Number)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)6
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)5

[back to top]

Percent Change From Baseline in Total Hip Bone Mineral Density (BMD)

The primary endpoint is the percent change in bone mineral density (BMD) at total hip (as measured by DXA scan) from baseline (week 0) to week 48. (NCT01400412)
Timeframe: Week 0, week 48

Interventionpercentage change (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-1.51
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-2.40

[back to top]

Percentage Change in Expression of CD38+/HLA-DR+ on CD8+ T Cells From Baseline to Week 48

percentage change is defined as [ (week 48 - week 0) / week 0 ] * 100% (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionpercentage change (Median)
MVC Arm: DRV/r + MVC + FTC + TDF Placebo-59.5
TDF Arm: DRV/r + TDF + FTC + MVC Placebo-60.9

[back to top]

Percentage Change in Expression of CD38+/HLA-DR+ on CD4+ T Cells From Baseline to Week 48

percentage change is define as [ (week 48 - week 0) / week 0 ] * 100% (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionpercentage change (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-52.1
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-48.6

[back to top]

Percent Change in Expression of CD28+/CD57+ on CD8+ T Cells From Baseline to Week 48

percentage change is define as [ (week 48 - week 0) / week 0 ] * 100% (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionpercentage change (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-9.1
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-11.2

[back to top]

Percent Change in Expression of CD28+ on CD8+ T Cells From Baseline to Week 48

percentage change is define as [ (week 48 - week 0) / week 0 ] * 100% (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionpercentage change (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)11.9
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)14.0

[back to top]

CD8+ T-cell Change From Baseline to Week 48

CD8+ T-cell change from baseline to week 48 (NCT01400412)
Timeframe: At weeks 0 and 48

Interventioncell/mm^3 (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-6
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-109

[back to top]

Change in Levels of sCD163 From Baseline to Week 48

Change in levels of soluble CD163 from baseline to week 48 (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionng/ml (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-250
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-258

[back to top]

Change in Levels of sCD14 From Baseline

Change in levels of soluble CD14 from baseline (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionng/ml (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-103
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-10

[back to top]

Change in Level of IP-10 From Baseline to Week 48

Change in level of Interferon gamma-induced protein 10 (IP-10) from baseline to week 48 (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionpg/ml (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-198
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-170

[back to top]

Number of Participants Who Developed Grade 3 or 4 Primary Adverse Events

"Grade 3 or 4 primary adverse events includes primary signs/symptoms, primary laboratory abnormalities, or primary diagnoses.~See DAIDS AE Grading Table Version 1.0, Dec 2004 (Clarification, Aug 2009)" (NCT01400412)
Timeframe: From study treatment initiation to week 48

Interventionparticipants (Number)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)16
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)22

[back to top]

Number of Participants Who Died During the Study

Number of participants who died during the study (NCT01400412)
Timeframe: From study treatment initiation to week 48

Interventionparticipants (Number)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)0
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)0

[back to top]

Change in Levels of IL-6 From Baseline to Week 48

Change in levels of Interleukin 6 (IL-6) from baseline to week 48 (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionpg/ml (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-0.21
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-0.12

[back to top]

NK-104 AUC

(NCT01422369)
Timeframe: 16 Days

Interventionng * h/mL (Mean)
NK-104192.00

[back to top]

Number of Participants With at Least One Adverse Event.

(NCT01422369)
Timeframe: 16 Days

InterventionParticipants (Number)
All Subjects2

[back to top]

Number of Participants With Greater Than 95% Adherence at 48 Weeks

Characterize adherence to once-daily versus twice-daily darunavir/ritonavir containing regimens using the Modified Medication Adherence Self-Report Inventory (M-MASRI) scale (NCT01423812)
Timeframe: Within 48 weeks of randomization to study medications

InterventionParticipants (Count of Participants)
Once-daily Darunavir and Ritonavir22
Twice-daily Darunavir and Ritonavir20

[back to top]

Secondary Efficacy Endpoints

•Proportion of subjects with plasma HIV-1 RNA >200 c/mL at Week 24 (NCT01423812)
Timeframe: Within 24 weeks after randomization to study medication

InterventionParticipants (Count of Participants)
Once-daily Darunavir and Ritonavir0
Twice-daily Darunavir and Ritonavir0

[back to top]

Secondary Efficacy Endpoints

•Proportion of subjects with plasma HIV-1 RNA <40 c/mL at Week 24 (NCT01423812)
Timeframe: week 24

InterventionParticipants (Count of Participants)
Once-daily Darunavir and Ritonavir29
Twice-daily Darunavir and Ritonavir25

[back to top]

Primary Efficacy Endpoint for Virologic Suppression in HIV-infected Subjects

"Proportion of subjects with plasma HIV-1 RNA <40 c/mL at Week 48 using a Missing, Switch, or Discontinuation = Failure (MSDF) algorithm as codified by the FDA's snapshot algorithm" (NCT01423812)
Timeframe: 48 weeks after randomization to study medication

Interventionparticipants (Number)
Once-daily Darunavir/Ritonavir27
Twice-daily Darunavir/Ritonavir25

[back to top]

Change in Total Cholesterol From Baseline to 48 Weeks

Absolute change in lipid parameter (total cholesterol mg/dl) of once-daily versus twice-daily darunavir/ritonavir containing regimens over 48 weeks (NCT01423812)
Timeframe: Within 48 weeks of randomization baseline to study medications

Interventionmg/dl (Mean)
Once-daily Darunavir and Ritonavir-14.2
Twice-daily Darunavir and Ritonavir-3.8

[back to top]

Absolute Value Change in CD4+ From Baseline to Week 48

Absolute value increase in CD4+ cells/mm3 from baseline to week 48 (NCT01423812)
Timeframe: 48 weeks after randomization baseline to study medications

Interventioncells/mm3 (Mean)
Once-daily Darunavir and Ritonavir19
Twice-daily Darunavir and Ritonavir39

[back to top]

Assessment of Virologic Failure

•Assess the development of viral resistance in subjects experiencing virological failure (NCT01423812)
Timeframe: Within 48 weeks of randomization to study medications

Interventionparticipants with resistance for failure (Number)
Once-daily Darunavir and Ritonavir0
Twice-daily Darunavir and Ritonavir0

[back to top]

Area Under Curve From 0 to ∞ Hours (AUC0-∞)

AUC0-∞ represents the area under the concentration curve of the analyte in plasma from 0 extrapolated to infinity. (NCT01426958)
Timeframe: 0, 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 10, 12, 24, 36, 48, 72, 96, 120 hours post

Interventionng*h/mL (Geometric Mean)
Afatinib426
Afatinib + Concomittant Rtv515
Afatinib + Timed Rtv475

[back to top]

Area Under Curve From 0 to tz (AUC0-tz)

AUC0-tz represents the area under the concentration curve of the analyte in plasma from 0 to the time of the last quantifiable plasma contentration of the analyte. (NCT01426958)
Timeframe: 0, 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 10, 12, 24, 36, 48, 72, 96, 120 hours post

Interventionng*h/mL (Geometric Mean)
Afatinib392
Afatinib + Concomittant Rtv478
Afatinib + Timed Rtv438

[back to top]

Maximum Concentration (Cmax)

Cmax represents the maximum concentration of the analyte in plasma. (NCT01426958)
Timeframe: 0, 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 10, 12, 24, 36, 48, 72, 96, 120 hours post

Interventionng/mL (Geometric Mean)
Afatinib19.5
Afatinib + Concomittant Rtv20.7
Afatinib + Timed Rtv20.7

[back to top]

Change From Baseline in Global Neurocognitive Performance z-Score

Change in neurocognitive function of DRV/rtv monotherapy versus triple therapy containing DRV/rtv over 48 and 96 weeks. Neurocognitive function will be measured by Hopkins Verbal Learning Test (verbal learning and memory), Colour Trail Test (psychomotor speed and cognitive flexibility) and Grooved Pegboard Test (psychomotor speed and fine motor function). Higher values for change in z-score represent an improvement in Neurocognitive Performance (NP). (NCT01448707)
Timeframe: Baseline, Week 48 and 96

,
InterventionUnits on a Scale (Mean)
Change at Week 48Change at Week 96
DRV/r0.390.63
DRV/r + 2NRTIs0.420.57

[back to top]

Virologic Response (Food Drug and Administration [FDA] Snapshot, Switch = Failure)

The percentage of participants who have plasma human immunodeficiency virus type-1 (HIV-1) ribonucleic acid (RNA) levels <50 copies/milliliters [mL] after 96 weeks of follow-up after switching to DRV/ritonavir(rtv) monotherapy versus triple therapy containing DRV/rtv. Switch = Failure is defined as switch in background nucleoside/nucleotide reverse transcriptase inhibitors (N[t]RTIs) not permitted by the trial protocol. (NCT01448707)
Timeframe: Week 96

InterventionPercentage of Participants (Number)
DRV/r75.2
DRV/r + 2NRTIs85.3

[back to top]

Virologic Response (Food Drug and Administration [FDA] Snapshot, Switch = Failure)

The percentage of participants who have plasma human immunodeficiency virus type-1 (HIV-1) ribonucleic acid (RNA) levels <50 copies/milliliters [mL] after 48 weeks of follow-up. Switch = Failure is defined as switch in background nucleoside/nucleotide reverse transcriptase inhibitors (N[t]RTIs) not permitted by the trial protocol or plasma HIV-1 RNA assessment closest to target date of the analysis time point window (44-52 weeks) and next/confirmation of Plasma HIV-1 RNA in the analysis time point window above the threshold or discontinuation for any other reason. (NCT01448707)
Timeframe: Week 48

InterventionPercentage of Participants (Number)
DRV/r87.0
DRV/r + 2NRTIs95.0

[back to top]

Virologic Response (FDA Snapshot, Switch Included)

The percentage of participants who have plasma human immunodeficiency virus type-1 (HIV-1) ribonucleic acid (RNA) levels <50 copies/milliliters [mL] after 48 and 96 weeks of follow-up after switching to DRV/ritonavir(rtv) monotherapy versus triple therapy containing DRV/rtv. Switch included is defined as all participants who discontinued randomized medication were followed up on their subsequent treatment. (NCT01448707)
Timeframe: Week 48 and 96

,
InterventionPercentage of Participants (Number)
Week 48Week 96
DRV/r93.089.3
DRV/r + 2NRTIs96.589.9

[back to top]

Time to Loss of Virologic Response

Time (in days) it takes to show loss of response per time to loss of virologic response (TLOVR) algorithm: confirmed HIV-1 RNA >= 50 copies/mL or premature discontinuation. (NCT01448707)
Timeframe: Baseline up to Week 96 or early withdrawal

InterventionDays (Median)
DRV/rNA
DRV/r + 2NRTIsNA

[back to top]

Number of Participants Reporting Resistance Mutations With Confirmed Virologic Failure Who Have HIV RNA >400 Copies/mL and Genotype Resistance Results

The viral genotype of participants treated with DRV/rtv monotherapy versus triple therapy containing DRV/rtv over 48 and 96 weeks. Genotypic resistance (number of resistance mutations) at any time point when a participant had a confirmed plasma VL >400 copies/mL after randomization was performed per treatment group for the ITT population. Results were summarized based on individual treatment received: Darunavir resistance mutations, non-nucleoside reverse transcriptase inhibitor (NNRTI) mutations, nucleoside reverse transcriptase inhibitor (NRTI) mutations, protease inhibitor (PI) resistance mutations, PR mutations, RT mutations, extended NNRTI mutations, primary PI mutations. (NCT01448707)
Timeframe: Over 48 and 96 Weeks

,
InterventionParticipants (Number)
Participans with HIV RNA >= 400 copies/mLNumber of 0 Darunavir resistance mutationsNumber of 0 NNRTI mutationsNumber of 1 NNRTI mutationsNumber of 1 PI resistance mutationsNumber of 5 PI resistance mutationsNumber of 6 PI resistance mutationsNumber of 11 PR mutationsNumber of 15 PR mutationsNumber of 7 PR mutationsNumber of 14 RT mutationsNumber of 16 RT mutationsNumber of 33 RT mutationsNumber of extended 0 NNRTI mutationsNumber of extended 1 NNRTI mutationsNumber of primary 0 PI mutationsNumber of participants with no mutations
DRV/r12201010111012020
DRV/r + 2NRTIs21010101000100110

[back to top]

Number of Participants Reporting Treatment-Emergent Phenotypic Drug Resistance

The loss of treatment options of DRV/rtv monotherapy versus triple therapy containing DRV/rtv at Weeks 48 and 96, as defined by treatment-emergent phenotypic drug resistance. Drug resistance is classified as: 1) Confirmed HIV RNA >= 400 copies/mL, 2) Post-baseline phenotypic data and 3) Phenotypic resistance to any of the drug classes (NRTI, NNRTI, or PI). (NCT01448707)
Timeframe: At Weeks 48 and 96

,
InterventionParticipants (Number)
Confirmed HIV RNA >= 400 copies/mLPost-baseline phenotypic dataPhenotypic resistance to any of the drug classes
DRV/r120
DRV/r + 2NRTIs210

[back to top]

Time to Virologic Suppression (<50 Copies/mL) Through Week 48

The time to viral suppression (i.e. first viral load value <50 copies/mL) through Week 48 was derived and summarized using Kaplan-Meier plots. Participants who withdrew for any reason without having suppressed prior to the analysis were censored. Confidence intervals were estimated using the Brookmeyer-Crowley method. (NCT01449929)
Timeframe: From Baseline through Week 48

InterventionDays (Median)
DTG 50 mg QD28.0
DRV 800 mg + RTV 100 mg QD85.0

[back to top]

Change From Baseline in Fasting Low-density Lipoprotein (LDL) Cholesterol Through Week 48

Fasting LDL cholesterol change from Baseline was analyzed. Values represented are for adjusted means. Estimates are calculated from a repeated measures model including the following covariates: treatment, visit, Baseline plasma HIV-1 RNA, background dual NRTI therapy, Baseline LDL cholesterol, treatment*visit interaction and Baseline LDL cholesterol*visit interaction. Change from Baseline was calculated as the post-Baseline value minus the Baseline value. Only those participants with data available at the specified time points were analyzed. (NCT01449929)
Timeframe: From Baseline through Week 48

InterventionMillimoles per liter (mmol/L) (Mean)
DTG 50 mg QD0.07
DRV 800 mg + RTV 100 mg QD0.37

[back to top]

Number of Participants (Par.) With Detectable Virus That Has Genotypic or Phenotypic Evidence of Treatment-emergent Resistance to DTG, DRV+RTV and Other On-study ART at Time of Protocol Defined Virology Failure (PDVF)

An assessment was made of every change across all amino acids within the integrase (IN), reverse transcriptase (RT), and Protease (PRO) encoding region at Baseline and at time of suspected PDVF. PDVF is defined as the confirmed plasma HIV-1 RNA >200 c/mL >=Week 24. PDVF Genotypic Population included all participants in the mITT-E population with available on-treatment genotypic resistance data, at time of PDVF. Only those participants with data available at the specified time points were analyzed. (NCT01449929)
Timeframe: Baseline until PDVF up to Week 48

InterventionParticipants (Number)
DTG 50 mg OD0
DRV 800 mg + RTV 100 mg OD0

[back to top]

Percentage of Participants With Grade 2 or Higher Abnormalities in Fasting LDL Cholesterol Through Week 48

Hematology and clinical chemistry data were summarized according to the division of AIDS (DAIDS) table for grading the Severity of adverse events, version 1.0. Grade 1, Mild; Grade 2, Moderate; Grade 3 (G3), Severe; Grade 4 (G4), Life-threatening or disabling; Grade 5, Death. Data are presented for which an increase in fasting LDL cholesterol to Grade 2 or higher occurred. Only those participants with data available at the specified time points were analyzed. (NCT01449929)
Timeframe: From Baseline through Week 48

InterventionPercentage of Participants (Number)
DTG 50 mg QD2
DRV 800 mg + RTV 100 mg QD7

[back to top]

Percentage of Participants With Plasma HIV-1 RNA <400 c/mL at Week 48

"The percentage of participants with Plasma HIV-1 RNA <400 c/mL at Week 48 was assessed MSDF, as codified by the FDA snapshot algorithm. This algorithm treated all participants without HIV-1 RNA data at Week 48 as nonresponders, as well as participants who switched their concomitant ART prior to Week 48 as follows: background ART substitutions non-permitted per protocol (one background ART substitution was permitted for safety or tolerability); background ART substitutions permitted per protocol unless the decision to switch was documented as being before or at the first on-treatment visit where HIV-1 RNA was assessed. Otherwise, virologic success or failure was determined by the last available HIV-1 RNA assessment while the participant was on-treatment in the snapshot window (Week 48 +/- 6 weeks)." (NCT01449929)
Timeframe: Week 48

InterventionPercentage of participants (Number)
DTG 50 mg QD92
DRV 800 mg + RTV 100 mg QD87

[back to top]

Percentage of Participants With Plasma Human Immunodeficiency Virus-1 (HIV-1) Ribonucleic Acid (RNA) <50 Copies/Milliliter (c/mL) at Week 48

"Assessment was done using Missing, Switch or Discontinuation = Failure (MSDF), as codified by the Food and Drug Administration (FDA) snapshot algorithm.This algorithm treated all participants without HIV-1 RNA data at Week 48 as nonresponders, as well as participants who switched their concomitant ART prior to Week 48 as follows: background ART substitutions non-permitted per protocol (one background ART substitution was permitted for safety or tolerability); background ART substitutions permitted per protocol unless the decision to switch was documented as being before or at the first on-treatment visit where HIV-1 RNA was assessed. Otherwise, virologic success or failure was determined by the last available HIV-1 RNA assessment while the participant was on-treatment in the snapshot window (Week 48 +/- 6 weeks). Modified Intent-To-Treat Exposed (mITT-E) Population:all randomized participants who received at least one dose of investigational product" (NCT01449929)
Timeframe: Week 48

InterventionPercentage of participants (Number)
DTG 50 mg QD90
DRV 800 mg + RTV 100 mg QD83

[back to top]

Change From Baseline in Acquired Immune Deficiency Syndrome (AIDS) Clinical Trials Group (ACTG) Symptom Distress Module (SDM) Bother Score at Week 4, Week 24, and Week 48

SDM is a 20-item self-reported measure that addresses the presence and perceived distress linked to symptoms commonly associated with HIV or its treatment. Each item is rated from 0 to 4 where 0 (complete absence of symptom) and 4 (very bothersome symptom). Overall score calculated as the sum of the scores for each of the 20 items of the questionnaire and ranged from 0 (best health) and 80 (worst health). Values represented are for adjusted mean. Estimates are calculated from an ANCOVA model adjusting for age, sex, race, baseline viral load, background dual NRTI therapy and baseline symptom bother score. Change from Baseline was calculated as the post-Baseline value minus the Baseline value. A positive change from Baseline indicates a decline in a participant's quality of life over that period. (NCT01449929)
Timeframe: Baseline, Week 4, Week 24, and Week 48

,
InterventionScores on a scale (Mean)
Week 4, n=218, 210Week 24, n=222, 214Week 48, n= 222, 215
DRV 800 mg + RTV 100 mg QD-2.19-1.65-0.77
DTG 50 mg QD-3.20-2.71-2.46

[back to top]

Change From Baseline in CD4+ and CD8+ Cell Counts

Change from Baseline in CD4+ cell counts was assessed at Weeks 4, 8, 12, 16, 36 and 48. Change from Baseline in CD8+ cell counts was assessed at Weeks 4, 12, 24 and 48. Change from Baseline was calculated as the post-Baseline value minus the Baseline value. Only those participants available at the specified time points were analyzed (represented by n=X, X in the category titles). Different participants may have been analyzed for different visits and parameters, so the overall number of participants analyzed reflects everyone in the mITT-E Population. (NCT01449929)
Timeframe: Baseline and Weeks 4, 8, 12, 16, 36 and 48 for CD4+ and Baseline and Weeks 4, 12, 24 and 48 for CD8+

,
InterventionCells per millimeters cubed (cells/mm^3) (Mean)
CD4+ cell count, Week 4, n=237, 236CD4+ cell count, Week 8, n=236, 236CD4+ cell count, Week 12, n=234, 228CD4+ cell count, Week 16, n=227, 227CD4+ cell count, Week 24, n=233, 227CD4+ cell count, Week 36, n=232, 218CD4+ cell count, Week 48, n=227, 212CD8+ cell count, Week 4, n=235, 235CD8+ cell count, Week 12, n=231, 227CD8+ cell count, Week 24, n=231, 224CD8+ cell count, Week 48, n=224, 210
DRV 800 mg + RTV 100 mg QD75.6118.8131.8146.1164.3186.5215.4-3.7-68.9-132.9-162.1
DTG 50 mg QD80.1126.9135.2156.8165.1206.1243.8-47.4-42.0-108.0-109.5

[back to top]

Change From Baseline in EQ-5D Thermometer Scores at Week 24 and Week 48

The European Quality of Life -5 Dimensions (EQ-5D) is a 5-question quality of life instrument that provides a utility score and visual analogue scale score that describes the participants' health status. The primary reason for including the EQ-5D is to elicit utility values for potential cost-effectiveness analysis for submission to health technology assessment agencies. Thermometer score is based on a visual analogue scale (VAS) ranging from 100 (best imaginable health state) to 0 (worst imaginable health state).Values represented are for adjusted mean. Estimates are calculated from an ANCOVA model adjusting for age, sex, race, Baseline viral load, background dual NRTI therapy and Baseline EQ-5D thermometer score. Change from Baseline was calculated as the post-Baseline value minus the Baseline value. Only those participants available at the specified time points were analyzed (represented by n=X, X in the category titles). (NCT01449929)
Timeframe: Baseline, Week 24, and Week 48

,
InterventionScores on a scale (Mean)
Week 24, n=221, 216Week 48, n=224, 220
DRV 800 mg + RTV 100 mg QD5.966.95
DTG 50 mg QD4.955.78

[back to top]

Change From Baseline in European Quality of Life -5 Dimensions (EQ-5D) Utility Scores at Week 24 and Week 48

The EQ-5D is a 5-question quality of life instrument that provides a utility score and visual analogue scale score that describes the participants' health status. The primary reason for including the EQ-5D is to elicit utility values for potential cost-effectiveness analysis for submission to health technology assessment agencies. The EQ-5D total score ranges from 0 (worst health state) to 1 (perfect health state) and 1 reflects the best outcome. Values represented are for adjusted mean. Estimates are calculated from an ANCOVA model adjusting for age, sex, race, baseline viral load, background dual NRTI therapy and Baseline EQ-5D utility score. Change from Baseline was calculated as the post-Baseline value minus the Baseline value. Only those participants available at the specified time points were analyzed (represented by n=X, X in the category titles). (NCT01449929)
Timeframe: Baseline, Week 24, and Week 48

,
InterventionScores on a scale (Mean)
Week 24, n=217, 213Week 48, n=224, 217
DRV 800 mg + RTV 100 mg QD0.020.01
DTG 50 mg QD0.000.01

[back to top]

Number of Participants With HIV-1 Associated Disease Progression With the Indicated Shift to CDC Class C, or New CDC Class C or Death at Week 48

The number of participants with HIV-1 disease progression (AIDS or death) was assessed per the Centers for Disease Control and Prevention (CDC) 1993 revised classification system for HIV infection and expanded surveillance case definition for AIDS among adolescents and adults. The CDC classifies HIV infection as Category A (participants with asymptomatic HIV infection, acute HIV infection with accompanying illness, or persistent generalized lymphadenopathy), Category B (participants with symptomatic non-AIDS condition, i.e., conditions that are attributed to HIV infection or are indicative of a defect in cell-mediated immunity; or conditions are considered by physicians to have a clinical course or to require management that is complicated by HIV infection), and Category C (includes AIDS indicator conditions as defined by diagnostic or presumptive measures). (NCT01449929)
Timeframe: Week 48

InterventionParticipants (Number)
DTG 50 mg QD0
DRV 800 mg + RTV 100 mg QD0

[back to top]

Human Immunodeficiency Virus Treatment Satisfaction Questionnaire (HIVTSQ) Convenience Score at Week 4, Week 24, and Week 48

Participant treatment satisfaction was measured using the self-reported scale (HIVTSQ), which consists of 10 items (1-satisfaction, 2-HIV control, 3-adverse effects, 4-level of demand, 5-convenience, 6-flexibility, 7-knowledge, 8-life habits, 9-recommendability, and 10-willingness to continue). Items are scored from 0 (very dissatisfied) to 6 (very satisfied), other than item 4, which has an inverted score from 6 (very demanding) to 0 (very undemanding). The convenience score is the score for item 5 (range: 0-6). Only those participants available at the specified time points were analyzed (represented by n=X, X in the category titles). Different participants may have been analyzed for different visits, so the overall number of participants analyzed reflects everyone in the HIVTSQ mITT-E population. (NCT01449929)
Timeframe: Week 4, Week 24, and Week 48

,
InterventionScores on a scale (Mean)
Week 4, n=204, 190Week 24, n=211, 200,Week 48, n=212, 201
DRV 800 mg + RTV 100 mg QD5.25.45.4
DTG 50 mg QD5.65.65.7

[back to top]

Human Immunodeficiency Virus Treatment Satisfaction Questionnaire (HIVTSQ) Lifestyle/Ease Sub Score at Week 4, Week 24, and Week 48

Participant treatment satisfaction was measured using the self-reported scale (HIVTSQ), which consists of 10 items (1-satisfaction, 2-HIV control, 3-adverse effects, 4-level of demand, 5-convenience, 6-flexibility, 7-knowledge, 8-life habits, 9-recommendability, and 10-willingness to continue). Items are scored from 0 (very dissatisfied) to 6 (very satisfied), other than item 4, which has an inverted score from 6 (very demanding) to 0 (very undemanding). The lifestyle/ease score is the sum of items 4, 5, 6, 7 and 8 (range: 0-30). Only those participants available at the specified time points were analyzed (represented by n=X, X in the category titles). Different participants may have been analyzed for different visits, so the overall number of participants analyzed reflects everyone in the HIVTSQ mITT-E population. (NCT01449929)
Timeframe: Week 4, Week 24, and Week 48

,
InterventionScores on a scale (Mean)
Week 4, n=202, 190Week 24, n=210, 199Week 48, n=211, 201
DRV 800 mg + RTV 100 mg QD25.826.626.6
DTG 50 mg QD26.727.527.6

[back to top]

Human Immunodeficiency Virus Treatment Satisfaction Questionnaire (HIVTSQ) Total Score at Week 4, Week 24, and Week 48

Participant treatment satisfaction was measured using the self-reported scale (HIVTSQ), which consists of 10 items (1-satisfaction, 2-HIV control, 3-adverse effects, 4-level of demand, 5-convenience, 6-flexibility, 7-knowledge, 8-life habits, 9-recommendability, and 10-willingness to continue). Items are scored from 0 (very dissatisfied) to 6 (very satisfied), other than item 4, which has an inverted score from 6 (very demanding) to 0 (very undemanding). The treatment satisfaction score (range: 0-60) was the sum of the individual items. HIVTSQ mITT-E Population=Only participants from USA, France, Germany, Italy, Spain for whom valid translations were available from the mITT-E Population. Only those participants available at the specified time points were analyzed (represented by n=X, X in the category titles). Different participants may have been analyzed for different visits, so the overall number of participants analyzed reflects everyone in the HIVTSQ mITT-E population. (NCT01449929)
Timeframe: Week 4, Week 24, and Week 48

,
InterventionScores on a scale (Mean)
Week 4, n=206, 192Week 24, n= 211, 200Week 48, n=212, 201
DRV 800 mg + RTV 100 mg QD52.454.354.5
DTG 50 mg QD54.156.156.1

[back to top]

Number of Participants With the Indicated Grade 3 and Grade 4 Maximum Post-Baseline Chemistry and Hematology Laboratory Toxicities

Hematology and clinical chemistry data were summarized according to the division of AIDS (DAIDS) table for grading the Severity of adverse events, version 1.0. Grade 1, Mild; Grade 2, Moderate; Grade 3 (G3), Severe; Grade 4 (G4), Life-threatening or disabling; Grade 5, Death. Data are presented for only those parameters for which an increase to Grade 3 or Grade 4 occurred. (NCT01449929)
Timeframe: From Baseline through Week 48

,
InterventionParticipants (Number)
Alanine Amino Transferase, G3Alanine Amino Transferase, G4Aspartate Amino Transferase, G3Aspartate Amino Transferase, G4Cholesterol, G3Creatine Kinase, G3Creatine Kinase, G4Hyperglycaemia, G3Hypoglycaemia, G3LDL Cholesterol, G3Lipase, G3Lipase, G4Phosphorus, inorganic, G3Total Bilirubin, G3Triglycerides, G3Triglycerides, G4Hemoglobin, G3Hemoglobin, G4Platelet count, G4Total Neutrophils, G3Total Neutrophils, G4
DRV 800 mg + RTV 100 mg QD133035421650702100101
DTG 50 mg QD216208810252711011053

[back to top]

Change From Baseline in Plasma HIV-1 RNA (log10 c/mL) at Weeks 4, 8, 12, 16, 24, 36 and 48

Change from Baseline in plasma HIV-1 RNA (log10 c/mL) was assessed at Weeks 4, 8, 12, 16, 24, 36 and 48 . Change from Baseline was calculated as the post-Baseline value minus the Baseline value. Only those participants available at the specified time points were analyzed (represented by n=X, X in the category titles). Different participants may have been analyzed for different visits, so the overall number of participants analyzed reflects everyone in the mITT-E Population. (NCT01449929)
Timeframe: Baseline, Weeks 4, 8, 12, 16, 24, 36 and 48

,
InterventionLog10 copies per mL (Mean)
Week 4, n=238, 235Week 8, n=237, 236Week 12, n=234, 227Week 16, n=229, 228Week 24, n=234, 227Week 36, n=232, 218Week 48, n=227, 212
DRV 800 mg + RTV 100 mg OD-2.01-2.40-2.61-2.71-2.83-2.85-2.86
DTG 50 mg OD-2.80-2.86-2.88-2.86-2.86-2.87-2.89

[back to top]

Percentage of Participants With Sustained Virologic Response 12 Weeks (SVR12) Post-treatment

Sustained Virologic Response 12 (SVR12) is defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) less than the lower limit of quantification (< LLOQ; < 25 IU/mL) 12 weeks after the last dose of study drug. Participants with missing data were imputed as failures. (NCT01458535)
Timeframe: Post-treatment Day 1 to Post-treatment Week 12

Interventionpercentage of participants (Number)
ABT-450/r and ABT-267 Plus RBV in Genotype 1 Participants100
ABT-450/r and ABT-267 Plus RBV in Genotype 2 Participants80.0
ABT-450/r and ABT-267 Plus RBV in Genotype 3 Participants50.0
ABT-450/r and ABT-267 in Genotype 1 Participants60.0
ABT-450/r and ABT-267 in Genotype 2 Participants60.0
ABT-450/r and ABT-267 in Genotype 3 Participants9.1

[back to top]

Percentage of Participants With Hepatitis C Virus (HCV) Ribonucleic Acid (RNA) Below the Lower Limit of Quantitation (LLOQ) at Week 4 Rapid Virologic Response (RVR)

Analysis of percentage of participants with hepatitis C virus ribonucleic acid less than the lower limit of quantitation (< 25 IU/mL). Participants with missing data were imputed as failures. (NCT01458535)
Timeframe: Week 4

Interventionpercentage of participants (Number)
ABT-450/r and ABT-267 Plus RBV in Genotype 1 Participants100
ABT-450/r and ABT-267 Plus RBV in Genotype 2 Participants100
ABT-450/r and ABT-267 Plus RBV in Genotype 3 Participants90.0
ABT-450/r and ABT-267 in Genotype 1 Participants100
ABT-450/r and ABT-267 in Genotype 2 Participants90.0
ABT-450/r and ABT-267 in Genotype 3 Participants27.3

[back to top]

Percentage of Participants With Hepatitis C Virus (HCV) Ribonucleic Acid (RNA) < 1000 International Units Per Milliliter (IU/mL)

Analysis of participants with HCV RNA levels below 1000 IU/mL at Week 2. Participants with missing data were imputed as failures. (NCT01458535)
Timeframe: Week 2

Interventionpercentage of participants (Number)
ABT-450/r and ABT-267 Plus RBV in Genotype 1 Participants100
ABT-450/r and ABT-267 Plus RBV in Genotype 2 Participants100
ABT-450/r and ABT-267 Plus RBV in Genotype 3 Participants100
ABT-450/r and ABT-267 in Genotype 1 Participants100
ABT-450/r and ABT-267 in Genotype 2 Participants100
ABT-450/r and ABT-267 in Genotype 3 Participants100

[back to top]

Percentage of Participants Who Experienced Virologic Relapse Through End of Post Treatment Period (up to 48 Weeks)

Virologic relapse is defined as confirmed hepatitis C virus (HCV) ribonucleic acid (RNA) >= lower limit of quantitation (LLOQ) (2 consecutive measurements >= LLOQ) at any point in the post-treatment period among participants with HCV RNA < LLOQ at the end of treatment. Participants with missing data were imputed as failures. (NCT01458535)
Timeframe: Post-treatment Day 1 to Post-treatment Week 48

Interventionpercentage of participants (Number)
ABT-450/r and ABT-267 Plus RBV in Genotype 1 Participants0
ABT-450/r and ABT-267 Plus RBV in Genotype 2 Participants0
ABT-450/r and ABT-267 Plus RBV in Genotype 3 Participants28.6
ABT-450/r and ABT-267 in Genotype 1 Participants22.2
ABT-450/r and ABT-267 in Genotype 2 Participants22.2
ABT-450/r and ABT-267 in Genotype 3 Participants50.0

[back to top]

Percentage of Participants With Hepatitis C Virus (HCV) Ribonucleic Acid (RNA) Suppressed Below the Lower Limit of Quantitation (LLOQ) From Week 4 Through Week 12 [(Extended Rapid Virologic Response (eRVR)]

Analysis of the percentage of participants with hepatitis C virus ribonucleic acid less than the lower limit of quantitation (< 25 IU/mL). Participants with missing data were imputed as failures. (NCT01458535)
Timeframe: Week 4 through Week 12

Interventionpercentage of participants (Number)
ABT-450/r and ABT-267 Plus RBV in Genotype 1 Participants100
ABT-450/r and ABT-267 Plus RBV in Genotype 2 Participants90.0
ABT-450/r and ABT-267 Plus RBV in Genotype 3 Participants70.0
ABT-450/r and ABT-267 in Genotype 1 Participants90.0
ABT-450/r and ABT-267 in Genotype 2 Participants80.0
ABT-450/r and ABT-267 in Genotype 3 Participants18.2

[back to top]

Percentage of Participants With Virologic Failure During Treatment

Virologic failure during treatment is defined as a participant meeting any virologic stopping criteria, including 1) rebound (defined as the first day of 2 consecutive increases of at least 0.5 log10 IU/mL above nadir (local minimum value), or first day of 2 consecutive HCV RNA >= LLOQ for participants who previously achieved HCV RNA < LLOQ) during treatment, 2) participant who fails to suppress (defined as never achieving HCV RNA < LLOQ during treatment). (NCT01458535)
Timeframe: Day 1 through Week 12

,,,,,
Interventionpercentage of participants (Number)
Participant ReboundsParticipants who fail to Suppress
ABT-450/r and ABT-267 in Genotype 1 Participants10.00
ABT-450/r and ABT-267 in Genotype 2 Participants10.00
ABT-450/r and ABT-267 in Genotype 3 Participants72.70
ABT-450/r and ABT-267 Plus RBV in Genotype 1 Participants00
ABT-450/r and ABT-267 Plus RBV in Genotype 2 Participants10.00
ABT-450/r and ABT-267 Plus RBV in Genotype 3 Participants30.00

[back to top]

Percentage of Participants With Sustained Virologic Response 24 Weeks (SVR24) Post-Treatment

Sustained Virologic Response 24 (SVR24) is defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) less than the lower limit of quantification (LLOQ; < 25 IU/mL) 24 weeks after the last dose of study drug. Participants with missing data were imputed as failures. (NCT01458535)
Timeframe: Post-treatment Day 1 to Post-treatment Week 24

Interventionpercentage of participants (Number)
ABT-450/r and ABT-267 Plus RBV in Genotype 1 Participants100
ABT-450/r and ABT-267 Plus RBV in Genotype 2 Participants80.0
ABT-450/r and ABT-267 Plus RBV in Genotype 3 Participants40.0
ABT-450/r and ABT-267 in Genotype 1 Participants60.0
ABT-450/r and ABT-267 in Genotype 2 Participants60.0
ABT-450/r and ABT-267 in Genotype 3 Participants9.1

[back to top]

Percentage of Participants With Sustained Virologic Response 24 Weeks Post-dose for 8 Weeks Versus 12 Weeks of Treatment With 3 DAAs and Ribavirin

"The percentage of participants achieving sustained virologic response 24 weeks after the last dose of study drug (SVR24), defined as hepatitis C virus (HCV) ribonucleic acid (RNA) less than the lower limit of quantitation (LLOQ), without any confirmed quantifiable (≥ LLOQ) post-treatment value before that time point. HCV RNA levels were measured from plasma by a central laboratory. The LLOQ for the assay was 25 IU/mL.~The primary efficacy endpoint was the comparison between treatment-naïve participants following 8 weeks of treatment with 3 DAAs and ribavirin and those with 12 weeks of treatment with 3 DAAs and ribavirin (Group A versus Group G)." (NCT01464827)
Timeframe: Post Treatment Week 24

Interventionpercentage of participants (Number)
Group A87.5
Group B82.9
Group C84.6
Group D92.5
Group E88.6
Group F97.4
Group G95.0
Group H92.5
Group I90.0
Group J88.9
Group K91.3
Group L95.5
Group M91.3
Group N100.0

[back to top]

Percentage of Participants With Sustained Virologic Response 24 Weeks Post-dose in Treatment-naïve Versus Null-responders

This outcome measure compares the percentage of participants achieving sustained virologic response 24 weeks post-dose (HCV RNA < LLOQ at post-treatment Week 24) following treatment with 3 DAAs and ribavirin in participants who were treatment-naïve versus those who were null-responders to previous HCV therapy (Groups F + G + H + I versus Groups K + L + M + N). (NCT01464827)
Timeframe: Post-Treatment Week 24

Interventionpercentage of participants (Number)
Groups F + G + H + I93.7
Groups K + L + M + N94.3

[back to top]

Number of Participants With Adverse Events (AEs)

"An adverse event was defined as any untoward medical occurrence in a patient or clinical investigation participant administered a pharmaceutical product and that did not necessarily have a causal relationship with this treatment.~The investigator assessed the relationship of each AE to the use of direct-acting antiviral agents (DAAs) and to ribavirin, and rated the severity of each event as either:~Mild: The AE was transient and easily tolerated by the participant; Moderate: The AE caused the participant discomfort and interrupted usual activities; Severe: The AE caused considerable interference with the participant's usual activities and could have been incapacitating or life-threatening.~A serious adverse event was any event that resulted in death, was life-threatening, resulted in or prolonged hospitalization, resulted in a congenital anomaly or persistent or significant disability or was any other important medical event requiring medical or surgical intervention." (NCT01464827)
Timeframe: From the time of study drug administration until 30 days following discontinuation of study drug administration (up to 28 weeks).

,,,,,,,,
Interventionparticipants (Number)
Any adverse eventAny adverse event at least possibly DAA-relatedAny severe adverse eventAny serious adverse eventAny AE leading to discontinuation of study drugAny AE leading to interruption of study drugAny AE leading to ribavirin dose modificationAny fatal adverse events
Group A6758301020
Group B3629000120
Group C + D7153320240
Group E6851520100
Group F + G7157313090
Group H + I77683131100
Group J4235101030
Group K + L3930100010
Group M + N3728121030

[back to top]

Percentage of Participants With Sustained Virologic Response 24 Weeks Post-dose Following Treatment of Different Durations With 3 Direct-acting Antiviral Agents (DAAs) and Ribavirin

This outcome measure compares the percentage of participants achieving sustained virologic response 24 weeks after the last dose of study drug (HCV RNA < LLOQ at post-treatment Week 24) following treatment with 3 DAAs (ABT-450/ritonavir, ABT-267, and ABT-333) and ribavirin in both treatment naïve and null-responder participants for 8 weeks (Group A) versus 12 weeks (Groups F + G + K + L) versus 24 weeks (Groups H + I + M + N). (NCT01464827)
Timeframe: Post-Treatment Week 24

Interventionpercentage of participants (Number)
Group A87.5
Groups F + G + K + L95.2
Groups H + I + M + N92.7

[back to top]

Percentage of Participants With Sustained Virologic Response 24 Weeks Post-dose Following Treatment for 12 Weeks With 3 DAAs With Versus Without Ribavirin

This outcome measure compares the percentage of participants achieving sustained virologic response 24 weeks post-dose (HCV RNA < LLOQ at post-treatment Week 24) following treatment with 3 DAAs with or without ribavirin (Group E versus Groups F + G + K + L). (NCT01464827)
Timeframe: Post-Treatment Week 24

Interventionpercentage of participants (Number)
Group E88.6
Groups F + G + K + L95.2

[back to top]

Percentage of Participants With Sustained Virologic Response 24 Weeks Post-dose Following Treatment for 12 Weeks With 2 DAAs and Ribavirin Versus 3 DAAs and Ribavirin

This outcome measure compares the percentage of participants achieving sustained virologic response 24 weeks post-dose (HCV RNA < LLOQ at post-treatment Week 24) following treatment with 2 DAAs (ABT-450/ritonavir plus ABT-333 [Group B] or ABT-450/ritonavir plus ABT-267 [Groups C + D + J]) and ribavirin versus 3 DAAs (ABT-450/ritonavir plus ABT-333 and ABT-267) and ribavirin (Groups F + G + K + L). (NCT01464827)
Timeframe: Post-Treatment Week 24

Interventionpercentage of participants (Number)
Group B82.9
Groups C + D + J88.7
Groups F + G + K + L95.2

[back to top]

Proportion of Patients With Treatment Failure (TF)

Proportion of patients with treatment failure defined as having one of the following events: confirmed viral rebound (CVR) or treatment discontinuation for any cause. CVR was established when 2 consecutive viral load values (HIV-1 RNA)>50 copies/mL occurred within 2 weeks during follow-up. In case of CVR, patients treated with atazanavir/ritonavir monotherapy had to re-introduce their previous 2NRTIs (re-intensification) and, if not suppressed (HIV-1 RNA <50 copies /ml) after 12 weeks, discontinued from the study. Re-intensification was considered as treatment failure in the primary analysis conducted according to the intention-to-treat principle (intention-to-treat analysis with re-intensification equal failure, ITT=Failure) while it was not in the secondary analysis (intention-to-treat analysis with re-intensification equal success, ITT=Success). (NCT01511809)
Timeframe: Up to week 48

,
Interventionpercentage of patients (Number)
ITT=Failure analysisITT=Success analysis
Atazanavir/Ritonavir Monotherapy27.57.9
Atazanavir/Ritonavir Triple Therapy15.415.4

[back to top]

Mean Bone Mineral Density Changes From Baseline to 48 Weeks as Measured by DXA Scan

(NCT01513122)
Timeframe: 48 weeks

Interventionpercentage change (Mean)
Arm 1. Lopinavir / Ritonavir + 2-3N(t)RTI-5.2
Arm 2. Lopinavir /Ritonavir + Raltegravir-2.9

[back to top]

Mean Glucose Changes From Baseline to 48 Weeks

(NCT01513122)
Timeframe: 48 weeks

Interventionmmol/L (Mean)
Arm 1. Lopinavir / Ritonavir + 2-3N(t)RTI-0.04
Arm 2. Lopinavir /Ritonavir + Raltegravir-0.1

[back to top]

Mean Limbs Fat Changes From Baseline to 48 Weeks as Measured by DXA Scan

(NCT01513122)
Timeframe: 48 weeks

Interventionpercentage change (Mean)
Arm 1. Lopinavir / Ritonavir + 2-3N(t)RTI15.7
Arm 2. Lopinavir /Ritonavir + Raltegravir21.1

[back to top]

Mean Total Body Fat Changes From Baseline to 48 Weeks as Measured by DXA Scan

(NCT01513122)
Timeframe: 48 weeks

Interventionkg (Mean)
Arm 1. Lopinavir / Ritonavir + 2-3N(t)RTI1.4
Arm 2. Lopinavir /Ritonavir + Raltegravir2.1

[back to top]

Mean Total Cholesterol Changes From Baseline to 48 Weeks

(NCT01513122)
Timeframe: 48 weeks

Interventionmmol/L (Mean)
Arm 1. Lopinavir / Ritonavir + 2-3N(t)RTI0.4
Arm 2. Lopinavir /Ritonavir + Raltegravir0.6

[back to top]

Mean Triglycerides Changes From Baseline to 48 Weeks

(NCT01513122)
Timeframe: 48 weeks

Interventionmmol/L (Mean)
Arm 1. Lopinavir / Ritonavir + 2-3N(t)RTI0.6
Arm 2. Lopinavir /Ritonavir + Raltegravir0.8

[back to top]

Worst Sheehan Disability Scale (SDS) Score for the Safety Population

The Sheehan Disability Scale (SDS) assesses functional impairment in 3 inter-related domains: work/school, social and family life, using a rating scale for each item ranging from 0 (not at all) to 10 (extremely). (NCT01516970)
Timeframe: Month 3

,
Interventionunits on a scale (Mean)
Impairment in work/school/studiesImpairment in social lifeImpairment in family life
Darunavir/Ritonavir Postexposure Prophylaxis (DRV/r PEP)2.5662.4652.226
Standard of Care Postexposure Prophylaxis (SOCPEP)3.5033.4642.954

[back to top]

Percentage of Participants Who Developed Detectable HIV Antibodies

Seroconversion rate of HIV antibodies while receiving HIV PEP evaluated as the percentage of participants who developed detectable HIV antibodies (defined as positive) and percentage of participants who had not developed detectable HIV antibodies (defined as negative). Per protocol (PP) population included all participants in mITT (defined as all participants who were assigned to receive randomized HIV PEP and were not discontinued due to confirmation of the negative HIV infection status of the index person) excluding participants with: No indication for HIV PEP; Initiation of PEP >72 hours after injury; Discontinuation of HIV PEP due to confirmation of HIV negative status of index person and if index person bears resistant virus against HIV PEP components prescribed; incorrect HIV PEP; no intake of medication. (NCT01516970)
Timeframe: At Month 3

,
Interventionpercentage of participants (Number)
NegativePositive
Darunavir/Ritonavir Postexposure Prophylaxis (DRV/r PEP)99.30.7
Standard of Care Postexposure Prophylaxis (SOCPEP)1000

[back to top]

Number of Participants With Early Discontinuation From Randomized Human Immunodeficiency Virus Postexposure Prophylaxis (HIV PEP)

Number of participants with early discontinuation from randomized HIV PEP for any reason other than confirmation of the negative HIV infection status of the index person in participants receiving HIV PEP for at least 28 days and a maximum of 30 days was assessed. Per protocol (PP) population included all participants in modified intention-to-treat (mITT [defined as all participants who were assigned to receive randomized HIV PEP and were not discontinued due to confirmation of the negative HIV infection status of the index person]) excluding participants with: No indication for HIV PEP; Initiation of PEP >72 hours after injury; Discontinuation of HIV PEP due to confirmation of HIV negative status of index person and if index person bears resistant virus against HIV PEP components prescribed; incorrect HIV PEP; no intake of medication. (NCT01516970)
Timeframe: Up to 30 days

Interventionparticipants (Number)
Darunavir/Ritonavir Postexposure Prophylaxis (DRV/r PEP)10
Standard of Care Postexposure Prophylaxis (SOCPEP)15

[back to top]

Number of Participants With Treatment-Emergent Adverse Events (TEAEs)

An adverse event (AE) is defined to be non-treatment-emergent if the onset date of the AE was clearly before the date of first HIV PEP administration, otherwise it is considered treatment-emergent. (NCT01516970)
Timeframe: Up to Month 3

Interventionparticipants (Number)
Darunavir/Ritonavir Postexposure Prophylaxis (DRV/r PEP)131
Standard of Care Postexposure Prophylaxis (SOCPEP)125

[back to top]

Mean Maximal Decrease From Baseline in Hepatitis C Virus (HCV) Ribonucleic Acid (RNA) During ABT-267 Monotherapy

The baseline value was the last measurement before the first dose of ABT-267 monotherapy (Day 1). The maximal decrease during monotherapy was the change from baseline to the lowest log10 IU/mL HCV RNA level any time from the first dose of ABT-267 on Day 1 to the last log10 HCV RNA level before the first dose of ABT-267 combination therapy (Study Day 3). (NCT01563536)
Timeframe: Pre-dose on Days 1, 2, and 3

Interventionlog10 IU/mL (Least Squares Mean)
ABT-267 1.5 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV-1.6
ABT-267 25 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV-3.1

[back to top]

Percentage of Participants With End-of-Treatment Response

The percentage of participants with virologic response (plasma HCV RNA less than the lower limit of quantitation [< LLOQ]) at the end of combination therapy (12 weeks). (NCT01563536)
Timeframe: 12 weeks

Interventionpercentage of participants (Number)
ABT-267 1.5 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV83.3
ABT-267 25 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV83.3

[back to top]

Percentage of Participants With Rapid Virologic Response

The percentage of participants with virologic response (plasma HCV RNA less than the lower limit of quantitation [< LLOQ]) after 4 weeks of combination therapy. (NCT01563536)
Timeframe: 4 weeks

Interventionpercentage of participants (Number)
ABT-267 1.5 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV83.3
ABT-267 25 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV100

[back to top]

Time of Maximum Plasma Concentration (Tmax) of ABT-267 Following Monotherapy on Day 1

Blood samples were collected pre-dose (time 0) and at 2, 4, and 6 hours post-dose on Day 1 and pre-dose on Day 2 (ABT-267 monotherapy). The samples were analyzed for ABT-267 using validated analytical methods. Time of maximum plasma concentration (Tmax; measured in hours) was estimated using noncompartmental analyses. (NCT01563536)
Timeframe: Day 1 (pre-dose [time 0] and at 2, 4, and 6 hours post-dose) and Day 2 (pre-dose)

Interventionhours (Mean)
ABT-267 1.5 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV3.67
ABT-267 25 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV3.67

[back to top]

Mean Change in Viral Load From Baseline to Pre-dose on Day 2 and Day 3 of ABT-267 Monotherapy

The relationship between ABT-267 dose, ABT-267 concentration, and response was analyzed as the change in viral load (measured as log10 IU/mL) from baseline (pre-dose on Day 1) to pre-dose on Days 2 and 3. Plasma concentrations of ABT-267 pre-dose on Days 2 and 3 are presented above in 4. Primary Outcome: Plasma Concentration of ABT-267 Pre-dose (Ctrough) on Day 2 and Day 3. (NCT01563536)
Timeframe: Predose on Days 1, 2, and 3

,
Interventionlog10 IU/mL (Mean)
Day 2 Mean change in Viral Load from BaselineDay 3 Mean change in Viral Load from Baseline
ABT-267 1.5 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV-1.95-1.96
ABT-267 25 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV-2.85-3.10

[back to top]

Percentage of Participants With Extended Rapid Virologic Response

The percentage of participants with virologic response (plasma HCV RNA less than the lower limit of quantitation [< LLOQ]) at Weeks 4 through 12 of combination therapy. (NCT01563536)
Timeframe: Weeks 4 to 12

Interventionpercentage of participants (Number)
ABT-267 1.5 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV83.3
ABT-267 25 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV83.3

[back to top]

Resistance-Associated Variants and Phenotypic Resistance

Baseline (pre-dose on Day 1) samples were analyzed for resistance-associated amino acid (AA) variants using population sequencing. Phenotypic resistance to ABT-267 at Baseline was assessed by calculating the fold difference in the the half maximal effective concentration (EC50) compared with the EC50 for the appropriate reference replicon (1a-H77 or 1b-Con1). Day 3 samples were analyzed using population sequencing and were compared with the baseline and appropriate prototypic reference sequences to assess AA changes. Phenotypic resistance at Day 3 was assessed by calculating the fold difference in the EC50 compared with the EC50 for the corresponding Baseline sample. The number of participants with variants at resistance-associated AA positions and phenotypic resistance at Baseline and Day 3 are presented. (NCT01563536)
Timeframe: Day 1 Pre-dose (Baseline) and Day 3 Pre-dose

,
Interventionparticipants (Number)
Baseline Variants in NS5A (n=5, 6)Baseline Resistance >10-fold (n=5, 6)Day 3 Variants in NS5A (n=5, 2)Day 3 Resistance >10-fold (n=5, 2)
ABT-267 1.5 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV2161
ABT-267 25 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV0022

[back to top]

Number of Participants With Adverse Events (AEs)

"An adverse event was defined as any untoward medical occurrence in a patient or clinical investigation participant administered a pharmaceutical product and that did not necessarily have a causal relationship with this treatment.~The investigator assessed the relationship of each AE to the use of direct-acting antiviral agents (DAAs), and rated the severity of each event as either: Mild: The AE was transient and easily tolerated by the participant; Moderate: The AE caused the participant discomfort and interrupted usual activities; Severe: The AE caused considerable interference with the participant's usual activities and could have been incapacitating or life-threatening.~A serious adverse event was any event that resulted in death, was life-threatening, resulted in hospitalization or prolongation of hospitalization, resulted in a congenital anomaly or persistent or significant disability or was any other important medical event requiring medical or surgical intervention." (NCT01563536)
Timeframe: All AEs were collected from the time of study drug administration to 30 days after last dose of study drug (16 weeks).

,
Interventionparticipants (Number)
Any AEAny AE at least possibly related to DAAsAny severe AEAny serious AEAny AE leading to discontinuation of study drugAny AE leading to interruption of study drugAny AE leading to RBV dose modificationAny fatal AEDeaths
ABT-267 1.5 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV330000000
ABT-267 25 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV541210100

[back to top]

Percentage of Participants With Sustained Virologic Response 12 Weeks and 24 Weeks After Combination Therapy

The percentage of participants with sustained virologic response (plasma Hepatitis C virus ribonucleic acid [HCV RNA] level less than the lower limit of quantitation [< LLOQ]) 12 and 24 weeks after the last dose of combination study drug. The LLOQ for the assay was 25 IU/mL. (NCT01563536)
Timeframe: 12 and 24 weeks after last dose of combination study drug

,
Interventionpercentage of participants (Number)
12 Weeks Post-treatment24 Weeks Post-treatment
ABT-267 1.5 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV83.383.3
ABT-267 25 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV83.383.3

[back to top]

Plasma Concentration of ABT-267 Pre-dose (Ctrough) on Day 2 and Day 3

Blood samples were collected pre-dose on Day 2 (prior to second dose of ABT-267 monotherapy) and pre-dose on Day 3 (prior to first dose of combination therapy). The samples were analyzed for ABT-267 using validated analytical methods. Pre-dose plasma concentrations on Day 2 and Day 3 (Ctrough, measured in ng/mL) are reported. (NCT01563536)
Timeframe: Day 2 (pre-dose) and Day 3 (pre-dose)

,
Interventionng/mL (Mean)
Day 2 CtroughDay 3 Ctrough
ABT-267 1.5 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV00.228
ABT-267 25 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV5.26.62

[back to top]

Maximum Plasma Concentration (Cmax) of ABT-267 Following Monotherapy on Day 1

Blood samples were collected pre-dose (time 0) and at 2, 4, and 6 hours post-dose on Day 1 and pre-dose on Day 2 (ABT-267 monotherapy). The samples were analyzed for ABT-267 using validated analytical methods. Maximum plasma concentration (Cmax; measured in ng/mL) was estimated using noncompartmental analyses. (NCT01563536)
Timeframe: Day 1 (pre-dose [time 0] and at 2, 4, and 6 hours post-dose) and Day 2 (pre-dose)

Interventionng/mL (Mean)
ABT-267 1.5 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV1.66
ABT-267 25 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV41.0

[back to top]

Area Under the Plasma Concentration-time Curve From Time 0 to 24 Hours (AUC[24]) of ABT-267 Following Monotherapy on Day 1

Blood samples were collected pre-dose (time 0) and at 2, 4, and 6 hours post-dose on Day 1 and pre-dose on Day 2 (ABT-267 monotherapy). The samples were analyzed for ABT-267 using validated analytical methods. Area under the plasma concentration-time curve from time 0 to 24 hours (AUC[24]; measured in ng multiplied by hour/mL) was estimated using noncompartmental analyses. (NCT01563536)
Timeframe: Day 1 (pre-dose [time 0] and at 2, 4, and 6 hours post-dose) and Day 2 (pre-dose)

Interventionng*hr/mL (Mean)
ABT-267 1.5 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV18.0
ABT-267 25 mg, Then ABT-267, ABT-450/r, ABT-333, Plus RBV467

[back to top]

Part A: Plasma Pharmacokinetics of SOF, EFV, TFV, and FTC: Cmax at Day 7

"Cmax: maximum observed concentration of drug in plasma.~Data for this outcome measure were collected for participants in Part A only." (NCT01565889)
Timeframe: Pre-dose and 0.5, 1, 1.5, 2, 3, 4, 6, 8, 10, 12 hours postdose

,,,,
Interventionng/mL (Mean)
Pharmacokinetics of SOF (Cmax at Day 7)Pharmacokinetics of EFV (Cmax at Day 7)Pharmacokinetics of TFV (Cmax at Day 7)Pharmacokinetics of FTC (Cmax at Day 7)
Part A: SOF+EFV/FTC/TDF (Cohort 1)635.05423.8372.51533.1
Part A: SOF+EFV+ZDV/3TC (Cohort 2)285.13469.5NANA
Part A: SOF+RAL+FTC/TDF (Cohort 5)1189.2NA388.12079.5
Part A: SOF+RTV+ATV+FTC/TDF (Cohort 3)1228.9NA519.81797.9
Part A: SOF+RTV+DRV+FTC/TDF (Cohort 4)828.2NA441.81808.2

[back to top]

Incidence of Adverse Events Leading to Permanent Discontinuation of Study Drug(s)

The percentage of participants discontinuing any study drug due to an adverse event was summarized. (NCT01565889)
Timeframe: Up to 12 weeks

Interventionpercentage of participants (Number)
Part A: SOF+EFV/FTC/TDF (Cohort 1)0
Part A: SOF+EFV+ZDV/3TC (Cohort 2)0
Part A: SOF+RTV+ATV+FTC/TDF (Cohort 3)0
Part A: SOF+RTV+DRV+FTC/TDF (Cohort 4)0
Part A: SOF+RAL+FTC/TDF (Cohort 5)0
Part B: SOF+PEG+RBV8.7

[back to top]

Part B: Percentage of Participants With Sustained Virologic Response (SVR) at 12 Weeks After Discontinuation of Therapy (SVR12)

"SVR12 was defined as HCV RNA < the lower limit of quantitation (LLOQ; ie, 25 IU/mL) at 12 weeks after stopping study treatment.~Data for this outcome measure were collected for participants in Part B only." (NCT01565889)
Timeframe: Posttreatment Week 12

Interventionpercentage of participants (Number)
Part B: SOF+PEG+RBV91.3

[back to top]

Part A: Plasma Pharmacokinetics of SOF, EFV, Tenofovir (TFV), and FTC: AUCtau at Day 7

"AUCtau: concentration of drug over time (area under the plasma concentration versus time curve over the dosing interval).~Data for this outcome measure were collected for participants in Part A only." (NCT01565889)
Timeframe: Pre-dose and 0.5, 1, 1.5, 2, 3, 4, 6, 8, 10, 12 hours postdose

,,,,
Interventionh*ng/mL (Mean)
Pharmacokinetics of SOF (AUCtau at Day 7)Pharmacokinetics of EFV (AUCtau at Day 7)Pharmacokinetics of TFV (AUCtau at Day 7)Pharmacokinetics of FTC (AUCtau at Day 7)
Part A: SOF+EFV/FTC/TDF (Cohort 1)867.595094.42351.210144.8
Part A: SOF+EFV+ZDV/3TC (Cohort 2)627.653770.7NANA
Part A: SOF+RAL+FTC/TDF (Cohort 5)1687.1NA2657.110622.8
Part A: SOF+RTV+ATV+FTC/TDF (Cohort 3)2269.4NA3793.011564.9
Part A: SOF+RTV+DRV+FTC/TDF (Cohort 4)1421.5NA3996.813091.2

[back to top]

Part B: On-treatment HCV RNA

Data for this outcome measure were collected for participants in Part B only. (NCT01565889)
Timeframe: Up to 8 weeks

Interventionlog10 IU/mL (Mean)
Baseline (n = 23)Week 1 (n = 20)Week 2 (n = 22)Week 4 (n = 23)Week 6 (n = 23)Week 8 (n = 21)
Part B: SOF+PEG+RBV6.591.651.381.381.381.38

[back to top]

Part B: On-treatment HIV RNA

Data for this outcome measure were collected for participants in Part B only. (NCT01565889)
Timeframe: Up to 8 weeks

Interventioncopies/mL (Mean)
Baseline (n = 23)Week 1 (n = 20)Week 2 (n = 22)Week 4 (n = 22)Week 6 (n = 22)Week 8 (n = 21)
Part B: SOF+PEG+RBV252020191919

[back to top]

Part B: Percentage of Participants Experiencing Viral Breakthrough or Viral Relapse

"Viral breakthrough was defined as having confirmed detectable HCV RNA levels (HCV RNA > LLOQ) on treatment after having previously had undetectable HCV RNA levels (HCV RNA < LLOQ) while on treatment.~Viral relapse was defined as having achieved undetectable HCV RNA levels (HCV RNA < LLOQ) at end of treatment, but did not achieve an SVR.~Data for this outcome measure were collected for participants in Part B only." (NCT01565889)
Timeframe: Posttreatment Weeks 4 and 24

Interventionpercentage of participants (Number)
Viral breakthroughViral relapse
Part B: SOF+PEG+RBV08.7

[back to top]

Part B: Percentage of Participants With Sustained Virologic Response at 4 and 24 Weeks After Discontinuation of Therapy (SVR4 and SVR24)

"SVR4 and SVR24 was defined as HCV RNA < LLOQ at 4 and 24 weeks following the last dose of study drug, respectively.~Data for this outcome measure were collected for participants in Part B only." (NCT01565889)
Timeframe: Posttreatment Weeks 4 and 24

Interventionpercentage of participants (Number)
SVR4SVR24
Part B: SOF+PEG+RBV91.391.3

[back to top]

Percent of Participants Whose HIV Viral Load Was Less Than 400 Copies/mL at Week 48.

The percent of participants whose HIV viral load was less than 400 copies/mL at week 48 was calculated with an associated standard error. The confidence interval was calculated using Wilson's score method. Participants who were lost-to-follow-up or dead by week 48 or had missing results at week 48 were coded as having HIV viral load greater than 400 copies/mL. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: 48 weeks

Interventionpercentage of participants (Number)
A: Standard-dose LPV/r w/RBT58.3
B: Double-dose LPV/r w/RIF66.7
C: Standard-Dose LPV/r + RAL w/RBT60.9

[back to top]

CD4 Count Change From Baseline to Week 24

The difference in CD4 count from baseline to week 24 was calculated as the CD4 count at week 24 minus the CD4 count at baseline. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: Baseline and 24 weeks

Interventioncells/mm^3 (Median)
A: Standard-dose LPV/r w/RBT20
B: Double-dose LPV/r w/RIF56
C: Standard-Dose LPV/r + RAL w/RBT13

[back to top]

RAL Cmax and Cmin in Participants Enrolled in Arm C

Describe RAL plasma PK characteristics (Cmax and Cmin) in participants enrolled in Arm C, determined by non-compartmental analysis of 24-hour PK sampling. The pre-dose concentration was determined using a sample drawn 12 hours after the previous RAL dose and was used as the 12-hour RAL concentration. (NCT01601626)
Timeframe: At 2 weeks: pre-dose and at 2, 4, 5, 6, and 24 hours post-dose

Interventionng/mL (Median)
Maximum Concentration (Cmax)Minimum Concentration (Cmin)
C: Standard-Dose LPV/r + RAL w/RBT2830166

[back to top]

LPV Cmax and Cmin in Participants Enrolled in Arms A, B, and C

Describe LPV plasma pharmacokinetic (PK) characteristics (maximum concentration [Cmax] and minimum concentration [Cmin]) in participants enrolled in Arms A, B, and C, determined by non-compartmental analysis of 12-hour PK sampling. The pre-dose concentration was determined using a sample drawn 12 hours after the previous LPV dose and was used as the 12-hour LPV concentration. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: At 2 weeks: pre-dose and at 2, 4, 5, and 6 hours post-dose

,,
Interventionng/mL (Median)
Maximum Concentration (Cmax)Minimum Concentration (Cmin)
A: Standard-dose LPV/r w/RBT185319920
B: Double-dose LPV/r w/RIF181388033
C: Standard-Dose LPV/r + RAL w/RBT168028548

[back to top]

RBT AUC in Participants Enrolled in Arms A and C

Describe RBT plasma PK characteristics (area under the curve [AUC] between 0 and 24 hours) in participants enrolled in Arms A and C, determined by non-compartmental analysis of 24-hour PK sampling. The pre-dose concentration was determined using a sample drawn 24 hours after the previous RBT dose. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: At 2 weeks: pre-dose and at 2, 4, 5, 6, and 24 hours post-dose

Interventionhours*ng/mL (Median)
A: Standard-dose LPV/r w/RBT7374
C: Standard-Dose LPV/r + RAL w/RBT5516

[back to top]

RAL AUC in Participants Enrolled in Arm C

Describe RAL plasma PK characteristics (area under the curve [AUC] between 0 and 24 hours) in participants enrolled in Arm C, determined by non-compartmental analysis of 24-hour PK sampling. The pre-dose concentration was determined using a sample drawn 12 hours after the previous RAL dose and was used as the 12-hour RAL concentration. (NCT01601626)
Timeframe: At 2 weeks: pre-dose and at 2, 4, 5, 6, and 24 hours post-dose

Interventionhours*ng/mL (Median)
C: Standard-Dose LPV/r + RAL w/RBT11338

[back to top]

Percent of Participants Who Experienced a New AIDS-defining Illness or Died

New post-randomization diagnoses were considered AIDS-defining based on the CDC classification system. The percent of participants who experienced a new AIDS-defining illness or died was calculated with an associated standard error. Confidence intervals were calculated using Wilson's score method. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: After randomization and through week 72

Interventionpercentage of participants (Number)
A: Standard-dose LPV/r w/RBT4.2
B: Double-dose LPV/r w/RIF8.3
C: Standard-Dose LPV/r + RAL w/RBT4.3

[back to top]

RBT Cmax and Cmin in Participants Enrolled in Arms A and C

Describe RBT plasma PK characteristics (Cmax and Cmin) in participants enrolled in Arms A and C, determined by non-compartmental analysis of 24-hour PK sampling. The pre-dose concentration was determined using a sample drawn 24 hours after the previous RBT dose. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: At 2 weeks: pre-dose and at 2, 4, 5, 6, and 24 hours post-dose

,
Interventionng/mL (Median)
Maximum Concentration (Cmax)Minimum Concentration (Cmin)
A: Standard-dose LPV/r w/RBT461161
C: Standard-Dose LPV/r + RAL w/RBT349115

[back to top]

Percent of Participants Who Experienced Sputum Conversion at Week 8.

Sputum conversion was defined as culture MTB-negative at week 8 or AFB smear negative at week 8 (and culture contaminated or missing at week 8); there were no Xpert MTB/RIF results at week 8. The percent of participants experienced sputum conversion at week 8 was calculated with an associated standard error. The confidence interval was calculated using Wilson's score method. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: 8 weeks

Interventionpercentage of participants (Number)
A: Standard-dose LPV/r w/RBT87.5
B: Double-dose LPV/r w/RIF81.8
C: Standard-Dose LPV/r + RAL w/RBT70.0

[back to top]

Percent of Participants Who Experienced HIV Virologic Failure

Virologic failure was defined as the occurrence of two consecutive plasma HIV-1 RNA levels ≥1000 copies/mL at or after 16 weeks and within 24 weeks of treatment initiation or ≥400 copies/mL at or after 24 weeks of treatment, regardless of whether randomized ART was being taken at the time of virologic failure. Participants who were missing data due to being lost-to-follow-up or dead were coded as virologic failures. The percent of participants who experienced HIV virologic failure was calculated with an associated standard error. The confidence interval was calculated using Wilson's score method. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: At weeks 16, 24, 48, and 72

Interventionpercentage of participants (Number)
A: Standard-dose LPV/r w/RBT29.2
B: Double-dose LPV/r w/RIF50.0
C: Standard-Dose LPV/r + RAL w/RBT30.4

[back to top]

Percent of Participants Whose HIV Viral Load Was Less Than 50 Copies/mL at Week 48

The percent of participants whose HIV viral load was less than 50 copies/mL at week 48 was calculated with an associated standard error. Participants who were lost-to-follow-up or dead by week 48 or had missing RNA at week 48 were coded as having HIV viral load greater than 50 copies/mL. The confidence interval was calculated using Wilson's score method. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: 48 weeks

Interventionpercentage of participants (Number)
A: Standard-dose LPV/r w/RBT45.8
B: Double-dose LPV/r w/RIF54.2
C: Standard-Dose LPV/r + RAL w/RBT56.5

[back to top]

Percent of Participants Who Experienced a New AIDS-defining Illness

New post-randomization diagnoses were considered AIDS-defining based on the CDC classification system. The percent of participants who experienced a new AIDS-defining illness was calculated with an associated standard error. The confidence interval was calculated using Wilson's score method. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: After randomization and through week 72

Interventionpercentage of participants (Number)
A: Standard-dose LPV/r w/RBT0.0
B: Double-dose LPV/r w/RIF4.2
C: Standard-Dose LPV/r + RAL w/RBT0.0

[back to top]

Percent of Participants Who Interrupted or Discontinued at Least One TB Drug Due to Toxicity

The percent of participants who interrupted or discontinued at least one TB drug due to toxicity was calculated with an associated standard error. The confidence interval was calculated using Wilson's score method. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: After randomization and through to the discontinuation of the last TB drug

Interventionpercentage of participants (Number)
A: Standard-dose LPV/r w/RBT20.8
B: Double-dose LPV/r w/RIF8.3
C: Standard-Dose LPV/r + RAL w/RBT13.0

[back to top]

Percent of Participants Who Experienced TB Treatment Failure

TB treatment failure was defined as having a MTB-positive culture after 16 weeks of TB treatment for a participant who was documented to be taking TB medications. The percent of participants who experienced TB treatment failure was calculated with an associated standard error. The confidence interval was calculated using Wilson's score method. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: After 16 weeks and through week 72

Interventionpercentage of participants (Number)
A: Standard-dose LPV/r w/RBT0.0
B: Double-dose LPV/r w/RIF0.0
C: Standard-Dose LPV/r + RAL w/RBT0.0

[back to top]

Percent of Participants Who Interrupted or Discontinued at Least One HIV Drug Due to Toxicity

The percent of participants who interrupted or discontinued at least one HIV drug due to toxicity was calculated with an associated standard error. The confidence interval was calculated using Wilson's score method. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: After randomization and through week 72

Interventionpercentage of participants (Number)
A: Standard-dose LPV/r w/RBT20.8
B: Double-dose LPV/r w/RIF16.7
C: Standard-Dose LPV/r + RAL w/RBT21.7

[back to top]

Percent of Participants Who Died

The percent of participants who died was calculated with an associated standard error. Confidence intervals were calculated using Wilson's score method. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: After randomization and through week 72

Interventionpercentage of participants (Number)
A: Standard-dose LPV/r w/RBT4.2
B: Double-dose LPV/r w/RIF4.7
C: Standard-Dose LPV/r + RAL w/RBT4.3

[back to top]

Percent of Participants Who Experienced TB Relapse/Recurrence

TB relapse/recurrence was defined as having had 2 consecutive MTB-negative cultures and subsequently had clinical or radiographic deterioration consistent with active TB at or after week 24 and before week 72. The percent of participants who experienced TB relapse/recurrence was calculated with an associated standard error. The confidence interval was calculated using Wilson's score method. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: At or after 24 weeks and through week 72

Interventionpercentage of participants (Number)
A: Standard-dose LPV/r w/RBT0.0
B: Double-dose LPV/r w/RIF4.2
C: Standard-Dose LPV/r + RAL w/RBT4.3

[back to top]

Number of Participants Reporting a Grade 3 or 4 Sign or Symptom

The number of participants reporting a grade 3 (severe) or grade 4 (life-threatening) sign or symptom were summarized. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: After randomization and through week 72

InterventionParticipants (Count of Participants)
A: Standard-dose LPV/r w/RBT7
B: Double-dose LPV/r w/RIF5
C: Standard-Dose LPV/r + RAL w/RBT5

[back to top]

Number of Participants Reporting a Grade 3 or 4 Laboratory Abnormality

The number of participants reporting a grade 3 (severe) or grade 4 (life-threatening) laboratory abnormality were summarized. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: After randomization and through week 72

InterventionParticipants (Count of Participants)
A: Standard-dose LPV/r w/RBT6
B: Double-dose LPV/r w/RIF3
C: Standard-Dose LPV/r + RAL w/RBT5

[back to top]

LPV AUC in Participants Enrolled in Arms A, B, and C

Describe LPV plasma PK characteristics (area under the curve [AUC] between 0 and 12 hours) in participants enrolled in Arms A, B, and C, determined by non-compartmental analysis of 12-hour PK sampling. The pre-dose concentration was determined using a sample drawn 12 hours after the previous LPV dose and was used as the 12-hour LPV concentration. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: At 2 weeks: pre-dose and at 2, 4, 5, and 6 hours post-dose

Interventionhours*ng/mL (Median)
A: Standard-dose LPV/r w/RBT159796
B: Double-dose LPV/r w/RIF161772
C: Standard-Dose LPV/r + RAL w/RBT149247

[back to top]

Cumulative Probability of HIV Virologic Failure at Week 72

Virologic failure was defined as the occurrence of two consecutive plasma HIV-1 RNA levels ≥1000 copies/mL at or after 16 weeks and within 24 weeks of treatment initiation or ≥400 copies/mL at or after 24 weeks of treatment, regardless of whether randomized ART was being taken at the time of virologic failure. The percent of participants with HIV virologic failure at week 72 was calculated using a Kaplan-Meier estimator with an associated standard error. The confidence interval was calculated using a log-log transformation. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: At weeks 16, 24, 48, and 72

Interventioncumulative events per 100 participants (Number)
A: Standard-dose LPV/r w/RBT29.2
B: Double-dose LPV/r w/RIF50.0
C: Standard-Dose LPV/r + RAL w/RBT30.4

[back to top]

CD4 Count Change From Baseline to Week 8

The difference in CD4 count from baseline to week 8 was calculated as the CD4 count at week 8 minus the CD4 count at baseline. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: Baseline and 8 weeks

Interventioncells/mm^3 (Median)
A: Standard-dose LPV/r w/RBT7
B: Double-dose LPV/r w/RIF26
C: Standard-Dose LPV/r + RAL w/RBT37

[back to top]

CD4 Count Change From Baseline to Week 72

The difference in CD4 count from baseline to week 72 was calculated as the CD4 count at week 72 minus the CD4 count at baseline. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: Baseline and 72 weeks

Interventioncells/mm^3 (Median)
A: Standard-dose LPV/r w/RBT126
B: Double-dose LPV/r w/RIF212
C: Standard-Dose LPV/r + RAL w/RBT54

[back to top]

CD4 Count Change From Baseline to Week 48

The difference in CD4 count from baseline to week 48 was calculated as the CD4 count at week 48 minus the CD4 count at baseline. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: Baseline and 48 weeks

Interventioncells/mm^3 (Median)
A: Standard-dose LPV/r w/RBT99
B: Double-dose LPV/r w/RIF119
C: Standard-Dose LPV/r + RAL w/RBT74

[back to top]

Percent of Participants Who Experienced TB Relapse/Recurrence and Who Had TB Drug Resistance

TB relapse/recurrence was defined as having had 2 consecutive MTB-negative cultures and subsequently had clinical or radiographic deterioration consistent with active TB at or after week 24 and before week 72. The drug resistance was determined based on phenotypic methods. The percent of participants who experienced TB relapse/recurrence and who had TB drug resistance was calculated with an associated standard error. The confidence interval was calculated using Wilson's score method. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: At or after 24 weeks and through week 72

Interventionparticipants (Number)
B: Double-dose LPV/r w/RIF0
C: Standard-Dose LPV/r + RAL w/RBT0

[back to top]

Number of Participants Who Experienced MTB IRIS

The number of participants who experienced MTB immune reconstitution inflammatory syndrome (IRIS) was summarized. As stated in the Detailed Study Description of the Protocol Section, formal statistical comparisons were not undertaken because of limited sample size. (NCT01601626)
Timeframe: After randomization and through week 72

InterventionParticipants (Count of Participants)
A: Standard-dose LPV/r w/RBT1
B: Double-dose LPV/r w/RIF2
C: Standard-Dose LPV/r + RAL w/RBT3

[back to top]

Percentage of Participants Achieving Virologic Response 24 Weeks Post Treatment (SVR24)

SVR24 was defined as HCV RNA level less than the LLOQ 24 weeks after the last dose of study drugs (DAAs plus pegIFN alpha-2a and RBV). (NCT01609933)
Timeframe: 24 weeks after last dose of study drugs (DAAs plus pegIFN alpha-2a and RBV); approximately 48 weeks after subject's initial dose of study drug in Substudy 1

Interventionpercentage of participants (Number)
2-DAA + PegIFN/RBV78.1

[back to top]

Percentage of Participants Achieving Sustained Virologic Response 12 Weeks Post Treatment (SVR12)

SVR12 was defined as hepatitis C virus ribonucleic acid (HCV RNA) level less than lower limit of quantitation [LLOQ] 12 weeks after the last dose of study drugs (DAAs plus pegIFN alpha-2a and RBV). (NCT01609933)
Timeframe: 12 weeks after last dose of study drugs (DAAs plus pegIFN alpha-2a and RBV); approximately 36 weeks after subject's initial dose of study drug in Substudy 1

Interventionpercentage of participants (Number)
2-DAA + PegIFN/RBV81.3

[back to top]

Number of Participants With Adverse Events

An adverse event (AE) is defined as any untoward medical occurrence in a patient or clinical investigation subject administered a pharmaceutical product and which does not necessarily have a causal relationship with this treatment. The investigator assessed the relationship of each event to the use of study drug as either probably related, possibly related, probably not related or not related. A serious adverse event (SAE) is an event that results in death, is life-threatening, requires or prolongs hospitalization, results in a congenital anomaly, persistent or significant disability/incapacity or is an important medical event that, based on medical judgment, may jeopardize the subject and may require medical or surgical intervention to prevent any of the outcomes listed above. Treatment-emergent events (TEAEs/TESAEs) are defined as any event that began or worsened in severity after initiation of study drug through 30 days post-DAA dosing. For more details on AEs, see the AE section. (NCT01609933)
Timeframe: From first dose of study drug through 30 days after last dose of study drug (DAAs plus pegIFN alpha-2a and RBV) (up to 28 weeks).

Interventionparticipants (Number)
2-DAA TEAE2-DAA TESAE
2-DAA + PegIFN/RBV291

[back to top]

Percentage of Participants With Extended Rapid Virologic Response (eRVR)

eRVR was defined as HCV RNA level < LLOQ at Substudy 1 treatment weeks 4 through 12 without a confirmed HCV RNA >= LLOQ (NCT01609933)
Timeframe: Treatment weeks 4 through 12 of Substudy 1 (DAAs plus pegIFN alpha-2a and RBV)

Interventionpercentage of participants (Number)
2-DAA + PegIFN/RBV87.5

[back to top]

Change in log10(Pf Gametocyte Density) From Entry to Day 30

"Change in log10(Pf gametocyte density) as evaluated using a Hodges-Lehmann estimate from entry to day 30 is evaluated in two groups:~Randomized to nNRTI-based ART with continued Pf SCP at day 15~Randomized to LPV/r-based ART with continued Pf SCP at day 15~Analysis was not conducted in either group with clearance at day 15 due to the small sample size and high number of undetectable samples in both clearance groups at entry and day 30." (NCT01632891)
Timeframe: Entry, Day 30

Interventionlog10(gametocyte/µL) (Number)
nNRTI-based ART, Not Cleared-0.46
LPV/R-based ART, Not Cleared0.17

[back to top]

Change in log10(Pf Parasite Density) From Entry to Day 30

"Change is evaluated as log10(Pf parasite density) at day 30 minus log10(Pf parasite density) at entry.~Change is evaluated in four groups:~Randomized to nNRTI-based ART with continued Pf SCP at day 15~Randomized to nNRTI-based ART with clearance of Pf SCP at day 15~Randomized to LPV/r-based ART with continued Pf SCP at day 15~Randomized to LPV/r-based ART with clearance of Pf SCP at day 15" (NCT01632891)
Timeframe: Entry, Day 30

Interventionlog10(parasites/µL) (Median)
nNRTI-based ART, Not Cleared-2.26
nNRTI-based ART, Cleared-1.65
LPV/R-based ART, Not Cleared-1.82
LPV/R-based ART, Cleared-3.61

[back to top]

Number of Participants With Uncomplicated Clinical Malaria

Uncomplicated clinical malaria is defined as the presence of non-severe fever/symptoms and parasitemia without organ complication. (NCT01632891)
Timeframe: From study entry to day 30

InterventionParticipants (Count of Participants)
LPV/R-based ART2
nNRTI-based ART1

[back to top]

Proportion of Participants With Plasmodium Falciparum (Pf) Subclinical Parasitemia (SCP) Clearance

"Pf SCP clearance defined by polymerase chain reaction (PCR) < 10 parasites/µL on three consecutive occasions within a 24-hour period.~If a participant had missing data on day 15, they were considered as not having clearance." (NCT01632891)
Timeframe: Day 15 (3 samples collected, separated by at least 5 hours and all three collected within 24-hours)

,
InterventionProportion of participants (Number)
Proportion ClearedProportion Not Cleared
LPV/R-based ART0.230.77
nNRTI-based ART0.270.73

[back to top]

Time to First Pf SCP Clearance

Time to clearance is defined by time to first measurement with PCR < 10 parasites/µL, and is evaluated as the point estimate and 95% CI for the day when 50% of participants cleared parasite. (NCT01632891)
Timeframe: From study entry up to day 30

InterventionDays (Median)
LPV/R-based ART12
nNRTI-based ART14

[back to top]

Log10(Pf Parasite Density)

Pf parasite density was determined by PCR. If parasite density equals 0, the value is set to 0.01 before log10 transformation. The value 0.01 was chosen based on the smallest observed parasite density value of 0.017. (NCT01632891)
Timeframe: Entry, days 3, 6, 9, 12, 15, 20, 25, 30

,
Interventionlog10(parasites/µL) (Mean)
EntryDay 3Day 6Day 9Day 12Day 15Day 20Day 25Day 30
LPV/R-based ART2.481.921.771.651.591.590.650.280.14
nNRTI-based ART2.091.571.491.631.561.430.670.490.30

[back to top]

Number of Participants With Detectable Pf Gametocyte Density

Number of participants with detectable Pf gametocyte density as determined by PCR. Due to the large number of undetectable results, this outcome was measured as dichotomous. (NCT01632891)
Timeframe: Entry, days 3, 6, 9, 12, 15, 20, 25, 30

,
InterventionParticipants (Count of Participants)
EntryDay 3Day 6Day 9Day 12Day 15Day 20Day 25Day 30
LPV/R-based ART1110911610111211
nNRTI-based ART121512131114141613

[back to top] [back to top] [back to top] [back to top]

Change From Baseline in Patient Perception of Stress

Change in perception of stress was measured by the Perceived Stress Scale (PSS), a 10-item questionnaire that assesses the degree to which the participant considered situations as stressful. The PSS score ranges from 0 to 40, with higher scores indicating higher levels of perceived stress. (NCT01662336)
Timeframe: Baseline, Month 6 and Month 12

Interventionunits on a scale (Mean)
Change from Baseline to Month 6Change from Baseline to Month 12
Lopinavir/Ritonavir + KASA-0.3-0.8

[back to top]

Change From Baseline in Psychological Well-being

Change in psychological well-being was measured by the Center for Epidemiologic Studies Depression scale (CES-D), a 20-item questionnaire assessing the presence of depressive state during the previous week. The possible range of scores is 0 to 60, with higher scores indicating the presence of more symptomatology. (NCT01662336)
Timeframe: Baseline, Month 6 and Month 12

Interventionunits on a scale (Mean)
Change from Baseline to Month 6Change from Baseline to Month 12
Lopinavir/Ritonavir + KASA1.71.0

[back to top]

Cluster of Differentiation 4 (CD4) Positive Cell Counts at Each Visit

(NCT01662336)
Timeframe: Baseline, Month 6 and Month 12

Interventioncells/mm³ (Mean)
BaselineMonth 6Month 12
Lopinavir/Ritonavir + KASA547.0620.8620.2

[back to top]

Health Resource Utilization

Health resource utilization (HRU) was measured by a self-administered questionnaire that contained a series of questions aimed at measuring the patient's utilization of healthcare resources and economic impact of the disease. (NCT01662336)
Timeframe: Baseline, Month 6 and Month 12

,,
Interventionpercentage of participants (Number)
Emergency RoomAdmitted to HospitalAdmitted to a Long-Term Care FacilityVisit to a Doctor's Office/ ClinicAmbulance ServicePhysiotherapist/ RehabilitationPsychiatrist/ Psychologist/ CounselorNursing ServicesSpecialist
Baseline8.13.50.055.52.93.59.212.716.2
Month 125.53.70.950.51.88.312.811.014.7
Month 68.52.80.750.71.47.712.014.812.7

[back to top]

Healthcare Provider Satisfaction

For each participant, healthcare provider (HCP) satisfaction with the KASA program was measured by three questions assessing 1) the overall satisfaction with the KASA program, 2) subjective assessment on whether the KASA program was beneficial in maintaining adherence with HIV treatments, and 3) the likelihood of recommending KASA in the future. The scores for each question ranged from 0 to 100, with higher scores indicating higher satisfaction. (NCT01662336)
Timeframe: Month 6 and Month 12

,
Interventionunits on a scale (Mean)
Overall Satisfaction with KASA ProgramBenefits in Maintaining Treatment AdherenceLikelihood to Recommend KASA Program in Future
Month 1273.362.770.0
Month 674.165.674.7

[back to top]

Viral Load at Each Visit

(NCT01662336)
Timeframe: Baseline, Month 6 and Month 12

Interventioncopies/mL (Mean)
BaselineMonth 6Month 12
Lopinavir/Ritonavir + KASA111.355.747.2

[back to top] [back to top]

Percentage of Participants Adherent to Treatment at Month 12

Adherence was assessed by the Adherence Self-Efficacy Scale (ASES). The ASES is a 12 item tool that measures the patient's confidence to undertake treatment related activities and behaviors including medication regimen, diet and exercise. Each question was answered on a scale from 0 (cannot do at all) to 10 (certain can do). A summative score ranging from 0 to 120 was calculated, with higher scores indicating higher treatment self-efficacy. A participant was considered to have maintained adherence if the change in the ASES summative score at month 12 was greater than or equal to zero. Participants who discontinued from the study or were lost to follow-up were considered non-adherent. (NCT01662336)
Timeframe: Baseline and 12 months

Interventionpercentage of participants (Number)
Lopinavir/Ritonavir + KASA30.2

[back to top]

Percentage of Participants Adherent to Treatment at Month 6

Adherence was assessed by the Adherence Self-Efficacy Scale (ASES). The ASES is a 12 item tool that measures the patient's confidence to undertake treatment related activities and behaviors including medication regimen, diet and exercise. Each question is answered on a scale from 0 (cannot do at all) to 10 (certain can do). A summative score ranging from 0 to 120 was calculated, with higher scores indicating higher treatment self-efficacy. A participant was considered to have maintained adherence if the change in the ASES summative score at month 6 relative to Baseline was greater than or equal to zero. Participants who discontinued from the study or were lost to follow-up were considered non-adherent. (NCT01662336)
Timeframe: Baseline and 6 months

Interventionpercentage of participants (Number)
Lopinavir/Ritonavir + KASA43.3

[back to top]

Change From Baseline in Adherence Integration Subscale Score at Months 6 and 12

"Adherence was assessed by the Adherence Self-Efficacy Scale (ASES). The ASES is a 12-item tool that measures the patient's confidence to undertake treatment-related activities and behaviors including medication regimen, diet and exercise. Each question was answered on a scale from 0 (cannot do at all) to 10 (certain can do).~The 12 items converge to two subscales measuring adherence integration and adherence perseverance. The adherence integration subscale score ranges from 0 to 90, with higher scores indicating higher treatment self-efficacy." (NCT01662336)
Timeframe: Baseline, Month 6 and Month 12

Interventionunits on a scale (Mean)
Change from Baseline to Month 6Change from Baseline to Month 12
Lopinavir/Ritonavir + KASA-0.7-3.6

[back to top]

Change From Baseline in Adherence Perseverance Subscale Score at Months 6 and 12

Adherence was assessed by the Adherence Self-Efficacy Scale (ASES). The ASES is a 12-item tool that measures the patient's confidence to undertake treatment-related activities and behaviors including medication regimen, diet and exercise. Each question was answered on a scale from 0 (cannot do at all) to 10 (certain can do). The 12 items converge to two subscales measuring adherence integration and adherence perseverance. The adherence perseverance subscale score ranges from 0 to 30, with higher scores indicating higher treatment self-efficacy. (NCT01662336)
Timeframe: Baseline, Month 6 and Month 12

Interventionunits on a scale (Mean)
Change from Baseline to Month 6Change from Baseline to Month 12
Lopinavir/Ritonavir + KASA-0.1-1.0

[back to top]

Change From Baseline in Adherence Summative Score at Months 6 and 12

Adherence was assessed by the Adherence Self-Efficacy Scale (ASES). The ASES is a 12-item tool that measures the participant's confidence to undertake treatment-related activities and behaviors including medication regimen, diet and exercise. Each question was answered on a scale from 0 (cannot do at all) to 10 (certain can do). A summative score ranging from 0 to 120 was calculated, with higher scores indicating higher treatment self-efficacy. (NCT01662336)
Timeframe: Baseline, Month 6 and Month 12

Interventionunits on a scale (Mean)
Change from Baseline to Month 6Change from Baseline to Month 12
Lopinavir/Ritonavir + KASA-1.2-5.1

[back to top]

Change From Baseline in Coping Self-Efficacy

Change in coping self-efficacy was measured by the Coping Self-Efficacy Scale (CSE), a 26-item questionnaire that measures perceived self-efficacy in coping with daily psychological challenges. A summative score ranging from 0 to 260 was calculated, with higher scores indicating higher coping self-efficacy. (NCT01662336)
Timeframe: Baseline, Month 6 and Month 12

Interventionunits on a scale (Mean)
Change from Baseline to Month 6Change from Baseline to Month 12
Lopinavir/Ritonavir + KASA2.48.1

[back to top] [back to top] [back to top] [back to top] [back to top]

AUC Norethindrone

0, 1, 2, 3, 4, 6, 8, 12, 24, 48, 72, 96 hours post-dose on Day 21 (NCT01667978)
Timeframe: following 21 days of continuous ingestion

Interventionng*h/mL (Mean)
Protease Inhibitor37.81
Control25.21

[back to top]

Percentage of Participants With End of Treatment (EOT) Response

The percentage of participants with EOT response (plasma HCV RNA level < LLOQ at week 12 for the 12-week duration arms and Week 24 for the 24-week duration arms12). The LLOQ for the assay was 25 IU/mL. (NCT01672983)
Timeframe: 12 or 24 weeks after first dose of study drug

Interventionpercentage of participants (Number)
Arm 1100
Arm 2100
Arm 3100
Arm 4100
Arm 563.2
Arm 683.3

[back to top]

Percentage of Participants With Sustained Virologic Response 12 Weeks After Treatment (SVR12)

The percentage of participants with SVR12 (plasma HCV RNA level < LLOQ 12 weeks after the last dose of study drug). The LLOQ for the assay was 25 IU/mL. (NCT01672983)
Timeframe: 12 weeks after last dose of study drug

Interventionpercentage of participants (Number)
Arm 1100
Arm 288.9
Arm 3100
Arm 4100
Arm 557.9
Arm 672.2

[back to top]

Percentage of Participants With Sustained Virologic Response 24 Weeks After Treatment (SVR24)

The percentage of participants with SVR24 (plasma Hepatitis C virus ribonucleic acid [HCV RNA] level less than the lower limit of quantitation [< LLOQ] 24 weeks after the last dose of study drug). The LLOQ for the assay was 25 IU/mL. (NCT01672983)
Timeframe: 24 weeks after last dose of study drug

Interventionpercentage of participants (Number)
Arm 1100
Arm 288.9
Arm 3100
Arm 4100
Arm 557.9
Arm 672.2

[back to top]

Number of Participants With Adverse Events (AEs)

An AE is any untoward medical occurrence, which does not necessarily have a causal relationship with treatment. A serious adverse event (SAE) is an AE that results in death, is life-threatening, results in or prolongs hospitalization, results in congenital anomaly, persistent or significant disability/incapacity, spontaneous or elective abortion, or requires intervention to prevent a serious outcome. AEs were rated for severity as either Mild: transient and easily tolerated; Moderate: causes discomfort and interrupts usual activities; or Severe: causes considerable interference with usual activities, may be incapacitating or life-threatening. AEs related to study drug were assessed as being either probably or possibly related by the investigator. Treatment-emergent AEs (TEAEs) were collected from the first dose of study drug administration to 30 days after last dose; SAEs were collected from the time that informed consent was obtained to 30 days after last dose. (NCT01672983)
Timeframe: TEAEs: up to 16 weeks for the 12-week treatment groups and up to 28 weeks for the 24-week treatment groups; SAEs: up to 65 weeks for the 12-week treatment groups and up to 77 weeks for the 24-week treatment groups.

,,,
Interventionparticipants (Number)
Adverse EventsSerious Adverse Events
Arm 1 + Arm 5281
Arm 2 + Arm 6312
Arm 3162
Arm 4150

[back to top] [back to top]

Number of Participants With Laboratory Test Results Meeting the Criteria for Abnormal, Grades 1-4

Hematocrit (%): Grade (Gr) 1= ≥28.5- <31.5; Gr 2= ≥24- <28.5; Gr 3= ≥19.5- <24; Gr 4= <19.5. Hemoglobin (g/dL): Grade (Gr)1=8.5-10.0; Gr 2=7.5-8.4; Gr 3=6.50-7.4; Gr 4= <6.5. Platelets (/mm^3): Gr 1=100,000-124,999; Gr 2=50,000-99,999; Gr 3=25,000-49,999; Gr 4= <25,000. White blood cells (/mm^3): Gr 1=2000-2500; Gr 2=1500-1999; Gr 3=1000-1499; Gr 4= <1000. Neutrophils (/mm^3): Gr 1=1000-1500; Gr 2= ≥750-1000; Gr 3= ≥500-750; Gr 4= <500. Alanine transaminase (ALT), alkaline phosphatase (ALP), aspartate transaminase (AST) (*upper limit of normal [ULN]): Gr 1=1.5-2.5; Gr 2=2.6-5.0; Gr 3=5.1-10.0; Gr 4= >10.0. Total bilirubin (adult and pediatric >14 days) (*ULN): Gr 1=1.1-1.5; Gr 2=1.6-2.5; Gr 3=2.6-5.0; Gr 4= >5.0. Albumin (g/dL): Gr 1= 3.1- 5. Lipase (*ULN): Gr 1=1.1-1.5; Gr 2=1.6-3.0; Gr 3=3.1-5.0; Gr 4= >5.0. (NCT01691794)
Timeframe: After first dose to last dose plus 30 days (assessed up to February 2017, approximately 42 months)

,,
InterventionParticipants (Number)
HematocritHemoglobinPlateletsWhite blood cellsNeutrophils+bands (absolute)ALTASTALPTotal bilirubinAlbuminAmylaseLipase
Atazanavir, 150 mg + Ritonavir, 100 mg (Weight: 15 to <20 kg)000011013220
Atazanavir, 200 mg + Ritonavir, 100 mg (Weight: 20 to <40 kg)350313661126152715
Atazanavir, 300 mg + Ritonavir, 100 mg (Weight: ≥40 kg)001049512176148

[back to top]

Number of Participants With Laboratory Test Results Meeting the Criteria for Abnormal, Grades 1-4 (Continued)

Blood urea nitrogen (*upper limit of normal [ULN]): Grade (Gr) 1=1.25-2.5; Gr 2=2.6-5.0; Gr 3=5.1-10; Gr 4= >10. Uric acid (mg/dL): Gr 1=7.5-10.0; Gr 2=10.1-12; Gr 3=12.1-15.0; Gr 4= >15.0. Bicarbonate (mEqL): Gr 1= 19.0-21.0; Gr 2=15.0-18.0; Gr 3=41-45; Gr 4= >45. Calcium, low (mg/dL): Gr 1=7.8-8.4; Gr 2=7.0-7.7; Gr 3=6.1-6.9; Gr 4= <6.1.Potassium (mEq/L), high: Gr 1=5.6-6.0; Gr 2=6.1-6.5; Gr 3=6.6-7.0; Gr 4= >7.0. Potassium (mEq/L), low: Gr 1=3.1-3.4; Gr 2=2.5-2.9; Gr 3=2.0-2.4; Gr 4= <2.0. Sodium (mEq/L), low: Gr 1=130-135; Gr 2=125-129; Gr 3=121-124; Gr 4= <1. Total cholesterol, fasting (mg/dL): Gr 1=200-239; Gr 2=240-300; Gr 3= >300; Gr 4=Not applicable (NA). Low-density lipoprotein (LDL) cholesterol, fasting (mg/dL): Gr 1=130-159; Gr 2=160-190; Gr 3= >190; Gr 4= NA. Glucose, low (mg/dL): Gr 1= 55-64; Gr 2=40-54; Gr 3=30-39; Gr 4= <30. Glucose, fasting (mg/dL): Gr 1=110-125; Gr 2=126-250; Gr 3=251-500; Gr 4 >500. (NCT01691794)
Timeframe: After first dose to last dose plus 30 days (assessed up to February 2017, approximately 42 months)

,,
InterventionParticipants (Number)
Blood urea nitrogenUric acidBicarbonate, lowCalcium, lowPotassium, highPotassium, lowTotal cholesterol, fasting (n=3, 31, 21)Sodium, lowLDL cholesterol, fasting (n=3, 31, 21)Glucose, low (n=1, 11, 9)Glucose, fasting, high (3, 31, 22)Calcium, highChloride, highChloride, lowGlucose, Non-Fasting, HighSodium, highCreatinineTriglycerides, fasting
Atazanavir, 150 mg + Ritonavir, 100 mg (Weight: 15 to <20 kg)003000010000000000
Atazanavir, 200 mg + Ritonavir, 100 mg (Weight: 20 to <40 kg)322922210137332000110
Atazanavir, 300 mg + Ritonavir, 100 mg (Weight: ≥40 kg)0317310603010200100

[back to top]

AUC

24-hour area under the curve (AUC) for pitavastatin when coadministered with efavirenz and with darunavir/ritonavir and 24-hour AUC for efavirenz or darunavir when coadministered with pitavastatin (NCT01695954)
Timeframe: 0 to 24 hours

Interventionng*hr/mL (Mean)
Arm A: (Pitavastatin and Efavirenz)85.3
Arm B: (Pitavastatin and Darunavir)62.8

[back to top]

GMR of Cmax of Pitavastatin When Coadministered With Efavirenz or With Darunavir/Ritonavir

Geometric Mean Ratio (GMR) of Cmax for pitavastatin with Efavirenz vs. alone and GMR of Cmax for pitavastatin with darunavir/ritonavir vs. alone was reported. (NCT01695954)
Timeframe: Day 18

Interventionng/mL (Geometric Mean)
Arm A: (Pitavastatin and Efavirenz)1.20
Arm B: (Pitavastatin and Ritonavir-boosted Darunavir)0.93

[back to top]

GMR of 24- Hour AUC of Pitavastatin When Coadministered With Efavirenz or With Darunavir/Ritonavir Over 24 Hour AUC of Pitavastatin

Geometric Mean Ratio (GMR) of 24- Hour Area under the plasma drug concentration-time curve (AUC) of pitavastatin when coadministered with efavirenz or with darunavir/ritonavir over 24 Hour (AUC) of pitavastatin (NCT01695954)
Timeframe: 0 to 24 hours

,
InterventionRatio (Geometric Mean)
GMR of Pitavastatin with (EFV or DRV)/PitavastatinGMR of (EFV or DRV) with Pitavastatin/(EFV or DRV)
Arm A: (Pitavastatin and Efavirenz).89.90
Arm B: (Pitavastatin and Darunavir).911.08

[back to top]

Percentage of Participants Who Experienced Relapse˅Overall Without and With New HCV Infection

Relapse is defined as a confirmed HCV RNA ≥ LLOQ at any time during the post-treatment period for a participant who had HCV RNA < LLOQ at the end of treatment. Relapse˅overall was defined as a confirmed HCV RNA ≥ LLOQ between end of treatment and up to and including the last HCV RNA measurement collected in the post-treatment Period for a participant with HCV RNA < LLOQ at Final Treatment Visit who completed treatment. (NCT01773070)
Timeframe: Up to 3 years post-treatment

Interventionpercentage of participants (Number)
Relapse˅overall Without New HCV InfectionRelapse˅overall With New HCV Infection
All Participants2.20.2

[back to top]

Percentage of Participants Who Experienced Relapse12 Without and With New HCV Infection

Relapse is defined as a confirmed HCV RNA ≥ LLOQ at any time during the post-treatment period for a participant who had HCV RNA < LLOQ at the end of treatment. Relapse12 is defined as a confirmed HCV RNA ≥ LLOQ between end of treatment and 12 weeks after last actual dose of study drug (up to and including the SVR12 assessment time point) for a participant with HCV RNA < LLOQ at Final Treatment Visit who completed treatment. (NCT01773070)
Timeframe: From the end of treatment through 12 weeks post-treatment

Interventionpercentage of participants (Number)
Relapse12 Without New HCV InfectionRelapse12 With New HCV Infection
All Participants2.00

[back to top]

Percentage of Participants Who Experienced Relapse12overall With and Without New HCV Infection

Relapse is defined as a confirmed HCV ribonucleic acid (RNA) ≥ the lower limit of quantitation (LLOQ) at any time during the post-treatment period for a participant who had HCV RNA < LLOQ at the end of treatment. Relapse12overall is defined as a confirmed HCV RNA ≥ LLOQ at any time after the sustained virologic response at Week 12 post-dosing (SVR12) assessment time point for a participant who achieved SVR12 and had post-SVR12 HCV RNA data available. SVR12 is defined as HCV RNA < LLOQ in the SVR12 window (12 weeks after the last actual dose of study drug) without any confirmed quantifiable (≥ LLOQ) post-treatment value before or during that SVR window. New HCV infection is defined as re-infection with a different HCV isolate. (NCT01773070)
Timeframe: Up to 3 years post-treatment

Interventionpercentage of participants (Number)
Relapse12overall With New HCV InfectionRelapse12overall Without New HCV Infection
All Participants0.20.2

[back to top]

Percentage of Participants Who Experienced Relapse24 Without and With New HCV Infection

Relapse is defined as a confirmed HCV RNA ≥ LLOQ at any time during the post-treatment period for a participant who had HCV RNA < LLOQ at the end of treatment. Relapse24 is defined as a confirmed HCV RNA ≥ LLOQ within the sustained virologic response at Week 24 post-dosing (SVR24) window for a participant who achieved SVR12 and had HCV RNA data available in the SVR24 window. (NCT01773070)
Timeframe: From the end of treatment through 24 weeks post-treatment

Interventionpercentage of participants (Number)
Relapse24 Without New HCV InfectionRelapse24 With New HCV Infection
All Participants0.20

[back to top]

Number of HCV Genotype (GT)1a-Infected Participants With Persistence of Treatment-Emergent Substitutions in NS3, NS5A, or NS5B

The persistence of specific hepatitis C amino acid variants (treatment-emergent substitutions) associated with drug resistance in NS3, NS5A, or NS5B was evaluated in participants who had not achieved SVR12. Post-baseline time points were calculated relative to the last dose of study drug in the previous study. (NCT01773070)
Timeframe: from the last dose of study drug in the previous study up to 3 years post-treatment

InterventionParticipants (Count of Participants)
NS3: time of failureNS3: post-treatment week 48NS5A: time of failureNS5A: post-treatment week 48NS5A: post-treatment week 96NS5A: post-treatment week 132NS5B: time of failureNS5B: post-treatment week 24NS5B: post-treatment week 96
All Participants70131064731

[back to top]

Percentage of Participants With Sustained Virologic Response 24 Weeks Post-treatment (SVR24)

SVR24 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification [NCT01782495)
Timeframe: 24 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Arm A97.1
Arm B96.3
Arm C100
Arm D100
Arm E100
Arm F95.5
Arm G100
Arm H66.7
Arm I100
Arm J100
Arm K100

[back to top]

Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

SVR12 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification [NCT01782495)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Arm A97.1
Arm B96.3
Arm C100
Arm D100
Arm E100
Arm F95.5
Arm G100
Arm H66.7
Arm I100
Arm J100
Arm K100

[back to top]

Percentage of Participants With On-treatment Virologic Failure

On-treatment virologic failure was defined as confirmed increase of > 1 log(subscript)10(subscript) IU/mL above the lowest value post-baseline HCV RNA during treatment, or confirmed HCV RNA ≥ LLOQ at any point during treatment after HCV RNA < LLOQ, or HCV RNA ≥ LLOQ persistently during treatment with at least 6 weeks (≥ 36 days) of treatment. (NCT01782495)
Timeframe: Up to 12 weeks (for 12-week treatment) or 24 weeks (for 24-week treatment)

Interventionpercentage of participants (Number)
Arm A0
Arm B3.7
Arm C0
Arm D0
Arm E0
Arm F0
Arm G0
Arm H0
Arm I0
Arm J0
Arm K0

[back to top]

Percentage of Participants With Post-treatment Relapse

Post-treatment relapse was defined as confirmed HCV RNA ≥ LLOQ between the end of treatment and 12 weeks after the last dose of study drug among participants who completed treatment with HCV RNA levels < LLOQ at the end of treatment. (NCT01782495)
Timeframe: From the end of treatment through 12 weeks after the last dose of study drug

Interventionpercentage of participants (Number)
Arm A3.0
Arm B0
Arm C0
Arm D0
Arm E0
Arm F4.8
Arm G0
Arm H0
Arm I0
Arm J0
Arm K0

[back to top]

Change in Plasma Log10 HIV-1 Ribonucleic Acid (RNA) Levels From Baseline to Day 11

Antiviral activity of BMS-955176 was estimated by measuring the plasma HIV-1 RNA levels in the HIV-1 infected participants. Change in the plasma HIV-1 RNA levels were measured in the participants infected with HIV-1 clade B and C who received BMS-955176 monotherapy. Baseline was Day 1. Change from Baseline was post-Baseline individual values minus Baseline values. (NCT01803074)
Timeframe: Baseline (Day 1) and Day 11 after the final dose with BMS-955176

InterventionLog10 copies per milliliter (c/mL) (Mean)
Part A-Group 1: BMS-955176 (5 mg)-0.138
Part A-Group 2: BMS-955176 (10 mg)-0.567
Part A-Group 3: BMS-955176 (20 mg)-0.889
Part A-Group 4: BMS-955176 (40 mg)-1.279
Part A-Group 9: BMS-955176 (80 mg)-1.339
Part A-Group 10: BMS-955176 (120 mg)-1.326
Part B-Group 5: BMS-955176 (40 mg) + Atazanavir-1.216
Part B-Group 6: BMS-955176 (40 mg) + Atazanavir + Ritonavir-1.431
Part B-Group 7: Atazanavir+Ritonavir+Tenofovir+Emtricitabine-1.544
Part B-Group 12: BMS-955176 (80 mg) + Atazanavir-1.521
Part C-Group 8: BMS-955176 (40 mg)-1.29
Part C-Group 13: BMS-955176 (120 mg)-0.938
Placebo Clade B0.118
Placebo Clade C-0.172

[back to top]

Area Under The Plasma Concentration - Time Curve Over the Dosing Interval (AUC([Tau]) - Part A and C

AUC(tau) was defined as the area under the plasma concentration - time curve over the dosing interval. (NCT01803074)
Timeframe: Pre-dose Day 1 and Day 10

,,,,,,,
InterventionNanogram*hour/milliliter (Geometric Mean)
Day 1Day 10
Part A-Group 1: BMS-955176 (5 mg)1151.0622720.237
Part A-Group 10: BMS-955176 (120 mg)21872.7244182.4
Part A-Group 2: BMS-955176 (10 mg)2869.6265168.553
Part A-Group 3: BMS-955176 (20 mg)5132.95111751.82
Part A-Group 4: BMS-955176 (40 mg)10088.2322984.83
Part A-Group 9: BMS-955176 (80 mg)17057.2639341.11
Part C-Group 13: BMS-955176 (120 mg)26753.7453972.71
Part C-Group 8: BMS-955176 (40 mg)10936.925556.64

[back to top]

Maximum Decline From Baseline in Log10 HIV-1 RNA - Part B

Antiviral activity of BMS-955176 was estimated by measuring the plasma HIV-1 RNA levels in the HIV-1 infected participants. Maximum decline from Baseline in the plasma HIV-1 RNA levels were measured in the participants infected with HIV-1 clade B and C. Baseline was Day 1. Change from Baseline was post-Baseline individual values minus Baseline values. (NCT01803074)
Timeframe: Baseline (Day 1) up to Day 42

InterventionLog10 copies/mL (Median)
Part B-Group 5: BMS-955176 (40 mg) + Atazanavir-1.858
Part B-Group 6: BMS-955176 (40 mg) + Atazanavir + Ritonavir-2.202
Part B-Group 7: Atazanavir+Ritonavir+Tenofovir+Emtricitabine-2.39
Part B-Group 12: BMS-955176 (80 mg) + Atazanavir-2.228

[back to top]

Maximum Decline From Baseline in Log10 HIV-1 RNA - Part A and C

Antiviral activity of BMS-955176 was estimated by measuring the plasma HIV-1 RNA levels in the HIV-1 infected participants. Maximum decline from Baseline in the plasma HIV-1 RNA levels were measured in the participants infected with HIV-1 clade B and C. Baseline was Day 1. Change from Baseline was post-Baseline individual values minus Baseline values. (NCT01803074)
Timeframe: Baseline (Day 1) up to Day 24

InterventionLog10 copies/mL (Median)
Part A-Group 1: BMS-955176 (5 mg)-0.498
Part A-Group 2: BMS-955176 (10 mg)-0.976
Part A-Group 3: BMS-955176 (20 mg)-1.115
Part A-Group 4: BMS-955176 (40 mg)-1.701
Part A-Group 9: BMS-955176 (80 mg)-1.555
Part A-Group 10: BMS-955176 (120 mg)-1.654
Part C-Group 8: BMS-955176 (40 mg)-1.352
Part C-Group 13: BMS-955176 (120 mg)-1.257
Placebo Clade B-0.381
Placebo Clade C-0.419

[back to top]

Number of Participants With Clinically Significant Changes in Heart Rate

Heart rate was measured after the participants had been seated quietly for at least 5 minutes. Criteria used to determine heart rate that are outside of a pre-specified range, where changes from Baseline are based on matched postural positions and are calculated as parameter value - Baseline parameter value: Value >100 and change from Baseline > 30, or Value < 55 and change from Baseline < -15. (NCT01803074)
Timeframe: Day 1 to end of the study (Day 42)

InterventionParticipants (Count of Participants)
Part A-Group 1: BMS-955176 (5 mg)0
Part A-Group 2: BMS-955176 (10 mg)0
Part A-Group 3: BMS-955176 (20 mg)0
Part A-Group 4: BMS-955176 (40 mg)0
Part A-Group 9: BMS-955176 (80 mg)0
Part A-Group 10: BMS-955176 (120 mg)0
Part B-Group 5: BMS-955176 (40 mg) + Atazanavir0
Part B-Group 6: BMS-955176 (40 mg) + Atazanavir + Ritonavir0
Part B-Group 7: Atazanavir+Ritonavir+Tenofovir+Emtricitabine1
Part B-Group 12: BMS-955176 (80 mg) + Atazanavir0
Part C-Group 8: BMS-955176 (40 mg)0
Part C-Group 13: BMS-955176 (120 mg)2
Placebo Clade B1
Placebo Clade C0

[back to top]

Plasma Half-life: Part A and C

Half-life of the terminal log-linear phase, (T-half), was calculated as natural logarithm of 2 (ln2)/λ, where λ is the absolute value of the slope of the terminal log-linear phase. T-half was derived by non-compartmental methods, using a validated pharmacokinetic (PK) analysis program: KineticaTM 5.0 within eToolbox (version 2.7). (NCT01803074)
Timeframe: Baseline (Day 1) to Day 10

InterventionHours (Median)
Part A-Group 1: BMS-955176 (5 mg)32.134
Part A-Group 2: BMS-955176 (10 mg)31.967
Part A-Group 3: BMS-955176 (20 mg)27.382
Part A-Group 4: BMS-955176 (40 mg)33.475
Part A-Group 9: BMS-955176 (80 mg)29.171
Part A-Group 10: BMS-955176 (120 mg)34.574
Part C-Group 8: BMS-955176 (40 mg)31.565
Part C-Group 13: BMS-955176 (120 mg)35.278

[back to top]

Time to Reach Maximum Plasma Concentration (Tmax) - Part A and C

Time to reach the maximum plasma concentration was directly determined from concentration time data. (NCT01803074)
Timeframe: Pre-dose Day 1 and Day 10

,,,,,,,
InterventionHours (Median)
Day 1Day 10
Part A-Group 1: BMS-955176 (5 mg)33
Part A-Group 10: BMS-955176 (120 mg)32.5
Part A-Group 2: BMS-955176 (10 mg)2.513
Part A-Group 3: BMS-955176 (20 mg)34
Part A-Group 4: BMS-955176 (40 mg)43
Part A-Group 9: BMS-955176 (80 mg)3.53
Part C-Group 13: BMS-955176 (120 mg)3.533
Part C-Group 8: BMS-955176 (40 mg)3.53

[back to top]

Plasma Concentration 24 Hours Post-Dose (C24) - Part B

C24 was defined as the plasma concentration of BMS-955176 at 24 hours post-dose. (NCT01803074)
Timeframe: 24 hours post-dose

,,
InterventionNanogram/milliliter (Geometric Mean)
Day 1Day 28
Part B-Group 12: BMS-955176 (80 mg) + Atazanavir899.3642010.679
Part B-Group 5: BMS-955176 (40 mg) + Atazanavir462.3121099.313
Part B-Group 6: BMS-955176 (40 mg) + Atazanavir + Ritonavir520.0481163.177

[back to top]

Plasma Concentration 24 Hours Post-Dose (C24) - Part A and C

C24 was defined as the plasma concentration of BMS-955176 at 24 hours post-dose. (NCT01803074)
Timeframe: 24 hours post-dose

,,,,,,,
InterventionNanogram/milliliter (Geometric Mean)
Day 1Day 10
Part A-Group 1: BMS-955176 (5 mg)34.94681.642
Part A-Group 10: BMS-955176 (120 mg)624.7451288.985
Part A-Group 2: BMS-955176 (10 mg)79.002138.775
Part A-Group 3: BMS-955176 (20 mg)154.5325.934
Part A-Group 4: BMS-955176 (40 mg)286.268713.077
Part A-Group 9: BMS-955176 (80 mg)482.3491150.397
Part C-Group 13: BMS-955176 (120 mg)865.8671691.306
Part C-Group 8: BMS-955176 (40 mg)339.173779.438

[back to top]

Percent Change From Baseline in Cluster of Differentiation (CD) 4+ and CD8+ Lymphocyte Percent - Part B

Percent Change in the CD4+ and CD8+ cell counts from Baseline were measured in the participants infected with HIV-1 clade B and C who received BMS-955176 + ATV or BMS-955176 + ATV + RTV therapy. Baseline was Day 1. Change from Baseline was post-Baseline individual values minus Baseline values. (NCT01803074)
Timeframe: Baseline (Day 1) up to Day 42

,,,
InterventionPercent change (Mean)
CD4+CD8+
Part B-Group 12: BMS-955176 (80 mg) + Atazanavir-0.75-1.25
Part B-Group 5: BMS-955176 (40 mg) + Atazanavir2.4-2.8
Part B-Group 6: BMS-955176 (40 mg) + Atazanavir + Ritonavir3.25-6.25
Part B-Group 7: Atazanavir+Ritonavir+Tenofovir+Emtricitabine4.75-3.75

[back to top]

Area Under The Plasma Concentration - Time Curve Over the Dosing Interval (AUC[Tau]) - Part B

AUC(tau) was defined as the area under the plasma concentration - time curve over the dosing interval. (NCT01803074)
Timeframe: Pre-dose Day 1 and Day 28

,,
InterventionNanogram*hour/milliliter (Geometric Mean)
Day 1Day 28
Part B-Group 12: BMS-955176 (80 mg) + Atazanavir24478.3559915.72
Part B-Group 5: BMS-955176 (40 mg) + Atazanavir12147.2331406.32
Part B-Group 6: BMS-955176 (40 mg) + Atazanavir + Ritonavir12954.834225.08

[back to top]

Percent Change From Baseline in Cluster of Differentiation (CD) 4+ and CD8+ Lymphocyte Percent - Part A and C

Percent Change in the CD4+ and CD8+ cell counts from Baseline were measured in the participants infected with HIV-1 clade B and C who received BMS-955176 + ATV or BMS-955176 + ATV + RTV therapy. Baseline was Day 1. Change from Baseline was post-Baseline individual values minus Baseline values. (NCT01803074)
Timeframe: Baseline (Day 1) up to Day 24

,,,,,,,,,
InterventionPercent change (Mean)
CD4+CD8+
Part A-Group 1: BMS-955176 (5 mg)2.331.17
Part A-Group 10: BMS-955176 (120 mg)0.29-2.29
Part A-Group 2: BMS-955176 (10 mg)0.290.43
Part A-Group 3: BMS-955176 (20 mg)-1.290
Part A-Group 4: BMS-955176 (40 mg)0.861
Part A-Group 9: BMS-955176 (80 mg)2.13-0.25
Part C-Group 13: BMS-955176 (120 mg)3.17-4.25
Part C-Group 8: BMS-955176 (40 mg)0.50
Placebo Clade B-0.221.75
Placebo Clade C2.75-1.33

[back to top]

Degree of Fluctuation (DF): Part A and C

DF was calculated as the difference between Cmax and Cmin divided by Css-avg. DF was derived by non-compartmental methods, using a validated pharmacokinetic (PK) analysis program: KineticaTM 5.0 within eToolbox (version 2.7). (NCT01803074)
Timeframe: Baseline (Day 1) to Day 10

InterventionRatio (Geometric Mean)
Part A-Group 1: BMS-955176 (5 mg)0.766
Part A-Group 2: BMS-955176 (10 mg)0.912
Part A-Group 3: BMS-955176 (20 mg)0.758
Part A-Group 4: BMS-955176 (40 mg)0.78
Part A-Group 9: BMS-955176 (80 mg)0.779
Part A-Group 10: BMS-955176 (120 mg)0.818
Part C-Group 8: BMS-955176 (40 mg)0.723
Part C-Group 13: BMS-955176 (120 mg)0.727

[back to top] [back to top]

Number of Participants With Clinically Significant Grade 3/4 Laboratory Abnormalities From Baseline

Laboratory abnormalities were determined and graded using the Division of Acquired Immune Deficiency Syndrome (AIDS) Table for Grading the Severity of Adult and Pediatric Adverse Events, version 1.0, December 2004. (NCT01803074)
Timeframe: Day 1 to up to end of the study (Day 42)

,,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
Neutrophils (Absolute)Bilirubin (Total)
Part A-Group 1: BMS-955176 (5 mg)00
Part A-Group 10: BMS-955176 (120 mg)10
Part A-Group 2: BMS-955176 (10 mg)00
Part A-Group 3: BMS-955176 (20 mg)00
Part A-Group 4: BMS-955176 (40 mg)00
Part A-Group 9: BMS-955176 (80 mg)00
Part B-Group 12: BMS-955176 (80 mg) + Atazanavir10
Part B-Group 5: BMS-955176 (40 mg) + Atazanavir02
Part B-Group 6: BMS-955176 (40 mg) + Atazanavir + Ritonavir05
Part B-Group 7: Atazanavir+Ritonavir+Tenofovir+Emtricitabine03
Part C-Group 13: BMS-955176 (120 mg)00
Part C-Group 8: BMS-955176 (40 mg)00
Placebo Clade B00
Placebo Clade C00

[back to top]

Time to Maximum Decline in Log 10 HIV-1 RNA - Part A and C

Antiviral activity of BMS-955176 was estimated by measuring the plasma HIV-1 RNA levels in the HIV-1 infected participants. Time to maximum decline in the plasma HIV-1 RNA levels were measured in the participants infected with HIV-1 clade B and C. Baseline was Day 1. Change from Baseline was post-Baseline individual values minus Baseline values. (NCT01803074)
Timeframe: Baseline (Day 1) up to Day 24

InterventionHours (Median)
Part A-Group 1: BMS-955176 (5 mg)168
Part A-Group 2: BMS-955176 (10 mg)216
Part A-Group 3: BMS-955176 (20 mg)203.9
Part A-Group 4: BMS-955176 (40 mg)240.15
Part A-Group 9: BMS-955176 (80 mg)204
Part A-Group 10: BMS-955176 (120 mg)240.2
Part C-Group 8: BMS-955176 (40 mg)228.05
Part C-Group 13: BMS-955176 (120 mg)215.8
Placebo Clade B216.2
Placebo Clade C132.05

[back to top]

Number of Participants With Clinically Significant Changes in Electrocardiogram (ECG)

Participants with out of range ECG intervals were summarized. Criteria used to determine ECG results that are outside of a pre-specified range: PR (milliseconds [msec]): Value >200; QRS (msec): Value >120; QT (msec): Value >500 or change from Baseline >30; corrected QT interval Fridericia's formula (QTcF) (msec): Value >450 or change from Baseline >30. (NCT01803074)
Timeframe: Day 1 to end of the study (Day 42)

,,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
PR > 200 msecQRS > 120 msecQT > 500 msecQTcB > 450 msecQTcF > 450 msec
Part A-Group 1: BMS-955176 (5 mg)10000
Part A-Group 10: BMS-955176 (120 mg)20000
Part A-Group 2: BMS-955176 (10 mg)10000
Part A-Group 3: BMS-955176 (20 mg)10001
Part A-Group 4: BMS-955176 (40 mg)00000
Part A-Group 9: BMS-955176 (80 mg)00000
Part B-Group 12: BMS-955176 (80 mg) + Atazanavir10000
Part B-Group 5: BMS-955176 (40 mg) + Atazanavir01000
Part B-Group 6: BMS-955176 (40 mg) + Atazanavir + Ritonavir10000
Part B-Group 7: Atazanavir+Ritonavir+Tenofovir+Emtricitabine10000
Part C-Group 13: BMS-955176 (120 mg)00000
Part C-Group 8: BMS-955176 (40 mg)10000
Placebo Clade B00000
Placebo Clade C00010

[back to top]

Number of Participants With Abnormal Changes in Physical Examination

Participants with abnormal changes in physical examination is presented. (NCT01803074)
Timeframe: Day 1 to end of the study (Day 42)

,,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
HeightWeightBody mass index
Part A-Group 1: BMS-955176 (5 mg)000
Part A-Group 10: BMS-955176 (120 mg)000
Part A-Group 2: BMS-955176 (10 mg)000
Part A-Group 3: BMS-955176 (20 mg)000
Part A-Group 4: BMS-955176 (40 mg)000
Part A-Group 9: BMS-955176 (80 mg)000
Part B-Group 12: BMS-955176 (80 mg) + Atazanavir000
Part B-Group 5: BMS-955176 (40 mg) + Atazanavir000
Part B-Group 6: BMS-955176 (40 mg) + Atazanavir + Ritonavir000
Part B-Group 7: Atazanavir+Ritonavir+Tenofovir+Emtricitabine000
Part C-Group 13: BMS-955176 (120 mg)000
Part C-Group 8: BMS-955176 (40 mg)000
Placebo Clade B000
Placebo Clade C000

[back to top]

Maximum Observed Plasma Concentrations (Cmax) - Part B

Cmax was defined as the peak plasma concentration of a drug after administration, obtained directly from the plasma concentration-time curve. (NCT01803074)
Timeframe: Pre-dose Day 1 and Day 28

,,
InterventionNanogram/milliliter (Geometric Mean)
Day 1Day 28
Part B-Group 12: BMS-955176 (80 mg) + Atazanavir1493.3363159.181
Part B-Group 5: BMS-955176 (40 mg) + Atazanavir695.5961667.817
Part B-Group 6: BMS-955176 (40 mg) + Atazanavir + Ritonavir770.9751852

[back to top]

Time to Maximum Decline in Log 10 HIV-1 RNA - Part B

Antiviral activity of BMS-955176 was estimated by measuring the plasma HIV-1 RNA levels in the HIV-1 infected participants. Time to maximum decline in the plasma HIV-1 RNA levels were measured in the participants infected with HIV-1 clade B and C. Baseline was Day 1. Change from Baseline was post-Baseline individual values minus Baseline values. (NCT01803074)
Timeframe: Baseline (Day 1) up to Day 42

InterventionHours (Median)
Part B-Group 5: BMS-955176 (40 mg) + Atazanavir624
Part B-Group 6: BMS-955176 (40 mg) + Atazanavir + Ritonavir636.05
Part B-Group 7: Atazanavir+Ritonavir+Tenofovir+Emtricitabine588
Part B-Group 12: BMS-955176 (80 mg) + Atazanavir636.05

[back to top]

Maximum Observed Plasma Concentrations (Cmax) - Part A and C

Cmax was defined as the peak plasma concentration of a drug after administration, obtained directly from the plasma concentration-time curve. (NCT01803074)
Timeframe: Pre-dose Day 1 and Day 10

,,,,,,,
InterventionNanogram/milliliter (Geometric Mean)
Day 1Day 10
Part A-Group 1: BMS-955176 (5 mg)79.376170.778
Part A-Group 10: BMS-955176 (120 mg)1515.3892809.671
Part A-Group 2: BMS-955176 (10 mg)201.498337.379
Part A-Group 3: BMS-955176 (20 mg)349.466705.073
Part A-Group 4: BMS-955176 (40 mg)791.3171476.166
Part A-Group 9: BMS-955176 (80 mg)1155.4482466.447
Part C-Group 13: BMS-955176 (120 mg)1907.7473377.967
Part C-Group 8: BMS-955176 (40 mg)793.5691560.122

[back to top]

Number of Participants With Clinically Significant Changes in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)

Systolic BP (millimeter of mercury [mmHg]): value >140 and change from Baseline >20, or value <90 and change from Baseline <-20; Diastolic BP (mmHg): value >90 and change from Baseline >10, or value <55 and change from Baseline <-10. (NCT01803074)
Timeframe: Day 1 to end of the study (Day 42)

,,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
SBPDBP
Part A-Group 1: BMS-955176 (5 mg)00
Part A-Group 10: BMS-955176 (120 mg)00
Part A-Group 2: BMS-955176 (10 mg)01
Part A-Group 3: BMS-955176 (20 mg)01
Part A-Group 4: BMS-955176 (40 mg)00
Part A-Group 9: BMS-955176 (80 mg)00
Part B-Group 12: BMS-955176 (80 mg) + Atazanavir01
Part B-Group 5: BMS-955176 (40 mg) + Atazanavir00
Part B-Group 6: BMS-955176 (40 mg) + Atazanavir + Ritonavir11
Part B-Group 7: Atazanavir+Ritonavir+Tenofovir+Emtricitabine00
Part C-Group 13: BMS-955176 (120 mg)00
Part C-Group 8: BMS-955176 (40 mg)01
Placebo Clade B01
Placebo Clade C00

[back to top]

Change From Baseline in Cluster of Differentiation (CD) 4+ and CD8+ Lymphocyte Counts - Part A and C

Change in the CD4+ and CD8+ cell counts from Baseline were measured in the participants infected with HIV-1 clade B and C who received BMS-955176 + ATV or BMS-955176 + ATV + RTV therapy. Baseline was Day 1. Change from Baseline was post-Baseline individual values minus Baseline values. (NCT01803074)
Timeframe: Baseline (Day 1) up to Day 24

,,,,,,,,,
InterventionCells/microliter (Mean)
CD4+CD8+
Part A-Group 1: BMS-955176 (5 mg)-21.8-95
Part A-Group 10: BMS-955176 (120 mg)-56.7-161.3
Part A-Group 2: BMS-955176 (10 mg)14.6-8.3
Part A-Group 3: BMS-955176 (20 mg)-70.1-107.4
Part A-Group 4: BMS-955176 (40 mg)-23.6-57.3
Part A-Group 9: BMS-955176 (80 mg)-43.8-194.6
Part C-Group 13: BMS-955176 (120 mg)24.5-155.8
Part C-Group 8: BMS-955176 (40 mg)-53.7-214.4
Placebo Clade B-77.3-93.1
Placebo Clade C18-136.3

[back to top]

Apparent Total Body Clearance: Part A and C

Apparent total body clearance was derived by non-compartmental methods, using a validated pharmacokinetic (PK) analysis program: KineticaTM 5.0 within eToolbox (version 2.7). (NCT01803074)
Timeframe: Baseline (Day 1) to Day 10

InterventionMilliliters/minute (Geometric Mean)
Part A-Group 1: BMS-955176 (5 mg)30.635
Part A-Group 2: BMS-955176 (10 mg)32.246
Part A-Group 3: BMS-955176 (20 mg)28.364
Part A-Group 4: BMS-955176 (40 mg)29.005
Part A-Group 9: BMS-955176 (80 mg)33.892
Part A-Group 10: BMS-955176 (120 mg)45.267
Part C-Group 8: BMS-955176 (40 mg)26.086
Part C-Group 13: BMS-955176 (120 mg)37.056

[back to top]

Average Observed Plasma Concentration at Steady State (Css-avg): Part A and C

Css-avg was calculated by the quotient of AUC(TAU) and the dosing interval (24 h). Css-avg was derived by non-compartmental methods, using a validated pharmacokinetic (PK) analysis program: KineticaTM 5.0 within eToolbox (version 2.7). (NCT01803074)
Timeframe: Baseline (Day 1) to Day 10

InterventionNanogram/milliliter (Geometric Mean)
Part A-Group 1: BMS-955176 (5 mg)113.326
Part A-Group 2: BMS-955176 (10 mg)215.111
Part A-Group 3: BMS-955176 (20 mg)489.507
Part A-Group 4: BMS-955176 (40 mg)956.222
Part A-Group 9: BMS-955176 (80 mg)1639.471
Part A-Group 10: BMS-955176 (120 mg)1841.413
Part C-Group 8: BMS-955176 (40 mg)1065.435
Part C-Group 13: BMS-955176 (120 mg)2256.793

[back to top]

Accumulation Index (AI): Part A and C

Accumulation index was calculated by dividing the AUC(tau) or Cmax or C24 of BMS-955176 on Day 10 by the AUC(TAU) or Cmax or C24, respectively, of BMS-955176 on Day 1. (NCT01803074)
Timeframe: Baseline and Day 10

,,,,,,,
InterventionRatio (Geometric Mean)
CmaxC24AUC
Part A-Group 1: BMS-955176 (5 mg)2.1522.3362.363
Part A-Group 10: BMS-955176 (120 mg)1.8542.0632.02
Part A-Group 2: BMS-955176 (10 mg)1.6741.7571.801
Part A-Group 3: BMS-955176 (20 mg)2.0182.112.289
Part A-Group 4: BMS-955176 (40 mg)1.8562.4912.278
Part A-Group 9: BMS-955176 (80 mg)2.1352.3852.306
Part C-Group 13: BMS-955176 (120 mg)1.7711.9532.017
Part C-Group 8: BMS-955176 (40 mg)1.9662.2982.337

[back to top]

Time to Reach Maximum Plasma Concentration (Tmax) - Part B

Tmax was directly determined from concentration time data. (NCT01803074)
Timeframe: Pre-dose Day 1 and Day 28

,,
InterventionHours (Median)
Day 1Day 28
Part B-Group 12: BMS-955176 (80 mg) + Atazanavir54.5
Part B-Group 5: BMS-955176 (40 mg) + Atazanavir5.014.5
Part B-Group 6: BMS-955176 (40 mg) + Atazanavir + Ritonavir5.055

[back to top]

Change From Baseline in Cluster of Differentiation (CD) 4+ and CD8+ Lymphocyte Counts - Part B

Change in the CD4+ and CD8+ cell counts from Baseline were measured in the participants infected with HIV-1 clade B and C who received BMS-955176 + ATV or BMS-955176 + ATV + RTV therapy. Baseline was Day 1. Change from Baseline was post-Baseline individual values minus Baseline values. (NCT01803074)
Timeframe: Baseline (Day 1) up to Day 42

,,,
InterventionCells/microliter (Mean)
CD4+CD8+
Part B-Group 12: BMS-955176 (80 mg) + Atazanavir-89-147
Part B-Group 5: BMS-955176 (40 mg) + Atazanavir-133.2-442.8
Part B-Group 6: BMS-955176 (40 mg) + Atazanavir + Ritonavir-106.4-466.1
Part B-Group 7: Atazanavir+Ritonavir+Tenofovir+Emtricitabine33-216.3

[back to top]

Change in CD4 Percent

Change in CD4 percent from baseline (NCT01818258)
Timeframe: Weeks 0, 12, 24, 36 and 48

,
Interventionpercent (Mean)
Change in CD4 Percent at Week 12Change in CD4 Percent at Week 24Change in CD4 Percent at Week 36Change in CD4 Percent at Week 48
Normal Nutrition/Mild Malnutrition Cohort3.06.87.16.7
Severe Malnutrition Cohort3.39.310.312.7

[back to top]

Change in HIV Viral Load From Baseline

Change from baseline in plasma HIV RNA viral load (NCT01818258)
Timeframe: Weeks 0, 12, 24, 36 and 48

,
Interventionlog10 copies/mL (Mean)
Change in Log10 Viral Load between Baseline and Week 12Change in Log10 Viral Load between Baseline and Week 24Change in Log10 Viral Load between Baseline and Week 36Change in Log10 Viral Load between Baseline and Week 48
Normal Nutrition/Mild Malnutrition Cohort-2.1-2.5-2.5-2.6
Severe Malnutrition Cohort-1.4-1.7-1.8-1.8

[back to top]

Change in Mid-upper Arm Circumference

Change in mid-upper arm circumference (MUAC) from entry (NCT01818258)
Timeframe: Weeks 0, 24, and 48

,
Interventioncentimeters (Mean)
Week 24Week 48
Normal Nutrition/Mild Malnutrition Cohort1.21.6
Severe Malnutrition Cohort2.63.5

[back to top]

Change in WHO Weight-for-height Z-score

Change in WHO weight-for-height Z-score from entry. A Z-score indicates the number of standard deviations the measurement is away from the mean. A Z-score of 0 is equal to the mean of the reference population. Negative numbers indicate values lower than the reference population and positive numbers indicate values higher than the reference population. The reference population was determined by the World Health Organization for children from 0 up to 5 years. (NCT01818258)
Timeframe: Weeks 0, 24, and 48

,
InterventionZ-Score (Mean)
Week 24Week 48
Normal Nutrition/Mild Malnutrition Cohort0.10.4
Severe Malnutrition Cohort2.32.7

[back to top]

Free Fraction of LPV at Hour 2 Post Dose

Free fraction of steady-state lopinavir at 2 hours post dose (NCT01818258)
Timeframe: Weeks 1, 12 and 24

,
InterventionPercent of Unbound LPV (Mean)
Week 1Week 12Week 24
Normal Nutrition/Mild Malnutrition Cohort3.26.02.1
Severe Malnutrition Cohort0.82.23.1

[back to top]

Minimum Trough Concentration (Ctrough) of Lopinavir

Count (%) of participants with minimum trough concentration (Ctrough) of steady-state Lopinavir >= 1 ug/mL (NCT01818258)
Timeframe: Measured 0, 1, 2, 4, 8, and 12 hours post-dose on 1, 4, 8, 12, 16, 24, 36 and 48 weeks following study entry

,
InterventionParticipants (Count of Participants)
Week 1Week 4Week 8Week 12Week 16Week 24Week 36Week 48
Normal Nutrition/Mild Malnutrition Cohort2118181815191918
Severe Malnutrition Cohort1214131711141616

[back to top]

Plasma Clearance of Lamivudine

Steady-state plasma clearance (CL/F) of Lamivudine (3TC) (NCT01818258)
Timeframe: 0, 1, 2, 4, 8, and 12 hours post-dose on 1, 12, and 24 weeks following study entry

,
InterventionL/hours (Geometric Mean)
3TC CL/F at Week 13TC CL/F at Week 123TC CL/F at Week 24
Normal Nutrition/Mild Malnutrition Cohort8.46.88.8
Severe Malnutrition Cohort8.79.57.5

[back to top]

Plasma Clearance of Lopinavir

Steady-state plasma clearance (CL/F) of LPV (NCT01818258)
Timeframe: 0, 1, 2, 4, 8, and 12 hours post-dose on 1, 12, and 24 weeks following study entry

,
InterventionL/hours (Geometric Mean)
LPV CL/F at Week 1LPV CL/F at Week 12LPV CL/F at Week 24
Normal Nutrition/Mild Malnutrition Cohort2.01.61.7
Severe Malnutrition Cohort2.22.32.1

[back to top]

Plasma Clearance of Ritonavir

Steady-state plasma clearance (CL/F) of RTV (NCT01818258)
Timeframe: 0, 1, 2, 4, 8, and 12 hours post-dose on 1, 12, and 24 weeks following study entry

,
InterventionL/hours (Geometric Mean)
RTV CL/F at Week 1RTV CL/F at Week 12RTV CL/F at Week 24
Normal Nutrition/Mild Malnutrition Cohort15.811.211.5
Severe Malnutrition Cohort16.917.314.8

[back to top]

Plasma Clearance of Zidovudine

Steady-state plasma clearance (CL/F) of Zidovudine (ZDV) (NCT01818258)
Timeframe: 0, 1, 2, 4, 8, and 12 hours post-dose on 1, 12, and 24 weeks following study entry

,
InterventionL/hours (Geometric Mean)
ZDV CL/F at Week 1ZDV CL/F at Week 12ZDV CL/F at Week 24
Normal Nutrition/Mild Malnutrition Cohort58.381.864.0
Severe Malnutrition Cohort34.848.840.8

[back to top]

Steady-state Lamivudine Area Under the Curve

Steady-state area under the curve (AUC) of Lamivudine (3TC) (NCT01818258)
Timeframe: 0, 1, 2, 4, 8, and 12 hours post-dose on 1, 12, and 24 weeks following study entry

,
Interventionng*hours/mL (Geometric Mean)
3TC AUC at Week 13TC AUC at Week 123TC AUC at Week 24
Normal Nutrition/Mild Malnutrition Cohort5,520.27,233.05,849.2
Severe Malnutrition Cohort4,245.04,365.56,359.0

[back to top]

Steady-state Ritonavir Area Under the Curve

Steady-state area under the curve (AUC) for Ritonavir (RTV) (NCT01818258)
Timeframe: 0, 1, 2, 4, 8, and 12 hours post-dose on 1, 12, and 24 weeks following study entry

,
Interventionug*hours/mL (Geometric Mean)
RTV AUC Week 1RTV AUC Week 12RTV AUC Week 24
Normal Nutrition/Mild Malnutrition Cohort2.13.03.0
Severe Malnutrition Cohort1.61.82.3

[back to top]

Steady-state Zidovudine Area Under the Curve

Steady-state area under the curve (AUC) of zidovudine (ZDV) (NCT01818258)
Timeframe: 0, 1, 2, 4, 8, and 12 hours post-dose on 1, 12, and 24 weeks following study entry

,
Interventionng*hours/mL (Geometric Mean)
ZDV AUC at Week 1ZDV AUC at Week 12ZDV AUC at Week 24
Normal Nutrition/Mild Malnutrition Cohort1,774.01,335.71,609.3
Severe Malnutrition Cohort2,261.01,826.02,449.7

[back to top]

HIV Viral Load <400 Copies/mL

Count (%) of participants with plasma HIV RNA viral load <400 copies/mL (NCT01818258)
Timeframe: Baseline and weeks 12, 24, and 48

,
InterventionParticipants (Count of Participants)
BaselineWeek 12Week 24Week 48
Normal Nutrition/Mild Malnutrition Cohort2141818
Severe Malnutrition Cohort281111

[back to top] [back to top]

Grade 3 or Higher Adverse Events Through 24 Weeks

Number (percent) of participants with at least one grade 3 or higher adverse event (AE) regardless of the relationship to study drugs. (NCT01818258)
Timeframe: From week 0 to week 24

InterventionParticipants (Count of Participants)
Severe Malnutrition Cohort13
Normal Nutrition/Mild Malnutrition Cohort10

[back to top]

Steady-state Lopinavir Area Under the Curve

Steady-state area under the curve (AUC) for Lopinavir (LPV) (NCT01818258)
Timeframe: 0, 1, 2, 4, 8, and 12 hours post-dose on 1, 12, and 24 weeks following study entry

,
Interventionug*hours/mL (Geometric Mean)
Week 1Week 12Week 24
Normal Nutrition/Mild Malnutrition Cohort64.883.479.4
Severe Malnutrition Cohort49.853.064.6

[back to top]

Change in Percentage of Total Artery Diameter

computerized axial tomography angiography of the coronary arteries (CT-angio) before and after 12-months of Darunavir therapy (NCT01869634)
Timeframe: Baseline, 12 months

Intervention% of total artery diameter (Median)
HIV Positive Naive to ART57
Normal Control Volunteers53

[back to top]

Change in Systemic Immune Activation

Change in systemic immune activation, as measured by change in plasma cytokine levels (IL-6). (NCT01869634)
Timeframe: Baseline, 12 months

Interventionpg/ml (Median)
HIV Positive Naive to ART1.74
Normal Control Volunteers0.66

[back to top]

Number of CD4+ T-cells in the Lamina Propria/mm2 Before and After 12 Months of Therapy Compared to Age-matched Control Volunteers Without HIV

CD4+ T-cells in the lamina propria/mm2 before and after 12 months of therapy compared to age-matched control volunteers without HIV. (NCT01869634)
Timeframe: Baseline, 12 months

Interventioncells / mm^2 (Median)
Entry
Normal Control Volunteers478

[back to top]

Number of CD4+ T-cells in the Lamina Propria/mm2 Before and After 12 Months of Therapy Compared to Age-matched Control Volunteers Without HIV

CD4+ T-cells in the lamina propria/mm2 before and after 12 months of therapy compared to age-matched control volunteers without HIV. (NCT01869634)
Timeframe: Baseline, 12 months

Interventioncells / mm^2 (Median)
Entry12-months after ART
HIV Positive Naive to ART80213

[back to top]

RTV PK Parameter CLss/F Determined Based on RTV Levels From Individual Participants Enrolled in Arm C

This evaluates the effect of NuvaRing on the PK parameter CLss/F of RTV before NuvaRing placement (at study day 0) and three weeks later (on study day 21), prior to NuvaRing removal. CLss/F defines apparent oral clearance. (NCT01903031)
Timeframe: Intensive RTV PK samples at pre-dose, 1, 3, 4, 5, and 8 hours post-dose on study day 0 (before vaginal ring placement) and on study day 21 (3 weeks after vaginal ring placement)

Interventionhour (Median)
CLss/F day 0CLss/F day 21
NuvaRing With ATV/r Plus TDF and ≥1 NRTIs9.313.9

[back to top]

Ritonavir (RTV) PK Parameter AUC(0-24h) Calculated Based on Intensive RTV PK Samples Obtained From Individual Participants Enrolled in Arm C

This evaluates the effect of NuvaRing on the PK parameter AUC(0-24h) of RTV before NuvaRing placement (at study day 0) and three weeks later (on study day 21), prior to NuvaRing removal. AUC(0-24h) defines area under the concentration-time curve over the period of 24 hours (pre-dose concentration was used to impute concentration at 24h). (NCT01903031)
Timeframe: Intensive RTV PK samples at pre-dose, 1, 3, 4, 5, and 8 hours post-dose on study day 0 (before vaginal ring placement) and on study day 21 (3 weeks after vaginal ring placement)

Interventionh*ng/mL (Median)
AUC0-24h day 0AUC0-24h day 21
NuvaRing With ATV/r Plus TDF and ≥1 NRTIs10740.07210.7

[back to top]

EFV PK Parameter Maximum Plasma Concentration (Cmax) Determined Based on EFV Levels From Individual Participants Enrolled in Arm B

This evaluates the effect of NuvaRing on the EFV PK parameter Cmax obtained from both sampling periods, before NuvaRing placement (at study day 0) and three weeks later (on study day 21), prior to NuvaRing removal. Cmax defines maximum concentration observed within the first 8 hours of the 24 hour dosing interval. (NCT01903031)
Timeframe: Intensive EFV PK samples at pre-dose, 1, 3, 4, 5, and 8 hours post-dose on study day 0 (before vaginal ring placement) and on study day 21 (3 weeks after vaginal ring placement).

Interventionng/mL (Median)
Cmax day 0Cmax day 21
NuvaRing With EFV Plus ≥2 NRTIs4541.03786.0

[back to top]

Proportion of Participants With Progesterone Levels Greater Than 5 ng/mL.

This evaluates alterations in progesterone levels due to the potential PK interaction between NuvaRing and the ARVs EFV and ATV/r by examining progesterone levels at study days 0 (before vaginal ring placement), 7, 14, and 21 (before vaginal ring removal), and study day 28, without regard to menstrual cycle status at study entry. (NCT01903031)
Timeframe: Study days 0, 7, 14, 21 and 28

,,
Interventionproportion of participants (Number)
Proportion with progesterone >5 at day 0Proportion with progesterone >5 at day 7Proportion with progesterone >5 at day 14Proportion with progesterone >5 at day 21Proportion with progesterone >5 at day 28
NuvaRing and no ART0.080.080.000.000.00
NuvaRing With ATV/r Plus TDF and ≥1 NRTIs0.250.080.000.000.00
NuvaRing With EFV Plus ≥2 NRTIs0.040.240.040.000.00

[back to top]

EFV PK Parameter Clearance (CLss/F) Determined Based on EFV Levels From Individual Participants Enrolled in Arm B

This evaluates the effect of NuvaRing on the EFV PK parameter CLss/F obtained from both sampling periods, before NuvaRing placement (at study day 0) and three weeks later (on study day 21), prior to NuvaRing removal. CLss/F defines apparent oral clearance (NCT01903031)
Timeframe: Intensive EFV PK samples at pre-dose, 1, 3, 4, 5, and 8 hours post-dose on study day 0 (before vaginal ring placement) and on study day 21 (3 weeks after vaginal ring placement).

InterventionL/h (Median)
CLss/F day 0CLss/F day 21
NuvaRing With EFV Plus ≥2 NRTIs8.710.4

[back to top]

EFV PK Parameter Area Under the Concentration-Time Curve (AUC0-24hours) Calculated Based on Intensive EFV PK Samples Obtained From Individual Participants Enrolled in Arm B

This evaluates the effect of NuvaRing on the PK parameter AUC(0-24h) of EFV before NuvaRing placement (at study day 0) and three weeks later (on study day 21), prior to NuvaRing removal. AUC(0-24h) defines area under the concentration-time curve over the period of 24 hours (pre-dose concentration was used to impute concentration at 24h). (NCT01903031)
Timeframe: Intensive EFV PK samples at pre-dose, 1, 3, 4, 5, and 8 hours post-dose on study day 0 (before vaginal ring placement) and on study day 21 (3 weeks after vaginal ring placement).

Interventionh*ng/mL (Median)
AUC0-24h day 0AUC0-24h day 21
NuvaRing With EFV Plus ≥2 NRTIs68949.157795.9

[back to top]

ATV PK Parameter Time to Cmax (Tmax) Determined Based on ATV Levels From Individual Participants Enrolled in Arm C

This evaluates the effect of NuvaRing on the ATV PK parameter Tmax obtained from both sampling periods, before NuvaRing placement (at study day 0) and three weeks later (on study day 21), prior to NuvaRing removal. Tmax defines time to maximum concentration since dose is initiated. (NCT01903031)
Timeframe: Intensive ATV PK samples at pre-dose, 1, 3, 4, 5, and 8 hours post-dose on study day 0 (before vaginal ring placement) and on study day 21 (3 weeks after vaginal ring placement).

Interventionhour (Median)
Tmax day 0Tmax day 21
NuvaRing With ATV/r Plus TDF ≥1 NRTIs2.93.0

[back to top]

ATV PK Parameter Cmin Determined Based on ATV Levels From Individual Participants Enrolled in Arm C

This evaluates the effect of NuvaRing on the ATV PK parameter Cmin obtained from both sampling periods, before NuvaRing placement (at study day 0) and three weeks later (on study day 21), prior to NuvaRing removal. Cmin defines minimum concentration observed within the first 8 hours of the 24 hour dosing interval. (NCT01903031)
Timeframe: Intensive ATV PK samples at pre-dose, 1, 3, 4, 5, and 8 hours post-dose on study day 0 (before vaginal ring placement) and on study day 21 (3 weeks after vaginal ring placement).

Interventionng/mL (Median)
Cmin day 0Cmin day 21
NuvaRing With ATV/r Plus TDF ≥1 NRTIs796.7599.4

[back to top]

Etonogestrel Concentrations Obtained on Study Days 7 and 14

This evaluates the effect of EFV and ATV/r on etonogestrel by measuring etonogestrel concentrations on all three study arms 7 and 14 days after NuvaRing administration. The assay lower limit of quantification for etonogestrel was 250 pg/mL; values < 250 were assigned a value of half the lower limit (ie, 125 pg/mL). (NCT01903031)
Timeframe: Study days 7 and 14

,,
Interventionpg/mL (Median)
Concentration at Day 7Concentration at Day 14
NuvaRing and no ART1970.002070.00
NuvaRing With ATV/r Plus TDF and ≥1 NRTIs3250.003530.00
NuvaRing With EFV Plus ≥2 NRTIs427.00437.00

[back to top]

ATV PK Parameter CLss/F Determined Based on ATV Levels From Individual Participants Enrolled in Arm C

This evaluates the effect of NuvaRing on the ATV PK parameter CLss/F obtained from both sampling periods, before NuvaRing placement (at study day 0) and three weeks later (on study day 21), prior to NuvaRing removal. CLss/f defines apparent oral clearance. (NCT01903031)
Timeframe: Intensive ATV PK samples at pre-dose, 1, 3, 4, 5, and 8 hours post-dose on study day 0 (before vaginal ring placement) and on study day 21 (3 weeks after vaginal ring placement).

InterventionL/h (Median)
CLss/F day 0CLss/F day 21
NuvaRing With ATV/r Plus TDF ≥1 NRTIs6.88.2

[back to top]

Ethinyl Estradiol Concentrations Obtained on Study Days 7 and 14.

This evaluates the effect of EFV and ATV/r on ethinyl estradiol by measuring ethinyl estradiol concentrations on all three study arms 7 and 14 days after NuvaRing administration. The assay lower limit of quantification for ethinyl estradiol was 5 pg/mL; values < 5 were assigned a value of half the lower limit (ie, 2.5 pg/mL). (NCT01903031)
Timeframe: Study days 7 and 14

,,
Interventionpg/mL (Median)
Concentration at Day 7Concentration at Day 14
NuvaRing and no ART18.0519.70
NuvaRing With ATV/r Plus TDF and ≥1 NRTIs15.7016.55
NuvaRing With EFV Plus ≥2 NRTIs9.9810.50

[back to top]

EFV PK Parameter Minimum Plasma Concentration (Cmin) Determined Based on EFV Levels From Individual Participants Enrolled in Arm B

This evaluates the effect of NuvaRing on the EFV PK parameter Cmin obtained from both sampling periods, before NuvaRing placement (at study day 0) and three weeks later (on study day 21), prior to NuvaRing removal. Cmin defines minimum concentration observed within the first 8 hours of the 24 hour dosing interval. (NCT01903031)
Timeframe: Intensive EFV PK samples at pre-dose, 1, 3, 4, 5, and 8 hours post-dose on study day 0 (before vaginal ring placement) and on study day 21 (3 weeks after vaginal ring placement).

Interventionng/mL (Median)
Cmin day 0Cmin day 21
NuvaRing With EFV Plus ≥2 NRTIs2121.51766.0

[back to top]

ATV PK Parameter Cmax Determined Based on ATV Levels From Individual Participants Enrolled in Arm C

This evaluates the effect of NuvaRing on the ATV PK parameter Cmax obtained from both sampling periods, before NuvaRing placement (at study day 0) and three weeks later (on study day 21), prior to NuvaRing removal. Cmax defines maximum concentration observed within the first 8 hours of the 24 hour dosing interval. (NCT01903031)
Timeframe: Intensive ATV PK samples at pre-dose, 1, 3, 4, 5, and 8 hours post-dose on study day 0 (before vaginal ring placement) and on study day 21 (3 weeks after vaginal ring placement).

Interventionng/mL (Median)
Cmax day 0Cmax day 21
NuvaRing With ATV/r Plus TDF ≥1 NRTIs4291.03583.0

[back to top]

Proportion of Participants With Plasma HIV-1 RNA Levels <40 Copies/mL

This evaluates the short-term impact of Nuvaring on virologic suppression in participants who have been administered Nuvaring alone or together with EFV or ATV/r by measuring proportion of participants with plasma HIV-1 RNA levels <40 copies/mL at study day 0 (before vaginal ring placement) and study day 21 (three weeks after vaginal ring placement). An FDA-approved HIV-1 RNA assay was required. (NCT01903031)
Timeframe: Study day 0 and study day 21

,,
Interventionproportion of participants (Number)
Proportion with HIV-1 RNA <40 copies/mL at day 0Proportion with HIV-1 RNA <40 copies/mL at day 21
NuvaRing and no ART0.220.17
NuvaRing With ATV/r Plus TDF and ≥1 NRTIs0.890.85
NuvaRing With EFV Plus ≥2 NRTIs0.930.85

[back to top]

RTV PK Parameter Tmax Determined Based on RTV Levels From Individual Participants Enrolled in Arm C

This evaluates the effect of NuvaRing on the PK parameter Tmax of RTV before NuvaRing placement (at study day 0) and three weeks later (on study day 21), prior to NuvaRing removal. Tmax defines time to maximum concentration since dose is initiated. (NCT01903031)
Timeframe: Intensive RTV PK samples at pre-dose, 1, 3, 4, 5, and 8 hours post-dose on study day 0 (before vaginal ring placement) and on study day 21 (3 weeks after vaginal ring placement)

Interventionhour (Median)
Tmax day 0Tmax day 21
NuvaRing With ATV/r Plus TDF and ≥1 NRTIs3.03.0

[back to top]

RTV PK Parameter Cmin Determined Based on RTV Levels From Individual Participants Enrolled in Arm C

This evaluates the effect of NuvaRing on the PK parameter Cmin of RTV before NuvaRing placement (at study day 0) and three weeks later (on study day 21), prior to NuvaRing removal. Cmin defines minimum concentration observed within the first 8 hours of the 24 hour dosing interval. (NCT01903031)
Timeframe: Intensive RTV PK samples at pre-dose, 1, 3, 4, 5, and 8 hours post-dose on study day 0 (before vaginal ring placement) and on study day 21 (3 weeks after vaginal ring placement)

Interventionng/mL (Median)
Cmin day 0Cmin day 21
NuvaRing With ATV/r Plus TDF and ≥1 NRTIs70.051.9

[back to top]

Ethinyl Estradiol Concentrations at Study Day 21

This evaluates the effect of EFV and ATV/r on ethinyl estradiol by measuring ethinyl estradiol concentrations on all three study arms 21 days after NuvaRing administration. The PK blood sample for measurement of ethinyl estradiol on study day 21 was taken before the NuvaRing was removed. The assay lower limit of quantification for ethinyl estradiol was 5 pg/mL ; values < 5 were assigned a value of half the lower limit (ie, 2.5 pg/mL). (NCT01903031)
Timeframe: Day 21

Interventionpg/mL (Median)
NuvaRing and no ART21.30
NuvaRing With EFV Plus ≥2 NRTIs11.40
NuvaRing With ATV/r Plus TDF and ≥1 NRTIs16.05

[back to top]

Etonogestrel Concentrations at Study Day 21

This evaluates the effect of EFV and ATV/r on etonogestrel by measuring etonogestrel concentrations on all three study arms 21 days after NuvaRing administration. The pharmacokinetic (PK) blood sample for measurement of etonogestrel on study day 21 was taken before the NuvaRing was removed. The assay lower limit of quantification for etonogestrel was 250 pg/mL; values < 250 were assigned a value of half the lower limit (ie, 125 pg/mL). (NCT01903031)
Timeframe: Day 21

Interventionpg/mL (Median)
NuvaRing and no ART1860.00
NuvaRing With EFV Plus ≥2 NRTIs429.00
NuvaRing With ATV/r Plus TDF and ≥1 NRTIs3290.00

[back to top] [back to top]

ATV PK Parameter AUC(0-24h) Calculated Based on Intensive Atazanavir (ATV) PK Samples Obtained From Individual Participants Enrolled in Arm C

This evaluates the effect of NuvaRing on the PK parameter AUC(0-24h) of ATV before NuvaRing placement (at study day 0) and three weeks later (on study day 21), prior to NuvaRing removal. AUC(0-24h) defines area under the concentration-time curve over the period of 24 hours (pre-dose concentration was used to impute concentration at 24h). (NCT01903031)
Timeframe: Intensive ATV PK samples at pre-dose, 1, 3, 4, 5, and 8 hours post-dose on study day 0 (before vaginal ring placement) and on study day 21 (3 weeks after vaginal ring placement)

Interventionh*ng/mL (Median)
AUC0-24h day 0AUC0-24h day 21
NuvaRing With ATV/r Plus TDF and ≥1 NRTIs44313.736764.7

[back to top]

RTV PK Parameter Cmax Determined Based on RTV Levels From Individual Participants Enrolled in Arm C

This evaluates the effect of NuvaRing on the PK parameter Cmax of RTV before NuvaRing placement (at study day 0) and three weeks later (on study day 21), prior to NuvaRing removal. Cmax defines maximum concentration observed within the first 8 hours of the 24 hour dosing interval. (NCT01903031)
Timeframe: Intensive RTV PK samples at pre-dose, 1, 3, 4, 5, and 8 hours post-dose on study day 0 (before vaginal ring placement) and on study day 21 (3 weeks after vaginal ring placement)

Interventionng/mL (Median)
Cmax day 0Cmax day 21
NuvaRing With ATV/r Plus TDF and ≥1 NRTIs1437.01063.0

[back to top]

Number of Participants With Post-Baseline HIV-1 Disease Progression-Randomized Phase

Number of participants with post-Baseline HIV-1disease progression were assessed during study period. The CDC Classification System for HIV Infection is the medical classification system used by the United States Centers for Disease Control and Prevention (CDC) to classify HIV disease and infection. The clinical categories of HIV infection are defined as follows: Category A: Mildly symptomatic, Category B: Moderately symptomatic, Category C: Severely symptomatic. The Baseline value was defined as the latest pre-dose assessment (Day 1) value. Only those participants available at the specified time points were analyzed. Different participants may have been analyzed at different time points, so the overall number of participants analyzed reflects everyone in the ITT population. (NCT01910402)
Timeframe: Up to week 48

,
InterventionParticipants (Count of Participants)
CDC Class A to CDC Class CCDC Class B to CDC Class CCDC Class C to new CDC Class CCDC Class A, B or C to Death
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase4201
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase5101

[back to top]

Number of Participants With Treatment Emergent Resistances for ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200mg QD (Randomized Phase)

Number of participants, who meet confirmed virologic withdrawal criteria, with treatment emergent genotypic resistance to integrase strand transfer inhibitor (INSTI), Non-nucleoside reverse transcriptase inhibitor (NNRTI), nucleoside reverse transcriptase inhibitor (NRTI), protease inhibitors (PI) will be summarized. The Baseline value was defined as the latest pre-dose assessment (Day 1) value. On-treatment Genotypic Resistance Population comprised of all participants in the ITTE population with available On-treatment genotypic resistance data at the time confirmed virologic withdrawal criterion was met. (NCT01910402)
Timeframe: Up to week 48

InterventionParticipants (Count of Participants)
INSTI; n= 3NNRTI; n=4NRTI; n=4PI; n=4
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200mg QD-Randomized Phase1010

[back to top]

Number of Participants With Treatment Emergent Resistances for DTG 50 mg/ABC 600 mg/3TC 300 mg QD (Randomized + Continuation Phase)

Number of participants, who meet confirmed virologic withdrawal criteria, with treatment emergent genotypic resistance to integrase strand transfer inhibitor (INSTI), Non-nucleoside reverse transcriptase inhibitor (NNRTI), nucleoside reverse transcriptase inhibitor (NRTI), protease inhibitors (PI) will be summarized. The Baseline value was defined as the latest pre-dose assessment (Day 1) value. On-treatment Genotypic Resistance Population comprised of all participants in the ITTE population with available On-treatment genotypic resistance data at the time confirmed virologic withdrawal criterion was met. (NCT01910402)
Timeframe: Up to week 432

InterventionParticipants (Count of Participants)
INSTI; n= 6NNRTI; n=8NRTI; n=8PI; n=8
DTG 50 mg/ABC 600 mg/3TC 300 mg QD0110

[back to top]

Percentage of Participants With Plasma HIV-1 RNA <50 and <400 c/mL Over Time-Randomized Phase

Percentage of participants with plasma HIV-1 RNA <50 and <400 c/mL were assessed at Baseline, Weeks 4, 12, 24 , 36 and 48 using the Snapshot algorithm (Missing, Switch or Discontinuation = Failure). The Baseline value was defined as the latest pre-dose assessment (Day 1) value. Percentage values are rounded off. (NCT01910402)
Timeframe: Baseline (Day 1), Week 4, Week 12, Week 24, Week 36 and Week 48

,
InterventionPercentage of participants (Number)
HIV-1 RNA <50 c/mL, Baseline (Day 1)HIV-1 RNA <50 c/mL, Week 4HIV-1 RNA <50 c/mL, Week 12HIV-1 RNA <50 c/mL, Week 24HIV-1 RNA <50 c/mL, Week 36HIV-1 RNA <50 c/mL, Week 48HIV-1 RNA <400 c/mL, Baseline (Day 1)HIV-1 RNA <400 c/mL, Week 4HIV-1 RNA <400 c/mL, Week 12HIV-1 RNA <400 c/mL, Week 24HIV-1 RNA <400 c/mL, Week 36HIV-1 RNA <400 c/mL, Week 48
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase0134977777115484828176
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase0648185858219091888683

[back to top]

Percentage of Participants With Plasma HIV-1 RNA <50 c/mL in Continuation Phase

Plasma samples were collected for quantitative analysis of HIV-1 RNA. Percentage of participants with plasma HIV-1 RNA <50 c/mL were reported. Percentage values are rounded off. (NCT01910402)
Timeframe: Week 96 and Week 432

InterventionPercentage of participants (Number)
Week 96, n=99Week 432, n=3
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Continuation Phase100100

[back to top]

Percentage of Participants With Plasma HIV-1 RNA <50 Copies/mL at Week 48 by Subgroups

Percentage of participants with plasma HIV-1 RNA <50 copies/mL at Week 48 by subgroups (age, race, country, Baseline plasma HIV-1 RNA [BPHR], Baseline CD4+ cell count [BCCC], Baseline Centers for Disease Control and Prevention [CDC] category and HIV-1 subtype) were assessed using the Snapshot algorithm (Missing, Switch or Discontinuation = Failure). Analysis was performed using a stratified analysis with CMH weights, adjusting for Baseline plasma HIV-1 RNA ( =100,000 c/mL) and CD4+ cell count (=<350 cells/mm^3 or >350 cells/mm^3). Different participants may have been analyzed at different time points, so the overall number of participants analyzed reflects everyone in the ITT population. Percentage values are rounded off. (NCT01910402)
Timeframe: Week 48

InterventionPercentage of participants (Number)
Age, <50 Years, n=212, 212Age, >=50 Years, n=36, 35Race, White, n=115, 107Race, Non-White, n=133,140Race, African-American/African Heritage, n=102,108Non-African-American/African Heritage, n=146, 139BPHR, <1000, n=5, 10BPHR, 1000 to <10,000, n=66, 62BPHR, 10,000 to <50,000, n=83, 81BPHR, 50,000 to <=100,000, n=25, 28BPHR, >100,000, n=69, 66BCCC, <200, n=64, 49BCCC, >=200, n=184, 198BCCC, <50, n=9, 15BCCC, 50 to <200, n=55, 34BCCC, 200 to <350, n=66, 74BCCC, 350 to <500, n=56, 65BCCC, >=500, n=62, 59CDC category, A, n=210, 208CDC category, B, n=27, 30CDC category, C, n=11, 9HIV-1 subtype: B vs Non-B, B, n=95, 111HIV-1 subtype: B vs Non-B, non-B, n=140, 131Argentina, n=24, 20Canada, n=11, 9France, n=7, 8Italy, n=17, 11Mexico, n=6, 5Portugal, n=4, 5Russia, n=28, 22South Africa, n=33, 33Spain, n=23, 31Thailand, n=19, 21USA, n=62, 69United Kingdom, n=14, 11
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase809286787488608384808081826784897977818191808492911008810075896770957493

[back to top]

Percentage of Participants With Plasma HIV-1 RNA <50 Copies/mL at Week 48 by Subgroups

Percentage of participants with plasma HIV-1 RNA <50 copies/mL at Week 48 by subgroups (age, race, country, Baseline plasma HIV-1 RNA [BPHR], Baseline CD4+ cell count [BCCC], Baseline Centers for Disease Control and Prevention [CDC] category and HIV-1 subtype) were assessed using the Snapshot algorithm (Missing, Switch or Discontinuation = Failure). Analysis was performed using a stratified analysis with CMH weights, adjusting for Baseline plasma HIV-1 RNA ( =100,000 c/mL) and CD4+ cell count (=<350 cells/mm^3 or >350 cells/mm^3). Different participants may have been analyzed at different time points, so the overall number of participants analyzed reflects everyone in the ITT population. Percentage values are rounded off. (NCT01910402)
Timeframe: Week 48

InterventionPercentage of participants (Number)
Age, <50 Years, n=212, 212Age, >=50 Years, n=36, 35Race, White, n=115, 107Race, Non-White, n=133,140Race, African-American/African Heritage, n=102,108Non-African-American/African Heritage, n=146, 139BPHR, <1000, n=5, 10BPHR, 1000 to <10,000, n=66, 62BPHR, 10,000 to <50,000, n=83, 81BPHR, 50,000 to <=100,000, n=25, 28BPHR, >100,000, n=69, 66BCCC, <200, n=64, 49BCCC, >=200, n=184, 198BCCC, <50, n=9, 15BCCC, 50 to <200, n=55, 34BCCC, 200 to <350, n=66, 74BCCC, 350 to <500, n=56, 65BCCC, >=500, n=62, 59CDC category, A, n=210, 208CDC category, B, n=27, 30CDC category, C, n=11, 9HIV-1 subtype: B vs Non-B, B, n=95, 111HIV-1 subtype: B vs Non-B, non-B, n=140, 131Argentina, n=24, 20Canada, n=11, 9France, n=7, 8Italy, n=17, 11Mexico, n=6, 5Portugal, n=4, 5Puerto Rico, n=0, 2Russia, n=28, 22South Africa, n=33, 33Spain, n=23, 31Thailand, n=19, 21USA, n=62, 69United Kingdom, n=14, 11
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase7174806467758077746464697260747374687177566973808975646060100827677526764

[back to top]

Change From Baseline in Plasma HIV-1 RNA at Indicated Time Points-Continuation Phase

Change from the Baseline in plasma HIV-1 RNA were assessed at indicated time points. The Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1), Week 96 and Week 432

InterventionLog10 copies/mL (Mean)
Week 96, n=99Week 432, n=3
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Continuation Phase-2.911-3.107

[back to top]

Change From Baseline in TC/HDL Ratio at Week 48

Change from Baseline in mean total cholesterol (TC)/HDL ratio is summarized at Week 48. The Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. Adjusted mean is the estimated mean change from Baseline in fasted TC/HDL at Week 48 in each arm calculated from a model adjusted for the following covariates: treatment, Baseline plasma HIV-1 RNA, Baseline CD4+ cell count, age and triglycerides/HDL at Baseline. Participants on lipid lowering therapy at Baseline were excluded from analysis. Measurements collected after a participant initiates lipid lowering therapy were set to missing. Missing values were imputed using multiple imputation under a multivariate normal model adjusting for Baseline plasma HIV-1 RNA, Baseline CD4+ cell count, fasted triglycerides and TC/HDL ratio at Baseline, Week 12 and Week 36. (NCT01910402)
Timeframe: Baseline (Day 1) and Week 48

InterventionRatio (Least Squares Mean)
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase-0.264
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase-0.158

[back to top]

Change From Baseline in Triglycerides at Week 48

Change from Baseline in mean triglycerides is summarized at Week 48. The Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. Adjusted mean is the estimated mean change from Baseline in fasted triglycerides at Week 48 in each arm calculated from a model adjusted for the following covariates: treatment, Baseline plasma HIV-1 RNA, Baseline CD4+ cell count, age and triglycerides at Baseline. Participants on lipid lowering therapy at Baseline were excluded from analysis. Measurements collected after a participant initiates lipid lowering therapy were set to missing. Missing values were imputed using multiple imputation under a multivariate normal model adjusting for Baseline plasma HIV-1 RNA, Baseline CD4+ cell count, fasted triglycerides and TC/HDL ratio at Baseline, Week 12 and Week 36. (NCT01910402)
Timeframe: Baseline (Day 1) and Week 48

InterventionMillimoles per liter (Least Squares Mean)
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase0.045
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase0.070

[back to top]

Number of Participants Who Withdrew From Treatment Due to AEs-Continuation Phase

An AE is defined as any untoward medical occurrence in a participant temporally associated with the use of a medicinal product (MP), whether or not considered related to the MP. An AE can therefore be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease (new or exacerbated) temporally associated with the use of an MP. An SAE is defined as any untoward medical occurrence that, at any dose, results in death, is life threatening, requires hospitalization or prolongation of existing hospitalization, results in disability/incapacity, is a congenital anomaly/birth defect, is an important medical event that jeopardizes the participant or may require medical or surgical intervention to prevent one of the other outcomes listed in the above definition, or is associated with liver injury and impaired liver function. (NCT01910402)
Timeframe: From Weeks 48 to 432

InterventionParticipants (Count of Participants)
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Continuation Phase4

[back to top]

Number of Participants Who Withdrew From Treatment Due to AEs-Randomized Phase

An AE is defined as any untoward medical occurrence in a participant temporally associated with the use of a medicinal product (MP), whether or not considered related to the MP. An AE can therefore be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease (new or exacerbated) temporally associated with the use of an MP. An SAE is defined as any untoward medical occurrence that, at any dose, results in death, is life threatening, requires hospitalization or prolongation of existing hospitalization, results in disability/incapacity, is a congenital anomaly/birth defect, is an important medical event that jeopardizes the participant or may require medical or surgical intervention to prevent one of the other outcomes listed in the above definition, or is associated with liver injury and impaired liver function. (NCT01910402)
Timeframe: Up to Week 48

InterventionParticipants (Count of Participants)
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase10
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase17

[back to top]

Percentage of Participants With Plasma HIV-1 RNA <50 Copies/mL at Week 48

Percentage of participants with plasma human immunodeficiency virus type 1(HIV-1) ribonucleic acid (RNA) <50 copies per milliliter (c/mL) were assessed at Week 48 using the Snapshot algorithm. Analysis was performed using a stratified analysis with Cochran-Mantel-Haenszel (CMH) weights, adjusting for Baseline plasma HIV-1 RNA ( =100,000 c/mL) and CD4+ cell count (=<350 cells per millimeter cube (cells/mm^3) or >350 cells/mm^3). Intent-to-Treat Exposed (ITT-E) Population comprised of all randomized participants who received at least one dose of study medication. Percentage values are rounded off. (NCT01910402)
Timeframe: Week 48

InterventionPercentage of participants (Number)
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase82
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase71

[back to top]

Absolute Values in CD4+ Cell Count at Indicated Timepoints-Continuation Phase

Absolute values in CD4+ cell count were assessed at indicated time points. (NCT01910402)
Timeframe: Week 96 and Week 432

InterventionCells per cubic millimeter (Mean)
Week 96, n=99Week 432, n=3
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Continuation Phase635.3553.0

[back to top]

Absolute Values in CD4+ Cell Count at Indicated Timepoints-Randomized Phase

Absolute values in CD4+ cell count were assessed at indicated time points. The Baseline value was defined as the latest pre-dose assessment (Day 1) value. (NCT01910402)
Timeframe: Baseline (Day 1), Week 4, Week 12, Week 24, Week 36 and Week 48

,
InterventionCells per cubic millimeter (Mean)
Baseline (Day 1), n=248, 247Week 4, n=245, 237Week 12, n=236, 224Week 24, n=226, 210Week 36, n=219, 204Week 48, n=208, 191
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase380.3455.1506.2542.5569.2608.5
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase369.7465.0509.5563.8592.8608.8

[back to top]

Absolute Values in Plasma HIV-1 RNA at Indicated Time Points-Continuation Phase

Absolute Values in plasma HIV-1 RNA were assessed at indicated time points. (NCT01910402)
Timeframe: Week 96 and Week 432

InterventionLog10 copies/mL (Mean)
Week 96, n=99Week 432, n=3
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Continuation Phase1.5911.590

[back to top]

Absolute Values in Plasma HIV-1 RNA at Indicated Time Points-Randomized Phase

Absolute Values in plasma HIV-1 RNA were assessed at indicated time points. The Baseline value was defined as the latest pre-dose assessment (Day 1) value. (NCT01910402)
Timeframe: Baseline (Day 1), Week 4, Week 12, Week 24, Week 36 and Week 48

,
InterventionLog10 copies/mL (Mean)
Baseline (Day 1), n=248, 247Week 4, n=245, 238Week 12, n=236, 226Week 24, n=225, 212Week 36, n=221, 204Week 48, n=207, 192
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase4.4412.5161.9081.7101.6581.657
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase4.4811.8951.7481.7241.6661.619

[back to top]

Bone Specific Alkaline Phosphatase, Osteocalcin, Procollagen 1 N-terminal Propeptide, Type 1 Collagen C-Telopeptide, Vitamin D Ratio of Week 48 Results Over Baseline

Bone markers were assessed at indicated timepoints. Bone specific alkaline phosphatase (BSAP), osteocalcin and procollagen 1 N-terminal propeptide (PTP), Type 1 Collagen C-Telopeptide, vitamin D ratio of Week 48 results over Baseline is calculated. Bone biomarkers were analyzed based on log transformed data. Estimates of adjusted mean and difference were calculated from an Analysis of covariance (ANCOVA) model adjusting for age, baseline viral load Baseline CD4+ cell count, Baseline biomarker level, body mass index category, smoking status and baseline Vitamin D use. Adjusted mean of log-transformed change from Baseline are transformed back to Week 48/Baseline ratio for each treatment group. Adjusted difference of log-transformed change from Baseline between treatment groups is transformed back to the ratio of Week 48/Baseline ratio in DTG/ABC/3TC FDC to ATV+RTV+TDF/FTC FDC. (NCT01910402)
Timeframe: Baseline (Day 1) and Week 48

,
InterventionRatio (Number)
BSAP, n=202, 183PTP, n=202, 184Osteocalcin, n=194, 178Type 1 Collagen C-Telopeptide, n=202, 184Vitamin D, n=206, 186
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase1.6291.7522.0391.9181.158
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase1.1881.2141.2821.2570.987

[back to top]

Change From Baseline at Week 48 in SF-12 Total Score, MCS and PCS

The 12-Item Short Form Health Survey (SF-12) is 12 item abbreviated form of SF-36 survey. SF-12 questions make up 8 scales: Physical Functioning, Role Physical, Bodily Pain, General Health, Vitality, Social Functioning, Role Emotional, Mental Health. SF-12 is a self-reported outcome measure assessing psychological wellness and the impact of health on an individual's everyday life. SF-12 total score ranges from 20 to 60 and higher score indicate a higher level of functioning. SF-12 total score was calculated by a clinician scoring 12-question survey filled by participants. Transformed physical component summary score (PCS) and transformed mental component summary score (MCS) are derived using the sum of all 12 items and scored onto a 0-100 scale such that a higher score indicates a better health state and better functioning. The Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1) and Week 48

,
InterventionScore on a scale (Mean)
Total Score, Week 48, n=205, 192MCS, Week 48, n=205, 192PCS, Week 48, n=205, 192
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase0.12.3291.444
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase0.02.3971.905

[back to top]

Change From Baseline in Alanine Aminotransferase, Alkaline Phosphatase, Aspartate Aminotransferase, Creatine Kinase at Indicated Time Points

Clinical chemistry parameters were assessed at Baseline (Day 1), Week 4, 12, 24, 36 and Week 48. Change from Baseline in alanine aminotransferase, alkaline phosphatase, aspartate aminotransferase, creatine kinase is summarized. Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1), Week 4, Week 12, Week 24, Week 36 and Week 48

,
InterventionInternational units per liter (Mean)
Alanine aminotransferase, Week 4, n= 245, 237Alanine aminotransferase, Week 12, n= 236, 226Alanine aminotransferase, Week 24, n= 225, 212Alanine aminotransferase, Week 36, n= 219, 204Alanine aminotransferase, Week 48, n= 208, 192Alkaline phosphatase, Week 4, n= 245, 237Alkaline phosphatase, Week 12, n= 236, 226Alkaline phosphatase, Week 24, n= 225, 212Alkaline phosphatase, Week 36, n= 219, 204Alkaline phosphatase, Week 48, n= 208, 192Aspartate aminotransferase, Week 4, n= 244, 237Aspartate aminotransferase, Week 12, n= 236, 226Aspartate aminotransferase, Week 24, n= 224, 212Aspartate aminotransferase, Week 36, n= 219, 204Aspartate aminotransferase, Week 48, n= 208, 192Creatine Kinase, Week 4, n= 245, 237Creatine Kinase, Week 12, n= 236, 226Creatine Kinase, Week 24, n= 225, 212Creatine Kinase, Week 36, n= 219, 204Creatine Kinase, Week 48, n= 208, 192
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase-3.4-2.3-3.7-5.3-1.59.415.122.420.421.9-3.6-4-5.1-6.5-3.735.67.35.87.23.8
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase-3.3-5.2-5.4-4.9-5.7-1.5-2.10.50.62.9-3.3-6.2-6.3-6.4-7.5-0.36.910.311.923.8

[back to top]

Change From Baseline in Albumin at Indicated Timepoints

Clinical chemistry parameters were assessed at Baseline (Day 1), Week 4, 12, 24, 36 and Week 48. Change from Baseline in albumin is summarized. Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1), Week 4, Week 12, Week 24, Week 36 and Week 48

,
InterventionGrams per liter (Mean)
Week 4, n= 245, 237Week 12, n= 236, 226Week 24, n= 225, 212Week 36, n= 219, 204Week 48, n= 208, 192
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase-0.50.10.80.61.3
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase0.10.51.41.41.7

[back to top]

Change From Baseline in Basophils, Eosinophils, Lymphocytes, Monocytes at Indicated Time Points

Hematology parameters were assessed at Baseline (Day 1), Week 4, 12, 24, 36 and Week 48. Change from Baseline in basophils, eosinophils, lymphocytes, monocytes is summarized. Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1), Week 4, Week 12, Week 24, Week 36 and Week 48

,
Intervention10^9 cells per liter (Mean)
Basophils, Week 4, n= 241, 234Basophils, Week 12, n= 228, 216Basophils, Week 24, n= 221, 208Basophils, Week 36, n= 214, 203Basophils, Week 48, n= 206, 189Eosinophils, Week 4, n= 241, 234Eosinophils, Week 12, n= 228, 216Eosinophils, Week 24, n= 221, 208Eosinophils, Week 36, n= 214, 203Eosinophils, Week 48, n= 206, 189Lymphocytes, Week 4, n= 241, 234Lymphocytes, Week 12, n= 228, 216Lymphocytes, Week 24, n= 221, 208Lymphocytes, Week 36, n= 214, 203Lymphocytes, Week 48, n= 206, 189Monocytes, Week 4, n= 241, 234Monocytes, Week 12, n= 228, 216Monocytes, Week 24, n= 221, 208Monocytes, Week 36, n= 214, 203Monocytes, Week 48, n= 206, 189
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase0.0030.0030.0030.0030.0060.021-0.0010.0050.0140.0070.1190.1560.1920.1780.261-0.015-0.031-0.015-0.028-0.024
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase0.0030.0020.0040.0040.0050.0400.0370.0280.0480.0300.2080.2570.3170.3620.359-0.001-0.0100.008-0.0060.001

[back to top]

Change From Baseline in Bilirubin and Creatinine at Indicated Timepoints

Clinical chemistry parameters were assessed at Baseline (Day 1), Weeks 4, 12, 24, 36 and 48. Change from Baseline in bilirubin and creatinine are summarized. Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1), Week 4, Week 12, Week 24, Week 36 and Week 48

,
InterventionMicromoles per liter (Mean)
Bilirubin, Week 4, n= 244, 237Bilirubin, Week 12, n= 236, 226Bilirubin, Week 24, n= 225, 212Bilirubin, Week 36, n= 219, 204Bilirubin, Week 48, n= 208, 192Creatinine, Week 4, n= 245, 237Creatinine, Week 12, n= 236, 226Creatinine, Week 24, n= 225, 212Creatinine, Week 36, n= 219, 204Creatinine, Week 48, n= 208, 192
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase27.222.82523.823.74.895.835.85.375.86
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase-0.8-0.6-0.2-0.2-0.38.49.29.1610.089.29

[back to top]

Change From Baseline in Bone Specific Alkaline Phosphatase, Osteocalcin and Procollagen 1 N-terminal Propeptide at Indicated Timepoints

Bone markers were assessed at Baseline (Day 1), Weeks 24, 48. Change from Baseline in bone specific alkaline phosphatase (BSAP), osteocalcin and procollagen 1 N-terminal propeptide (PTP) is summarized. The Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1), Weeks 24 and 48

,
InterventionMicrograms per liter (Mean)
BSAP, Week 24, n=219, 207BSAP, Week 48, n=202, 184Osteocalcin, Week 24, n=209, 197Osteocalcin, Week 48, n=194, 178PTP, Week 24, n=223, 206PTP, Week 48, n=205, 186
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase6.007.6014.3816.3032.034.1
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase1.332.643.735.1510.111.2

[back to top]

Change From Baseline in Carbon Dioxide, Electrolytes, Lipids, Glucose, Urea at Indicated Time Points

Clinical chemistry parameters were assessed at Baseline (Day 1), Weeks 4, 12, 24, 36 and 48. Change from Baseline in carbon dioxide, electrolytes (chloride, hyperkalemia, hypernatremia, hypokalemia, hyponatremia, phosphate, potassium, sodium), lipids (cholesterol [CHLS], high density lipoprotein [HDL] CHLS direct, low density lipoprotein (LDL) CHLS calculation, LDL CHLS direct, triglycerides), glucose (hyperglycaemia, hypoglycaemia) and urea are summarized. Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. Laboratory parameters were assessed in Safety Population which comprised of all participants who received at least one dose of study treatment. (NCT01910402)
Timeframe: Baseline (Day 1), Week 4, Week 12, Week 24, Week 36 and Week 48

InterventionMillimoles per liter (Mean)
Carbon Dioxide, Week 4, n= 244, 237Carbon Dioxide, Week 12, n= 236, 226Carbon Dioxide, Week 24, n= 224, 212Carbon Dioxide, Week 36, n= 219, 204Carbon Dioxide, Week 48, n= 208, 192Chloride, Week 4, n= 245, 237Chloride, Week 12, n= 236, 226Chloride, Week 24, n= 225, 212Chloride, Week 36, n= 219, 204Chloride, Week 48, n= 208, 192CHLS, Week 4, n= 1, 3CHLS, Week 12, n= 224, 221CHLS, Week 24, n= 218, 201CHLS, Week 36, n= 205, 191CHLS, Week 48, n= 195, 175Glucose, Week 12, n= 226, 224Glucose, Week 24, n= 219, 204Glucose, Week 36, n= 211, 196Glucose, Week 48, n= 197, 180HDL CHLS, Direct, Week 4, n= 1, 3HDL CHLS, Direct, Week 12, n= 224, 221HDL CHLS, Direct, Week 24, n= 218, 201HDL CHLS, Direct, Week 36, n= 205, 191HDL CHLS, Direct, Week 48, n= 195, 175Hyperglycaemia, Week 12, n= 226, 224Hyperglycaemia, Week 24, n= 219, 204Hyperglycaemia, Week 36, n= 211, 196Hyperglycaemia, Week 48, n= 197, 180Hyperkalemia, Week 4, n= 244, 237Hyperkalemia, Week 12, n= 236, 226Hyperkalemia, Week 24, n= 224, 212Hyperkalemia, Week 36, n= 219, 204Hyperkalemia, Week 48, n= 208, 192Hypernatremia, Week 4, n= 245, 237Hypernatremia, Week 12, n= 236, 226Hypernatremia, Week 24, n= 225, 212Hypernatremia, Week 36, n= 219, 204Hypernatremia, Week 48, n= 208, 192Hypoglycaemia, Week 12, n= 226, 224Hypoglycaemia, Week 24, n= 219, 204Hypoglycaemia, Week 36, n= 211, 196Hypoglycaemia, Week 48, n= 197, 180Hypokalemia, Week 4, n= 244, 237Hypokalemia, Week 12, n= 236, 226Hypokalemia, Week 24, n= 224, 212Hypokalemia, Week 36, n= 219, 204Hypokalemia, Week 48, n= 208, 192Hyponatremia, Week 4, n= 245, 237Hyponatremia, Week 12, n= 236, 226Hyponatremia, Week 24, n= 225, 212Hyponatremia, Week 36, n= 219, 204Hyponatremia, Week 48, n= 208, 192LDL CHLS Calculation, Week 4, n= 1, 3LDL CHLS Calculation, Week 12, n= 221, 219LDL CHLS Calculation, Week 24, n= 213, 201LDL CHLS Calculation, Week 36, n= 201, 188LDL CHLS Calculation, Week 48, n= 190, 175LDL CHLS, Direct, Week 12, n= 0, 1Phosphate, Week 4, n= 245, 237Phosphate, Week 12, n= 236, 226Phosphate, Week 24, n= 225, 212Phosphate, Week 36, n= 219, 204Phosphate, Week 48, n= 208, 192Potassium, Week 4, n= 244, 237Potassium, Week 12, n= 236, 226Potassium, Week 24, n= 224, 212Potassium, Week 36, n= 219, 204Potassium, Week 48, n= 208, 192Sodium, Week 4, n= 245, 237Sodium, Week 12, n= 236, 226Sodium, Week 24, n= 225, 212Sodium, Week 36, n= 219, 204Sodium, Week 48, n= 208, 192Triglycerides, Week 4, n= 1, 3Triglycerides, Week 12, n= 224, 221Triglycerides, Week 24, n= 218, 201Triglycerides, Week 36, n= 205, 191Triglycerides, Week 48, n= 195, 175Urea, Week 4, n= 245, 237Urea, Week 12, n= 236, 226Urea, Week 24, n= 225, 212Urea, Week 36, n= 219, 204Urea, Week 48, n= 208, 192
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase0.60.80.30.60.4-0.50.2-0.100-0.017-0.058-0.00100.1090.220.260.340.2400.0050.0530.0360.0810.220.260.340.240.120.10.060.130.04-0.50.10.20.20.50.220.260.340.240.120.10.060.130.04-0.50.10.20.20.5-0.123-0.14-0.111-0.099-0.021-0.44-0.0320.0260.0260.00900.120.10.060.130.04-0.50.10.20.20.50.2370.1670.1250.1570.1070.10.160.12-0.030.02

[back to top]

Change From Baseline in Carbon Dioxide, Electrolytes, Lipids, Glucose, Urea at Indicated Time Points

Clinical chemistry parameters were assessed at Baseline (Day 1), Weeks 4, 12, 24, 36 and 48. Change from Baseline in carbon dioxide, electrolytes (chloride, hyperkalemia, hypernatremia, hypokalemia, hyponatremia, phosphate, potassium, sodium), lipids (cholesterol [CHLS], high density lipoprotein [HDL] CHLS direct, low density lipoprotein (LDL) CHLS calculation, LDL CHLS direct, triglycerides), glucose (hyperglycaemia, hypoglycaemia) and urea are summarized. Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. Laboratory parameters were assessed in Safety Population which comprised of all participants who received at least one dose of study treatment. (NCT01910402)
Timeframe: Baseline (Day 1), Week 4, Week 12, Week 24, Week 36 and Week 48

InterventionMillimoles per liter (Mean)
Carbon Dioxide, Week 4, n= 244, 237Carbon Dioxide, Week 12, n= 236, 226Carbon Dioxide, Week 24, n= 224, 212Carbon Dioxide, Week 36, n= 219, 204Carbon Dioxide, Week 48, n= 208, 192Chloride, Week 4, n= 245, 237Chloride, Week 12, n= 236, 226Chloride, Week 24, n= 225, 212Chloride, Week 36, n= 219, 204Chloride, Week 48, n= 208, 192CHLS, Week 4, n= 1, 3CHLS, Week 12, n= 224, 221CHLS, Week 24, n= 218, 201CHLS, Week 36, n= 205, 191CHLS, Week 48, n= 195, 175Glucose, Week 12, n= 226, 224Glucose, Week 24, n= 219, 204Glucose, Week 36, n= 211, 196Glucose, Week 48, n= 197, 180HDL CHLS, Direct, Week 4, n= 1, 3HDL CHLS, Direct, Week 12, n= 224, 221HDL CHLS, Direct, Week 24, n= 218, 201HDL CHLS, Direct, Week 36, n= 205, 191HDL CHLS, Direct, Week 48, n= 195, 175Hyperglycaemia, Week 12, n= 226, 224Hyperglycaemia, Week 24, n= 219, 204Hyperglycaemia, Week 36, n= 211, 196Hyperglycaemia, Week 48, n= 197, 180Hyperkalemia, Week 4, n= 244, 237Hyperkalemia, Week 12, n= 236, 226Hyperkalemia, Week 24, n= 224, 212Hyperkalemia, Week 36, n= 219, 204Hyperkalemia, Week 48, n= 208, 192Hypernatremia, Week 4, n= 245, 237Hypernatremia, Week 12, n= 236, 226Hypernatremia, Week 24, n= 225, 212Hypernatremia, Week 36, n= 219, 204Hypernatremia, Week 48, n= 208, 192Hypoglycaemia, Week 12, n= 226, 224Hypoglycaemia, Week 24, n= 219, 204Hypoglycaemia, Week 36, n= 211, 196Hypoglycaemia, Week 48, n= 197, 180Hypokalemia, Week 4, n= 244, 237Hypokalemia, Week 12, n= 236, 226Hypokalemia, Week 24, n= 224, 212Hypokalemia, Week 36, n= 219, 204Hypokalemia, Week 48, n= 208, 192Hyponatremia, Week 4, n= 245, 237Hyponatremia, Week 12, n= 236, 226Hyponatremia, Week 24, n= 225, 212Hyponatremia, Week 36, n= 219, 204Hyponatremia, Week 48, n= 208, 192LDL CHLS Calculation, Week 4, n= 1, 3LDL CHLS Calculation, Week 12, n= 221, 219LDL CHLS Calculation, Week 24, n= 213, 201LDL CHLS Calculation, Week 36, n= 201, 188LDL CHLS Calculation, Week 48, n= 190, 175LDL CHLS, Direct, Week 24, n= 1, 0LDL CHLS, Direct, Week 36, n= 1, 0Phosphate, Week 4, n= 245, 237Phosphate, Week 12, n= 236, 226Phosphate, Week 24, n= 225, 212Phosphate, Week 36, n= 219, 204Phosphate, Week 48, n= 208, 192Potassium, Week 4, n= 244, 237Potassium, Week 12, n= 236, 226Potassium, Week 24, n= 224, 212Potassium, Week 36, n= 219, 204Potassium, Week 48, n= 208, 192Sodium, Week 4, n= 245, 237Sodium, Week 12, n= 236, 226Sodium, Week 24, n= 225, 212Sodium, Week 36, n= 219, 204Sodium, Week 48, n= 208, 192Triglycerides, Week 4, n= 1, 3Triglycerides, Week 12, n= 224, 221Triglycerides, Week 24, n= 218, 201Triglycerides, Week 36, n= 205, 191Triglycerides, Week 48, n= 195, 175Urea, Week 4, n= 245, 237Urea, Week 12, n= 236, 226Urea, Week 24, n= 225, 212Urea, Week 36, n= 219, 204Urea, Week 48, n= 208, 192
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase-0.4-0.2-0.50-0.60.610.70.90.7-0.10.2980.3170.330.4470.30.170.170.18-0.10.1820.2010.2040.2310.30.170.170.18-0.010.03-0.040.03-0.0400.70.60.90.60.30.170.170.18-0.010.03-0.040.03-0.0400.70.60.90.60.080.1250.1110.1120.213-0.64-0.2300.020.0210.0290.016-0.010.03-0.040.03-0.0400.70.60.90.6-0.18-0.040.0360.0370.018-0.040.080.030.080.1

[back to top]

Change From Baseline in CD4+ Cell Count at Indicated Timepoints-Continuation Phase

Change from Baseline in cluster of differentiation 4(CD4+) cell count were assessed at indicated time points. The Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1), Week 96, Week 432

InterventionCells per cubic millimeter (Mean)
Week 96, n=99Week 432, n=3
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Continuation Phase286.5254.7

[back to top]

Change From Baseline in CD4+ Cell Count at Indicated Timepoints-Randomized Phase

Change from Baseline in cluster of differentiation 4 (CD4+) cell count were assessed at indicated time points. The Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1), Week 4, Week 12, Week 24, Week 36 and Week 48

,
InterventionCells per cubic millimeter (Mean)
Week 4, n=245, 237Week 12, n=236, 224Week 24, n=226, 210Week 36, n=219, 204Week 48, n=208, 191
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase73.7124.4163.0191.4230.7
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase94.9143.8200.6230.7248.8

[back to top]

Change From Baseline in Creatinine Clearance at Indicated Time Points

Clinical chemistry parameters were assessed at Baseline (Day 1), Week 4, 12, 24, 36 and Week 48. Change from Baseline in creatinine clearance is summarized. Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1), Week 4, Week 12, Week 24, Week 36 and Week 48

,
InterventionMilliliter per minute (Mean)
Week 4, n= 245, 237Week 12, n= 236, 226Week 24, n= 225, 212Week 36, n= 219, 204Week 48, n= 208, 192
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase-7.5-7-9.1-7.5-7.7
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase-16.3-17.3-16.2-16.8-15.9

[back to top]

Change From Baseline in Erythrocyte Mean Corpuscular Volume at Indicated Time Points

Hematology parameters were assessed at Baseline (Day 1), Week 4, 12, 24, 36 and Week 48. Change from Baseline in erythrocyte mean corpuscular volume (EMCV) is summarized. Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1), Week 4, Week 12, Week 24, Week 36 and Week 48

,
InterventionFemtoliter (Mean)
Week 4, n= 243, 234Week 12, n= 233, 220Week 24, n= 225, 211Week 36, n= 218, 203Week 48, n= 207, 190
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase0.51.93.13.13.7
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase0.93.45.56.07.1

[back to top]

Change From Baseline in Erythrocytes at Indicated Time Points

Hematology parameters were assessed at Baseline (Day 1), Week 4, 12, 24, 36 and Week 48. Change from Baseline in erythrocytes is summarized. Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1), Week 4, Week 12, Week 24, Week 36 and Week 48

,
Intervention10^12 cells per liter (Mean)
Week 4, n= 243, 234Week 12, n= 233, 220Week 24, n= 225, 211Week 36, n= 218, 203Week 48, n= 207, 190
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase-0.07-0.09-0.09-0.08-0.05
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase-0.04-0.07-0.08-0.10-0.10

[back to top]

Change From Baseline in Hematocrit Count at Indicated Time Points

Hematology parameters were assessed at Baseline (Day 1), Weeks 4, 12, 24, 36 and 48. Change from Baseline in hematocrit is summarized. Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1), Week 4, Week 12, Week 24, Week 36 and Week 48

,
InterventionProportion of red blood cells in blood (Mean)
Week 4, n= 243, 234Week 12, n= 233, 220Week 24, n= 225, 211Week 36, n= 218, 203Week 48, n= 207, 190
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase-0.00420.00000.00510.00620.0107
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase0.00030.00810.01570.01670.0212

[back to top]

Change From Baseline in Lipase at Indicated Timepoints

Clinical chemistry parameters were assessed at Baseline (Day 1), Week 4, 12, 24, 36 and Week 48. Change from Baseline in lipase is summarized. Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1), Week 4, Week 12, Week 24, Week 36 and Week 48

,
InterventionUnits per liter (Mean)
Week 4, n= 245, 237Week 12, n= 236, 226Week 24, n= 225, 212Week 36, n= 219, 204Week 48, n= 208, 192
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase-1.3-2.1-6-6.3-7.8
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase-1.2-2.2-6-6.3-6.5

[back to top]

Change From Baseline in Plasma HIV-1 RNA at Indicated Time Points-Randomized Phase

Change from the Baseline in plasma HIV-1 RNA were assessed at indicated time points. The Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1), Week 4, Week 12, Week 24, Week 36 and Week 48

,
InterventionLog10 copies/mL (Mean)
Week 4, n=245, 238Week 12, n=236, 226Week 24, n=225, 212Week 36, n=221, 204Week 48, n=207, 192
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase-1.923-2.541-2.726-2.772-2.752
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase-2.591-2.756-2.789-2.838-2.874

[back to top]

Change From Baseline in Total CHLS/HDL CHLS Ratio at Indicated Timepoints

Clinical chemistry parameters were assessed at Baseline (Day 1), Week 4, 12, 24, 36 and Week 48. Change from Baseline in Total CHLS/HDL CHLS ratio is summarized. Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1), Week 4, Week 12, Week 24, Week 36 and Week 48

,
InterventionRatio (Mean)
Week 4, n= 1, 4Week 12, n= 233, 223Week 24, n= 224, 209Week 36, n= 212, 198Week 48, n= 207, 186
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase0.2159-0.1092-0.1922-0.1433-0.1444
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase0.1264-0.2736-0.3098-0.3286-0.2886

[back to top]

Change From Baseline in Type I Collagen C-telopeptides at Indicated Timepoints

Bone markers were assessed at Baseline (Day 1), Weeks 24, 48. Change from Baseline in Type I collagen C-telopeptides (T-1 CCT) is summarized. The Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1), Weeks 24 and 48

,
InterventionNanograms per liter (Mean)
Week 24, n=221, 207Week 48, n=202, 185
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase272.4267.9
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase89.875.9

[back to top]

Change From Baseline in Urine Albumin Creatinine Ratio at Indicated Time Points

Change from Baseline in urine albumin creatinine ratio at Week 24 and Week 48 is summarized. The Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1), Week 24 and Week 48

,
Interventionmilligrams per millimole (Mean)
Week 24, n= 179, 186Week 48, n= 170, 164
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase-1.03-0.10
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase-1.15-0.68

[back to top]

Change From Baseline in Vitamin D, Vitamin D2 and Vitamin D3 at Week 24 and Week 48

Bone markers were assessed at Baseline (Day 1), Weeks 24, 48. Change from Baseline in vitamin D, vitamin D2 and vitamin D3 is summarized. The Baseline value was defined as the latest pre-dose assessment (Day 1) value. Change from Baseline was calculated as post-dose visit value minus Baseline value. (NCT01910402)
Timeframe: Baseline (Day 1), Weeks 24 and 48

,
InterventionNanomoles per liter (Mean)
Vitamin D, Week 24, n=223, 208Vitamin D, Week 48, n=206, 186Vitamin D2, Week 24, n=223, 208Vitamin D2, Week 48, n=206, 186Vitamin D3, Week 24, n=223, 208Vitamin D3, Week 48, n=206, 186
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase16.38.91.00.915.27.9
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase1.8-1.90.30.11.5-1.9

[back to top]

HIVTSQs Total Score at Indicated Timepoints

The HIV treatment satisfaction questionnaire (HIVTSQ) is a 10-item self-reported scale that measures overall satisfaction with treatment and by specific domains e.g. convenience, flexibility. The HIVTSQ items are summed up to produce a treatment satisfaction total score (0 to 60) and an individual satisfaction rating for each item (0 to 6) and two subscales: general satisfaction/clinical and lifestyle/ease subscales. The higher the score, the greater the improvement in treatment satisfaction as compared to the past few weeks. A smaller score represents a decline in treatment satisfaction compared to the past few weeks. Statistical analysis was performed based on Wilcoxon rank sum test. (NCT01910402)
Timeframe: Weeks 4, 12, 24 and 48

,
InterventionScore on a scale (Mean)
Week 4, n=243, 239Week 12, n=236, 226Week 24, n=225, 211Week 48, n=206, 191
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase51.953.654.355.4
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase54.056.156.857.0

[back to top]

Number of Participants With AEs by Maximum Toxicity-Continuation Phase

Number of participants with Grade 1-4 AEs were assessed in Continuation Phase. AEs are categorized into following grades as per The Division of Aqcuired Immuno Deficiency Syndrome (AIDS) Table for Grading the Severity of Adult and Pediatric Adverse Events (DAIDS AE Grading Table)- Grade 1- mild, Grade 2- moderate; Grade 3- severe and Grade 4- potentially life-threatening. Higher the grade, more severe the symptoms. (NCT01910402)
Timeframe: From Weeks 48 to 432

InterventionParticipants (Count of Participants)
Grade 1Grade 2Grade 3Grade 4
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Continuation Phase324876

[back to top]

Number of Participants With AEs by Maximum Toxicity-Randomized Phase

Number of participants with Grade 1-4 AEs by maximum toxicity were assessed from the start of study treatment and until end of the Randomization phase. AEs are categorized into following grades as per The Division of Aqcuired Immuno Deficiency Syndrome (AIDS) Table for Grading the Severity of Adult and Pediatric Adverse Events (DAIDS AE Grading Table)- Grade 1- mild, Grade 2- moderate; Grade 3- severe and Grade 4- potentially life-threatening. Higher the grade, more severe the symptoms. (NCT01910402)
Timeframe: Up to Week 48

,
InterventionParticipants (Count of Participants)
Grade 1Grade 2Grade 3Grade 4
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase6091379
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase7994183

[back to top]

Number of Participants With Any Adverse Events (AEs), and Serious Adverse Events (SAEs)-Randomized Phase

An AE is defined as any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease (new or exacerbated) temporally associated with the use of a medicinal product. An SAE is any untoward medical occurrence that, at any dose results in death, is life-threatening, requires hospitalization or prolongation of existing hospitalization, results in disability/incapacity, or is a congenital anomaly/birth defect or other events that may jeopardize the participant or may require medical or surgical intervention to prevent one of the outcome listed above, liver injury and impaired liver function and grade 4 laboratory abnormalities. Number of participants with any AEs, and SAEs have been presented. (NCT01910402)
Timeframe: Up to Week 48

,
InterventionParticipants (Count of Participants)
Any AEsAny SAEs
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase19720
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase19512

[back to top]

Number of Participants With Any AEs, and SAEs in Continuation Phase

An AE is defined as any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease (new or exacerbated) temporally associated with the use of a medicinal product. An SAE is any untoward medical occurrence that, at any dose results in death, is life-threatening, requires hospitalization or prolongation of existing hospitalization, results in disability/incapacity, or is a congenital anomaly/birth defect or other events that may jeopardize the participant or may require medical or surgical intervention to prevent one of the outcome listed above, liver injury and impaired liver function and grade 4 laboratory abnormalities. Number of participants with any AEs, and SAEs have been presented. (NCT01910402)
Timeframe: From Weeks 48 to 432

InterventionParticipants (Count of Participants)
Any AEsAny SAEs
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Continuation Phase9313

[back to top]

Number of Participants With Maximum Post-Baseline Emergent Chemistry Toxicities-Continuation Phase

Number of participants with grades 1-4 emergent chemistry toxicities were assessed in Continuation Phase. Chemistry toxicities were categorized into following grades as per The Division of AIDS Table for Grading the Severity of Adult and Pediatric Adverse Events (DAIDS AE Grading Table)- Grade 1- mild, Grade 2- moderate; Grade 3- severe and Grade 4- potentially life-threatening. Higher the grade, more severe the symptoms. Data has been reported for clinical chemistry parameters including hyperglycaemia, hypernatremia, hypoglycaemia, hypokalemia, hyponatremia, alanine aminotransferase, albumin, alkaline phosphatase, aspartate aminotransferase, bilirubin, carbon dioxide, cholesterol, creatine kinase, creatinine, LDL cholesterol calculation, LDL cholesterol direct, lipase, phosphate, potassium, sodium, triglycerides and glucose. (NCT01910402)
Timeframe: From Weeks 48 to 432

InterventionParticipants (Count of Participants)
Hyperglycaemia, Grade 1, n=143Hyperglycaemia, Grade 2, n=143Hyperglycaemia, Grade 3, n=143Hyperglycaemia, Grade 4, n=143Hypernatremia, Grade 1, n=146Hypernatremia, Grade 2, n=146Hypernatremia, Grade 3, n=146Hypernatremia, Grade 4, n=146Hypoglycaemia, Grade 1, n=143Hypoglycaemia, Grade 2, n=143Hypoglycaemia, Grade 3, n=143Hypoglycaemia, Grade 4, n=143Hypokalemia, Grade 1, n=146Hypokalemia, Grade 2, n=146Hypokalemia, Grade 3, n=146Hypokalemia, Grade 4, n=146Hyponatremia, Grade 1, n=146Hyponatremia, Grade 2, n=146Hyponatremia, Grade 3, n=146Hyponatremia, Grade 4, n=146Alanine aminotransferase, Grade 1, n=146Alanine aminotransferase, Grade 2, n=146Alanine aminotransferase, Grade 3, n=146Alanine aminotransferase, Grade 4, n=146Alkaline phosphatase, Grade 1, n=146Alkaline phosphatase, Grade 2, n=146Alkaline phosphatase, Grade 3, n=146Alkaline phosphatase, Grade 4, n=146Aspartate aminotransferase, Grade 1, n=146Aspartate aminotransferase, Grade 2, n=146Aspartate aminotransferase, Grade 3, n=146Aspartate aminotransferase, Grade 4, n=146Bilirubin, Grade 1, n=146Bilirubin, Grade 2, n=146Bilirubin, Grade 3, n=146Bilirubin, Grade 4, n=146Carbon dioxide, Grade 1, n=146Carbon dioxide, Grade 2, n=146Carbon dioxide, Grade 3, n=146Carbon dioxide, Grade 4, n=146Cholesterol, Grade 1, n=71Cholesterol, Grade 2, n=71Cholesterol, Grade 3, n=71Cholesterol, Grade 4, n=71Creatine kinase, Grade 1, n=146Creatine kinase, Grade 2, n=146Creatine kinase, Grade 3, n=146Creatine kinase, Grade 4, n=146Creatinine, Grade 1, n=146Creatinine, Grade 2, n=146Creatinine, Grade 3, n=146Creatinine, Grade 4, n=146LDL cholesterol calculation, Grade 1, n=70LDL cholesterol calculation, Grade 2, n=70LDL cholesterol calculation, Grade 3, n=70LDL cholesterol calculation, Grade 4, n=70LDL cholesterol direct, Grade 1, n=2LDL cholesterol direct, Grade 2, n=2LDL cholesterol direct, Grade 3, n=2LDL cholesterol direct, Grade 4, n=2Lipase, Grade 1, n=146Lipase, Grade 2, n=146Lipase, Grade 3, n=146Lipase, Grade 4, n=146Phosphate, Grade 1, n=146Phosphate, Grade 2, n=146Phosphate, Grade 3, n=146Phosphate, Grade 4, n=146Potassium, Grade 1, n=146Potassium, Grade 2, n=146Potassium, Grade 3, n=146Potassium, Grade 4, n=146Sodium, Grade 1, n=146Sodium, Grade 2, n=146Sodium, Grade 3, n=146Sodium, Grade 4, n=146Triglycerides, Grade 1, n=71Triglycerides, Grade 2, n=71Triglycerides, Grade 3, n=71Triglycerides, Grade 4, n=71Glucose, Grade 1, n=143Glucose, Grade 2, n=143Glucose, Grade 3, n=143Glucose, Grade 4, n=143
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Continuation Phase249302000100113000360007302500010202413058700993061115001582010009611215201300037000010024931

[back to top]

Number of Participants With Maximum Post-Baseline Emergent Chemistry Toxicities-Randomized Phase

Number of participants with Grade 1-4 emergent chemistry toxicities were assessed from the start of study treatment and end of Randomized Phase. Chemistry toxicities were categorized into following grades as per The Division of AIDS Table for Grading the Severity of Adult and Pediatric Adverse Events (DAIDS AE Grading Table)- Grade 1- mild, Grade 2- moderate; Grade 3- severe and Grade 4- potentially life-threatening. Higher the grade, more severe the symptoms. Data has been reported for clinical chemistry parameters including hyperglycaemia, hyperkalemia, hypernatremia, hypoglycaemia, hypokalemia, hyponatremia, alanine aminotransferase, albumin, alkaline phosphatase, aspartate aminotransferase, bilirubin, carbon dioxide, cholesterol, creatine kinase, creatinine, LDL cholesterol calculation, LDL cholesterol direct, lipase, phosphate, potassium, sodium, triglycerides and glucose. (NCT01910402)
Timeframe: Up to Week 48

,
InterventionParticipants (Count of Participants)
Hyperglycaemia, Grade 1Hyperglycaemia, Grade 2Hyperglycaemia, Grade 3Hyperglycaemia, Grade 4Hyperkalemia, Grade 1Hyperkalemia, Grade 2Hyperkalemia, Grade 3Hyperkalemia, Grade 4Hypernatremia, Grade 1Hypernatremia, Grade 2Hypernatremia, Grade 3Hypernatremia, Grade 4Hypoglycaemia, Grade 1Hypoglycaemia, Grade 2Hypoglycaemia, Grade 3Hypoglycaemia, Grade 4Hypokalemia, Grade 1Hypokalemia, Grade 2Hypokalemia, Grade 3Hypokalemia, Grade 4Hyponatremia, Grade 1Hyponatremia, Grade 2Hyponatremia, Grade 3Hyponatremia, Grade 4Alanine aminotransferase, Grade 1Alanine aminotransferase, Grade 2Alanine aminotransferase, Grade 3Alanine aminotransferase, Grade 4Albumin, Grade 1Albumin, Grade 2Albumin, Grade 3Albumin, Grade 4Alkaline phosphatase, Grade 1Alkaline phosphatase, Grade 2Alkaline phosphatase, Grade 3Alkaline phosphatase, Grade 4Aspartate aminotransferase, Grade 1Aspartate aminotransferase, Grade 2Aspartate aminotransferase, Grade 3Aspartate aminotransferase, Grade 4Bilirubin, Grade 1Bilirubin, Grade 2Bilirubin, Grade 3Bilirubin, Grade 4Carbon dioxide, Grade 1Carbon dioxide, Grade 2Carbon dioxide, Grade 3Carbon dioxide, Grade 4Cholesterol, Grade 1Cholesterol, Grade 2Cholesterol, Grade 3Cholesterol, Grade 4Creatine kinase, Grade 1Creatine kinase, Grade 2Creatine kinase, Grade 3Creatine kinase, Grade 4Creatinine, Grade 1Creatinine, Grade 2Creatinine, Grade 3Creatinine, Grade 4LDL cholesterol calculation, Grade 1LDL cholesterol calculation, Grade 2LDL cholesterol calculation, Grade 3LDL cholesterol calculation, Grade 4LDL cholesterol direct, Grade 1LDL cholesterol direct, Grade 2LDL cholesterol direct, Grade 3LDL cholesterol direct, Grade 4Lipase, Grade 1Lipase, Grade 2Lipase, Grade 3Lipase, Grade 4Phosphate, Grade 1Phosphate, Grade 2Phosphate, Grade 3Phosphate, Grade 4Potassium, Grade 1Potassium, Grade 2Potassium, Grade 3Potassium, Grade 4Sodium, Grade 1Sodium, Grade 2Sodium, Grade 3Sodium, Grade 4Triglycerides, Grade 1Triglycerides, Grade 2Triglycerides, Grade 3Triglycerides, Grade 4Glucose, Grade 1Glucose, Grade 2Glucose, Grade 3Glucose, Grade 4
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase11930010000005100190005700074202200141007420528657554300319205101730021920100073211192019100570000200151030
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase17164110001000631017100441005611300032001241120006540052284031303010381370310012530571018100451000520221941

[back to top]

Number of Participants With Maximum Post-Baseline Emergent Hematology Toxicities-Continuation Phase

Number of participants with Grade 1-4 emergent hematology toxicities were assessed in Continuation Phase. Hematology toxicities were categorized into following grades as per The Division of AIDS Table for Grading the Severity of Adult and Pediatric Adverse Events (DAIDS AE Grading Table)- Grade 1- mild, Grade 2- moderate; Grade 3- severe and Grade 4- potentially life-threatening. Higher the grade, more severe the symptoms. Data has been reported for clinical chemistry parameters including hemoglobin, leukocytes, neutrophils and platelets. (NCT01910402)
Timeframe: From Weeks 48 to 432

InterventionParticipants (Count of Participants)
Hemoglobin, Grade 1Hemoglobin, Grade 2Hemoglobin, Grade 3Hemoglobin, Grade 4Leukocytes, Grade 1Leukocytes, Grade 2Leukocytes, Grade 3Leukocytes, Grade 4Neutrophils, Grade 1Neutrophils, Grade 2Neutrophils, Grade 3Neutrophils, Grade 4Platelets, Grade 1Platelets, Grade 2Platelets, Grade 3Platelets, Grade 4
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Continuation Phase51002010102113100

[back to top]

Number of Participants With Maximum Post-Baseline Emergent Hematology Toxicities-Randomized Phase

Number of participants with Grade 1-4 emergent hematology toxicities were assessed from the start of study treatment and end of Randomized Phase. Hematology toxicities were categorized into following grades as per The Division of AIDS Table for Grading the Severity of Adult and Pediatric Adverse Events (DAIDS AE Grading Table)- Grade 1- mild, Grade 2- moderate; Grade 3- severe and Grade 4- potentially life-threatening. Higher the grade, more severe the symptoms. Data has been reported for clinical chemistry parameters including hemoglobin, leukocytes, neutrophils and platelets. (NCT01910402)
Timeframe: Up to Week 48

,
InterventionParticipants (Count of Participants)
Hemoglobin, Grade 1Hemoglobin, Grade 2Hemoglobin, Grade 3Hemoglobin, Grade 4Leukocytes, Grade 1Leukocytes, Grade 2Leukocytes, Grade 3Leukocytes, Grade 4Neutrophils, Grade 1Neutrophils, Grade 2Neutrophils, Grade 3Neutrophils, Grade 4Platelets, Grade 1Platelets, Grade 2Platelets, Grade 3Platelets, Grade 4
ATV 300 mg+RTV 100 mg+TDF 300 mg/FTC 200 mg QD-Randomized Phase213106200129211200
DTG 50 mg/ABC 600 mg/3TC 300 mg QD-Randomized Phase72105100157016010

[back to top]

Number of Participants With Post-Baseline HIV-1 Disease Progression for DTG 50 mg/ABC 600 mg/3TC 300 mg QD (Randomized + Continuation Phase)

Number of participants with post-Baseline HIV-1disease progression were assessed during study period. The CDC Classification System for HIV Infection is the medical classification system used by the United States Centers for Disease Control and Prevention (CDC) to classify HIV disease and infection. The clinical categories of HIV infection are defined as follows: Category A: Mildly symptomatic, Category B: Moderately symptomatic, Category C: Severely symptomatic. The Baseline value was defined as the latest pre-dose assessment (Day 1) value. Only those participants available at the specified time points were analyzed. Different participants may have been analyzed at different time points, so the overall number of participants analyzed reflects everyone in the ITT population. (NCT01910402)
Timeframe: Up to week 432

InterventionParticipants (Count of Participants)
CDC Class A to CDC Class CCDC Class B to CDC Class CCDC Class C to new CDC Class CCDC Class A, B or C to Death
DTG 50 mg/ABC 600 mg/3TC 300 mg QD6102

[back to top]

Percentage of Participants in Part 1a With Plasma HIV-1 RNA Suppression at End of Treatment and 12 Weeks Post-Treatment

HIV virologic success was defined as HIV-1 RNA suppression (HIV-1 RNA value < 40 copies/mL). (NCT01939197)
Timeframe: End of treatment: HIV Week 12 window for 12-weeks of treatment (Treatment Day 71 - 98) or HIV Week 24 window (Treatment Day 155 - 182) for 24-weeks of treatment. Post-Treatment Week 12 (PTW12): HIV PTW12 window (Post-Treatment Day 57 - 126)

,
Interventionpercentage of participants (Number)
End of TreatmentPost-Treatment Week 12
Part 1a: Arm A93.596.8
Part 1a: Arm B90.693.8

[back to top]

Percentage of Participants in Part 1b With On-Treatment HCV Virologic Failure During the Treatment Period

Percentage of participants with on-treatment HCV virologic failure during the treatment period for each arm and overall in Part 1b. Virologic failure is defined as confirmed quantifiable HCV RNA among participants with previously unquantifiable HCV RNA during treatment. (NCT01939197)
Timeframe: up to 12 weeks

Interventionpercentage of participants (Number)
Part 1b: Arm C0
Part 1b: Arm D0
Part 1b: Total0

[back to top]

Percentage of Participants in Part 1b Achieving SVR12

SVR12 is defined as plasma HCV RNA < LLOQ 12 weeks after the last dose of study drug without any confirmed quantifiable (≥ LLOQ) post-treatment value before or during that SVR window. The 95% CI is calculated using the Wilson score method for binomial distribution. (NCT01939197)
Timeframe: 12 weeks after last dose of study drug

Interventionpercentage of participants (Number)
Part 1b: Arm C100
Part 1b: Arm D100
Part 1b: Total100

[back to top]

Percentage of Participants in Part 1a With Relapse12

Percentage of participants who experienced Relapse12 among participants who completed treatment with HCV RNA < LLOQ at final treatment visit and had at least one post-treatment HCV RNA value. Relapse12 is defined as confirmed HCV RNA ≥ LLOQ between end of treatment and 12 weeks after last actual dose of study drug (up to and including the SVR12 assessment time point) for a participant with HCV RNA < LLOQ at final treatment visit who completed treatment and had post-treatment data. Completion of treatment is defined as study drug duration ≥ 77 days for Arm A and ≥ 154 days for Arm B. The 95% CI is calculated using Wilson score method for the binomial distribution. (NCT01939197)
Timeframe: up to 12 or 24 weeks, based on treatment duration, after the last actual dose of study drug

Interventionpercentage of participants (Number)
Part 1a: Arm A3.3
Part 1a: Arm B0

[back to top]

Percentage of Participants in Part 1b With Relapse12 for Each Arm and Overall

Percentage of participants who experienced Relapse12 among participants who completed treatment with HCV RNA < LLOQ at final treatment visit and had at least one post-treatment HCV RNA value. Relapse12 is defined as confirmed HCV RNA ≥ LLOQ between end of treatment and 12 weeks after last actual dose of study drug (up to and including the SVR12 assessment time point) for a participant with HCV RNA < LLOQ at final treatment visit who completed treatment and had post-treatment data. Completion of treatment is defined as study drug duration ≥ 77 days for Arm C and Arm D. The 95% CI is calculated using Wilson score method for the binomial distribution. (NCT01939197)
Timeframe: up to 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Part 1b: Arm C0
Part 1b: Arm D0
Part 1b: Total0

[back to top]

Percentage of Participants in Part 2 With Relapse12

Percentage of participants who experienced Relapse12 among those who completed treatment with HCV RNA < LLOQ at final treatment visit and had ≥1 post-treatment HCV RNA value. Relapse12=confirmed HCV RNA ≥ LLOQ between end of treatment and 12 weeks after last actual dose of study drug (up to and including the SVR12 window) for a participant with HCV RNA < LLOQ at final treatment visit who completed treatment and had post-treatment data, excluding reinfection. Completion of treatment=study drug duration ≥ 77 days for participants who received 12 weeks of treatment and ≥154 days for participants who received 24 weeks of treatment. HCV reinfection=confirmed HCV RNA ≥ LLOQ after the end of treatment in a subject who had HCV RNA < LLOQ at final treatment visit, along with the post-treatment detection of a different HCV genotype, subtype, or clade compared with baseline, as determined by phylogenetic analysis. The 95% CI is calculated using Wilson score method for the binomial distribution. (NCT01939197)
Timeframe: up to 12 or 24 weeks, based on treatment duration, after the last actual dose of study drug

Interventionpercentage of participants (Number)
Part 2: GT1 Analysis Group0.5
Part 2: Arm E0
Part 2: Arm F0
Part 2: Arm I0.8
Part 2: Arm J0
Part 2: GT4 Analysis Group0
Group 2: Arm K0

[back to top]

Percentage of Participants in Arm F and Arm G of Part 2 Achieving SVR12

SVR12 is defined as plasma HCV RNA < LLOQ 12 weeks after the last dose of study drug without any confirmed quantifiable (≥ LLOQ) post-treatment value before or during that SVR window. The 95% CI is calculated using the Wilson score method for binomial distribution. (NCT01939197)
Timeframe: 12 weeks after last dose of study drug

Interventionpercentage of participants (Number)
Part 2: Arm F75.0
Part 2: Arm G80.0

[back to top]

Percentage of Participants in Part 1a With On-Treatment HCV Virologic Failure During the Treatment Period

Percentage of participants with on-treatment HCV virologic failure during the treatment period for each arm in Part 1a. Virologic failure is defined as confirmed quantifiable HCV RNA among participants with previously unquantifiable HCV RNA during treatment. (NCT01939197)
Timeframe: up to 12 or 24 weeks, based on treatment duration

Interventionpercentage of participants (Number)
Part 1a: Arm A0
Part 1a: Arm B3.1

[back to top]

Percentage of Participants in GT1 Analysis Group 1 in Part 2 Achieving Sustained Virologic Response 12 Weeks Post-Treatment (SVR12)

"SVR12 is defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) less than the lower limit of quantification (< LLOQ) 12 weeks after the last dose of study drug without any confirmed quantifiable (≥ LLOQ) post-treatment value before or during that SVR window. The 95% confidence interval (CI) is calculated using the Wilson score method for binomial distribution.~The primary efficacy endpoint was the non-inferiority of the percentage of participants in the GT1 Analysis Group in Part 2 achieving SVR12 compared to the historical SVR12 rate for sofosbuvir plus ribavirin (a non-inferiority threshold of the lower bound of the 95% CI of 74%)." (NCT01939197)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Part 2: GT1 Analysis Group97.0

[back to top]

Percentage of Participants in Part 2 With Plasma HIV-1 RNA Suppression at End of Treatment and 12 Weeks Post-Treatment

HIV virologic success was defined as HIV-1 RNA suppression (HIV-1 RNA value < 40 copies/mL). (NCT01939197)
Timeframe: End of treatment: HIV Week 12 window for 12-weeks of treatment (Treatment Day 71 - 98) or HIV Week 24 window (Treatment Day 155 - 182) for 24-weeks of treatment. PTW12: HIV PTW12 window (Post-Treatment Day 57 - 126)

,,,,,,
Interventionpercentage of participants (Number)
End of TreatmentPost-Treatment Week 12
Group 2: Arm K85.792.9
Part 2: Arm E90.597.6
Part 2: Arm F100100
Part 2: Arm I89.691.9
Part 2: Arm J78.989.5
Part 2: GT1 Analysis Group8993
Part 2: GT4 Analysis Group85.792.9

[back to top]

Percentage of Participants in Part 1b With Plasma HIV-1 RNA Suppression at End of Treatment and 12 Weeks Post-Treatment

HIV virologic success was defined as HIV-1 RNA suppression (HIV-1 RNA value < 40 copies/mL). (NCT01939197)
Timeframe: End of treatment: HIV Week 12 window (Treatment Day 78 - 98). PTW12: HIV PTW12 window (Post-Treatment Day 57 - 126)

,,
Interventionpercentage of participants (Number)
End of TreatmentPost-Treatment Week 12
Part 1b Total90.986.4
Part 1b: Arm C100100
Part 1b: Arm D83.375.0

[back to top]

Percentage of Participants With GT4 HCV in Part 2 Achieving SVR12, by Arm and Overall

SVR12 is defined as plasma HCV RNA < LLOQ 12 weeks after the last dose of study drug without any confirmed quantifiable (≥ LLOQ) post-treatment value before or during that SVR window. The 95% CI is calculated using the Wilson score method for binomial distribution. (NCT01939197)
Timeframe: 12 weeks after last dose of study drug

Interventionpercentage of participants (Number)
Part 2: GT4 Analysis Group96.4
Part 2: Arm K96.4

[back to top]

Percentage of Participants in Part 1a Achieving SVR12

SVR12 is defined as plasma HCV RNA < LLOQ 12 weeks after the last dose of study drug without any confirmed quantifiable (≥ LLOQ) post-treatment value before or during that SVR window. The 95% CI is calculated using the Wilson score method for binomial distribution. (NCT01939197)
Timeframe: 12 weeks after last dose of study drug

Interventionpercentage of participants (Number)
Part 1a: Arm A93.5
Part 1a: Arm B90.6

[back to top]

Percentage of Participants in Part 2 With On-Treatment HCV Virologic Failure During the Treatment Period

Percentage of participants with on-treatment HCV virologic failure during the treatment period for arms in Part 2. Virologic failure is defined as confirmed quantifiable HCV RNA among participants with previously unquantifiable HCV RNA during treatment. (NCT01939197)
Timeframe: up to 12 or 24 weeks, based on treatment duration

Interventionpercentage of participants (Number)
Part 2: GT1 Analysis Group0.5
Part 2: Arm E0
Part 2: Arm F25.0
Part 2: Arm I0
Part 2: Arm J0
Part 2: GT4 Analysis Group0
Group 2: Arm K0

[back to top]

Percentage of Non-cirrhotic Treatment-Naïve Participants Who Are Eligible for Interferon (IFN)-Based Therapy and Who Have High Viral Load in the DB Active Treatment Group With a Sustained Virologic Response 12 Weeks Post-treatment

The percentage noncirrhotic treatment-naïve participants who were eligible for IFN-based therapy and who had high viral load at baseline (HCV RNA ≥ 100,000 IU/mL) in the DB active treatment group with sustained virologic response (plasma Hepatitis C virus ribonucleic acid [HCV RNA] level less than the lower limit of quantitation [< LLOQ]) 12 weeks after the last dose of active study drug (SVR12). Among noncirrhotic treatment-naïve participants who were eligible for IFN-based therapy and who had high viral load at baseline, superiority of Arm A to a clinically relevant threshold based on historical SVR rate with telaprevir plus pegylated interferon alpha/ribavirin (pegIFN/RBV) in treatment-naïve, non-cirrhotic patients with high viral load; the lower bound of 95% confidence interval (LCB) had to exceed 63% to achieve superiority. The 95% confidence interval was calculated using the normal approximation to the binomial distribution. (NCT02023099)
Timeframe: 12 weeks after the last dose of study drug

Interventionpercentage of participants (Number)
Substudy 1, Arm A: DB 2-DAA94.6

[back to top]

Percentage of Participants in the Active Treatment Group With On-treatment Virologic Failure During Treatment

"On-treatment virologic failure among all randomized non-cirrhotic participants who received at least one dose of DB ABT-450/r/ABT-267 and all enrolled participants with compensated cirrhosis who received at least one dose of OL ABT-450/r/ABT-267. On-treatment virologic failure is defined as the occurrence of at least one of the following:~confirmed HCV RNA ≥ LLOQ (defined as 2 consecutive HCV RNA measurements ≥ LLOQ) at any point during treatment after HCV RNA < LLOQ (rebound), or~confirmed increase from nadir in HCV RNA (defined as 2 consecutive HCV RNA measurements > 1 log10 IU/mL above nadir) at any time point during treatment (rebound), or~HCV RNA ≥ LLOQ persistently during treatment with at least 6 weeks (≥ 36 days) of treatment (failure to suppress).~The 95% confidence interval was calculated using the Wilson's score method." (NCT02023099)
Timeframe: up to 12 weeks

Interventionpercentage of participants (Number)
Substudy 1, Arm A: DB 2-DAA0.5
Substudy 2, Arm C: OL 2-DAA2.4

[back to top]

Percentage of Participants in the Active Treatment Group With Sustained Virologic Response 12 Weeks Post-treatment

Sustained virologic response (plasma HCV RNA level < LLOQ) 12 weeks after the last dose of study drug for all randomized non-cirrhotic participants who received at least one dose of DB ABT-450/r/ABT-267 and for all enrolled participants with compensated cirrhosis who received at least one dose of open-label ABT-450/r/ABT-267. The 95% confidence interval was calculated using the Wilson's score method. (NCT02023099)
Timeframe: 12 weeks after last dose of study drug

Interventionpercentage of participants (Number)
Substudy 1, Arm A: DB 2-DAA94.9
Substudy 2, Arm C: OL 2-DAA90.5

[back to top]

Percentage of Participants in Substudy 1 Arm A Active Treatment Group With Post-treatment Relapse, by Subpopulation

"Post-treatment relapse among all randomized non-cirrhotic participants who received at least one dose of DB ABT-450/r/ABT-267 and completed treatment, in the following subpopulations: noncirrhotic treatment-naïve (T-naïve) participants with a high baseline (BL) viral load (HCV RNA ≥ 100,000 IU/mL) who are eligible for IFN-based therapy (IFN-BT), a low BL viral load (HCV RNA < 100,000 IU/mL), or who are ineligible for IFN-BT; noncirrhotic treatment-experienced (T-exp) participants who relapsed after prior IFN-BT, who were nonresponders to prior IFN-BT, or who were intolerant to IFN-BT.~Relapse was defined as confirmed HCV RNA ≥ LLOQ (defined as 2 consecutive HCV RNA measurements ≥ LLOQ) between the final treatment visit and 12 weeks after the last dose of study drugs among participants completing treatment and with HCV RNA < LLOQ at the final treatment visit and at least one post-treatment HCV RNA value. The 95% confidence interval was calculated using the Wilson's score method." (NCT02023099)
Timeframe: within 12 weeks after last dose of study drug

Interventionpercentage of participants (Number)
T-naïve: high BL viral load, IFN-eligible; n=109T-naïve: low BL viral load; n=6T-naïve: IFN-ineligible; n=22T-exp. w/prior IFN-BT: relapser; n=21T-exp. w/prior IFN-BT: nonresponder; n=28T-exp. w/prior IFN-BT: IFN-intolerant; n=25
Substudy 1, Arm A: DB 2-DAA2.804.5004.0

[back to top]

Percentage of Participants in Substudy 1 Arm A Active Treatment Group With Sustained Virologic Response 12 Weeks Post-Treatment, by Subpopulation

Sustained virologic response (plasma HCV RNA level < LLOQ) 12 weeks after the last dose of study drug for all randomized non-cirrhotic participants who received at least one dose of DB ABT-450/r/ABT-267 in the following subpopulations: noncirrhotic T-naïve participants with a high BL viral load (HCV RNA ≥ 100,000 IU/mL) who are eligible for IFN-BT; noncirrhotic T-naïve participants with low BL viral load (HCV RNA < 100,000 IU/mL); noncirrhotic T-naïve participants who are ineligible for IFN-BT; noncirrhotic T-exp participants who relapsed after prior IFN-BT; noncirrhotic T-exp participants who were nonresponders to prior IFN-BT; noncirrhotic T-exp participants who were intolerant to IFN-BT. The 95% confidence interval was calculated using the Wilson's score method. (NCT02023099)
Timeframe: 12 weeks after last dose of study drug

Interventionpercentage of participants (Number)
T-naïve: all; n=139T-naïve: high BL viral load, IFN-eligible; n=112T-naïve: low BL viral load; n=6T-naïve: IFN-ineligible; n=23T-exp. w/prior IFN-BT: all; n=76T-exp. w/prior IFN-BT: relapser; n=22T-exp. w/prior IFN-BT: nonresponder; n=28T-exp. w/prior IFN-BT: IFN-intolerant; n=26
Substudy 1, Arm A: DB 2-DAA94.294.610091.396.195.510092.3

[back to top]

Percentage of Participants in the Substudy 1 Arm A Active Treatment Group With On-treatment Virologic Failure During Treatment, by Subpopulation

On-treatment virologic failure among all randomized non-cirrhotic participants who received at least one dose of DB ABT-450/r/ABT-267 in the following subpopulations: noncirrhotic treatment-naïve (T-naïve) participants with a high baseline (BL) viral load (HCV RNA ≥ 100,000 IU/mL) who are eligible for IFN-based therapy (IFN-BT), a low viral load (HCV RNA < 100,000 IU/mL), or who are ineligible for IFN-BT; noncirrhotic treatment-experienced (T-exp) participants who relapsed after prior IFN-BT, who were nonresponders to prior IFN-BT, or who were intolerant to IFN-BT. On-treatment virologic failure is defined in Outcome measure 2. The 95% confidence interval was calculated using the Wilson's score method. (NCT02023099)
Timeframe: up to 12 weeks

Interventionpercentage of participants (Number)
T-naïve: high BL viral load, IFN-eligible; n=112T-naïve: low BL viral load; n=6T-naïve: IFN-ineligible; n=23T-exp. w/prior IFN-BT: relapser; n=22T-exp. w/prior IFN-BT: nonresponder; n=28T-exp. w/prior IFN-BT: IFN-intolerant; n=26
Substudy 1, Arm A: DB 2-DAA000003.8

[back to top]

Percentage of Participants in the Active Treatment Group With Post-treatment Relapse

Post-treatment relapse among all randomized non-cirrhotic participants who received at least one dose of DB ABT-450/r/ABT-267 and completed treatment, and all enrolled participants with compensated cirrhosis who received at least one dose of OL ABT-450/r/ABT-267 and completed treatment. Relapse was defined as confirmed HCV RNA ≥ LLOQ (defined as 2 consecutive HCV RNA measurements ≥ LLOQ) between the final treatment visit and 12 weeks after the last dose of study drugs among participants completing treatment and with HCV RNA < LLOQ at the final treatment visit and at least one post-treatment HCV RNA value. The 95% confidence interval was calculated using the Wilson's score method. (NCT02023099)
Timeframe: within 12 weeks after last dose of study drug

Interventionpercentage of participants (Number)
Substudy 1, Arm A: DB 2-DAA2.4
Substudy 2, Arm C: OL 2-DAA5.0

[back to top]

Percentage of Participants With SVR12 Weeks Post-treatment for Each Treatment Arm Within Different Subpopulations

The percentage of participants with SVR12 in each treatment arm within the following subpopulations: noncirrhotic participants; noncirrhotic treatment-experienced (T-exp) participants; noncirrhotic participants who relapsed after prior IFN-based therapy (relapsers); noncirrhotic T-exp participants who were non-responders to prior IFN-based therapy; noncirrhotic T-exp participants who were intolerant to IFN-based therapy; participants with compensated cirrhosis. (NCT02023112)
Timeframe: 12 weeks after last dose of study drug

,
Interventionpercentage of participants (Number)
All participants; n=85, 86Noncirrhotic participants; n=80, 80Noncirrhotic T-exp; n=32, 33Noncirrhotic T-exp Relapser; n=15, 16Noncirrhotic T-exp Nonresponder; n=5, 6Noncirrhotic T-exp IFN-intolerant; n=12, 11Cirrhotic Participants; n=5, 6
ABT-450/r/ABT-267 Plus RBV for 12 Weeks72.972.568.880.040.066.780.0
ABT-450/r/ABT-267 Plus RBV for 16 Weeks81.485.075.893.850.063.633.3

[back to top]

Percentage of Participants in Each Treatment Arm With On-treatment Virologic Failure During the Treatment Period

On-treatment virologic failure was defined as rebound (confirmed HCV RNA ≥ LLOQ after HCV RNA < LLOQ during treatment, or confirmed increase from nadir in HCV RNA [> 1 log10 IU/mL above nadir] at any time point during treatment) or failure to suppress HCV during treatment (all on-treatment values of HCV RNA ≥ LLOQ with at least 6 weeks of treatment). (NCT02023112)
Timeframe: 12 or 16 weeks (end of treatment period)

,
Interventionpercentage of participants (Number)
OverallReboundFailure to suppress
ABT-450/r/ABT-267 Plus RBV for 12 Weeks8.38.30
ABT-450/r/ABT-267 Plus RBV for 16 Weeks8.58.50

[back to top]

Percentage of Participants With Post-treatment Relapse

Relapse by post-treatment Week 12 was defined as confirmed HCV RNA ≥ LLOQ between end of treatment and 12 weeks after last actual dose of study drug for a participant with HCV RNA < LLOQ at the final treatment visit and who completed study treatment. Completion of treatment was defined as a study drug duration ≥ 77 days for the 12-week treatment arm or ≥ 105 days for the 16-week treatment arm. (NCT02023112)
Timeframe: within 12 weeks after the last dose of study drug

Interventionpercentage of participants (Number)
ABT-450/r/ABT-267 Plus RBV for 12 Weeks12.2
ABT-450/r/ABT-267 Plus RBV for 16 Weeks0

[back to top]

Percentage of Non-cirrhotic, Treatment-naive Participants in Each Treatment Group With a Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

The percentage of participants with sustained virologic response (plasma Hepatitis C virus ribonucleic acid [HCV RNA] level less than the lower limit of quantitation [< LLOQ]) 12 weeks after the last dose of study drug. (NCT02023112)
Timeframe: 12 weeks after last dose of study drug

Interventionpercentage of participants (Number)
ABT-450/r/ABT-267 Plus RBV for 12 Weeks75.0
ABT-450/r/ABT-267 Plus RBV for 16 Weeks91.5

[back to top]

Percentage of Participants in Each Treatment Arm With On-treatment Virologic Failure During the Treatment Period for Each Treatment Arm Within Different Subpopulations

"The percentage of participants with on-treatment virologic failure in each treatment arm within the following subpopulations: noncirrhotic participants; noncirrhotic treatment-experienced (T-exp) participants; participants with compensated cirrhosis.~On-treatment virologic failure was defined as rebound (confirmed HCV RNA ≥ LLOQ after HCV RNA < LLOQ during treatment, or confirmed increase from nadir in HCV RNA [> 1 log10 IU/mL above nadir] at any time point during treatment) or failure to suppress HCV during treatment (all on-treatment values of HCV RNA ≥ LLOQ with at least 6 weeks of treatment)." (NCT02023112)
Timeframe: 12 or 16 weeks (end of treatment period)

,
Interventionpercentage of participants (Number)
All Participants: Overall; n=85, 86All Participants: Rebound; n=85, 86All Participants: Failure to Suppress; n=85, 86Noncirrhotic: Overall; n=80, 80Noncirrhotic: Rebound; n=80, 80Noncirrhotic: Failure to Suppress; n=80, 80Noncirrhotic T-exp: Overall; n=32, 33Noncirrhotic T-exp: Rebound; n=32, 33Noncirrhotic T-exp: Failure to Suppress; n=32, 33Cirrhotic: Overall; n=5, 6Cirrhotic: Rebound; n=5, 6Cirrhotic: Failure to Suppress; n=5, 6
ABT-450/r/ABT-267 Plus RBV for 12 Weeks15.315.34.715.015.03.825.025.09.420.020.020.0
ABT-450/r/ABT-267 Plus RBV for 16 Weeks16.315.14.713.812.53.821.218.29.150.050.016.7

[back to top]

Percentage of Participants With Post-treatment Relapse Within Different Subpopulations

"The percentage of participants with relapse by post-treatment Week 12 in each treatment arm within the following subpopulations: noncirrhotic participants; noncirrhotic treatment-experienced (T-exp) participants; participants with compensated cirrhosis.~Relapse by post-treatment Week 12 was defined as confirmed HCV RNA ≥ LLOQ between end of treatment and 12 weeks after last actual dose of study drug for a participant with HCV RNA < LLOQ at the final treatment visit and who completed study treatment. Completion of treatment was defined as a study drug duration ≥ 77 days for the 12-week treatment arm or ≥ 105 days for the 16-week treatment arm." (NCT02023112)
Timeframe: within 12 weeks after the last dose of study drug

,
Interventionpercentage of participants (Number)
All Participants; n=69, 70Noncirrhotic Participants; n=65, 68Noncirrhotic T-exp Participants; n=24, 25Cirrhotic Participants; n=4, 2
ABT-450/r/ABT-267 Plus RBV for 12 Weeks10.110.88.30
ABT-450/r/ABT-267 Plus RBV for 16 Weeks0000

[back to top]

The Percentage of Subjects With Post-Treatment Relapse

Percentage of subjects with confirmed quantifiable HCV RNA within 12 weeks of last dose among subjects with unquantifiable hepatitis C virus ribonucleic acid at the end of treatment. (NCT02068222)
Timeframe: Within 12 weeks after the last dose of study drug

Interventionpercentage of participants (Number)
ABT-450/r and ABT-530 Plus RBV0

[back to top]

The Percentage of Subjects With Virologic Failure During Treatment

Percentage of subjects with quantifiable HCV RNA throughout the entire treatment period, confirmed quantifiable HCV RNA after previously having unquantifiable HCV RNA, or a confirmed increase of at least one log10 in HCV RNA during treatment. (NCT02068222)
Timeframe: Up to Treatment Week 12

Interventionpercentage of participants (Number)
ABT-450/r and ABT-530 Plus RBV10

[back to top]

The Percentage of Subjects Who Achieve 24-week Sustained Virologic Response (SVR24)

SVR24 defined as HCV RNA LLOQ 24 weeks after last dose of study drug. (NCT02068222)
Timeframe: 24 weeks after last dose of study drug

Interventionpercentage of participants (Number)
ABT-450/r and ABT-530 Plus RBV90

[back to top]

The Percentage of Subjects Who Achieve 12-week Sustained Virologic Response (SVR12)

SVR12 defined as hepatitis C (HCV) ribonucleic acid (RNA) less than the lower limit of quantification (LLOQ) 12 weeks after the last actual dose of study drug. (NCT02068222)
Timeframe: 12 weeks after last dose of study drug

Interventionpercentage of participants (Number)
ABT-450/r and ABT-530 Plus RBV90

[back to top]

Change From Baseline in Estimated Glomerular Filtration Rate (eGFR)

Glomerular Filtration Rate (eGFR) was estimated from the Modification of Diet in Renal Disease (MDRD)-6 equation. The MDRD-6 equation = 198 × [serum creatinine(mg/dL)]^-0.858 × [age]-0.167 × [0.822 if patient is female] × [1.178 if patient is black] × [serum urea nitrogen concentration (mg/dL)]^-0.293 × [urine urea nitrogen excretion (g/d)]^0.249. (NCT02116660)
Timeframe: Baseline and Week 48

InterventionmL/min (Mean)
Raltegravir Plus Nevirapine Plus Lamivudine-1.1
Protease Inhibitor/Ritonavir Plus Tenofovir/Emtricitabine-5.5

[back to top]

HIV-1 RNA Viral Load

Percentage of subjects who have plasma HIV-1 RNA levels <400 cps/ml after 24 weeks of follow-up following a switch to DRV/r monotherapy versus continuing triple therapy containing 2 NRTIs + LPV/r (or ATV/r) (FDA Snapshot method). The FDA 'Snapshot' algorithm evaluates HIV RNA response using only the results at the week 24 time-point which also means that rebound at earlier time-points are not classified as treatment failure, unless it lead to discontinuation prior to the week 48. (NCT02155101)
Timeframe: 24 weeks

InterventionParticipants (Count of Participants)
ART With 2 NRTIs Plus LPV/r (or ATV/r)37
Darunavir72

[back to top]

HIV-1 RNA Viral Load

"Percentage of subjects who have plasma HIV-1 RNA levels <50 cps/ml after 12 weeks of follow-up following a switch to DRV/r monotherapy versus continuing triple therapy containing 2 NRTIs + LPV/r (or ATV/r), using the FDA Time to Loss of Virologic Response method. The FDA 'Snapshot' algorithm evaluates HIV RNA response using only the results at the week 12 time-point which also means that rebound at earlier time-points are not classified as treatment failure, unless it lead to discontinuation prior to the week 48." (NCT02155101)
Timeframe: 12 weeks

InterventionParticipants (Count of Participants)
ART With 2 NRTIs Plus LPV/r (or ATV/r)35
Darunavir73

[back to top]

HIV-1 RNA Viral Load

"Percentage of subjects who have plasma HIV-1 RNA levels <50 cps/ml after 24 weeks of follow-up following a switch to DRV/r monotherapy versus continuing triple therapy containing 2 NRTIs + LPV/r (or ATV/r), using the FDA Time to Loss of Virologic Response method. The FDA 'Snapshot' algorithm evaluates HIV RNA response using only the results at the week 24 time-point which also means that rebound at earlier time-points are not classified as treatment failure, unless it lead to discontinuation prior to the week 48." (NCT02155101)
Timeframe: 24 weeks

InterventionParticipants (Count of Participants)
ART With 2 NRTIs Plus LPV/r (or ATV/r)36
Darunavir62

[back to top]

Plasma Concentration Observed at 24 Hours Postdose (C24) of Daclatasvir

C24 was obtained from concentration time plot of daclatasvir by using noncompartmental method by a validated pharmacokinetic analysis program. (NCT02159352)
Timeframe: Predose (0 hour) on Day 2, 3 and 0, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24 hours on Day 4 (Period 1); Predose (0 hour) on Day 12, 13 and 0, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24 hour on Day 14 (Period 2)

Interventionng/mL (Geometric Mean)
Group 1: Daclatasvir (60 mg)225
Group 1: Daclatasvir (30 mg) + Darunavir/Ritonavir250
Group 2: Daclatasvir (60 mg)225
Group 2: Daclatasvir (30 mg) + Lopinavir/Ritonavir280

[back to top]

Time of Maximum Observed Plasma Concentration (Tmax) of Daclatasvir

Tmax was obtained from concentration-time plot of daclatasvir by using non-compartmental method by a validated pharmacokinetic analysis program. (NCT02159352)
Timeframe: Predose (0 hour) on Day 2, 3 and 0, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24 hours on Day 4 (Period 1); Predose (0 hour) on Day 12, 13 and 0, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24 hour on Day 14 (Period 2)

Interventionhours (Median)
Group 1: Daclatasvir (60 mg)2.00
Group 1: Daclatasvir (30 mg) + Darunavir/Ritonavir3.00
Group 2: Daclatasvir (60 mg)2.00
Group 2: Daclatasvir (30 mg) + Lopinavir/Ritonavir2.00

[back to top]

Dose-normalized Maximum Observed Plasma Concentration (Cmax/D) and Dose-normalized Plasma Concentration Observed at 24 Hours Postdose (C24/D) of Daclatasvir

Cmax/D and C24/D are obtained from concentration-time plot of daclatasvir by using noncompartmental method by a validated pharmacokinetic analysis program. (NCT02159352)
Timeframe: Predose (0 hour) on Day 2, 3 and 0, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24 hours on Day 4 (Period 1); Predose (0 hour) on Day 12, 13 and 0, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24 hour on Day 14 (Period 2)

,,,
Interventionng/mL/mg (Geometric Mean)
Cmax/DC24/D
Group 1: Daclatasvir (30 mg) + Darunavir/Ritonavir16.48.34
Group 1: Daclatasvir (60 mg)22.23.75
Group 2: Daclatasvir (30 mg) + Lopinavir/Ritonavir15.99.33
Group 2: Daclatasvir (60 mg)23.53.76

[back to top]

Number of Participants With Abnormalities in Electrocardiogram (ECG) Findings

Abnormalities in ECG findings included: PR ≥210 msec, QRS ≥120 msec, QT ≥500 msec, QTcF ≥450 msec, and second- or third-degree heart block. (NCT02159352)
Timeframe: From start of study treatment (Day 1) to study discharge (up to 15 days)

,,,
Interventionparticipants (Number)
PR >210QRS >120QT >500QTcF >450
Group 1: Daclatasvir (30 mg) + Darunavir/Ritonavir4001
Group 1: Daclatasvir (60 mg)2000
Group 2: Daclatasvir (30 mg) + Lopinavir/Ritonavir1000
Group 2: Daclatasvir (60 mg)0000

[back to top]

Number of Participants With Serious Adverse Events (SAEs) and Discontinuations Due to Adverse Events (AEs) and Who Died

AE was defined as any new unfavorable symptom, sign, or disease or worsening of a preexisting condition that does not necessarily have a causal relationship with treatment. SAE was defined as a medical event that at any dose resulted in death, persistent or significant disability/incapacity, or drug dependency/abuse; was life threatening, an important medical event, or a congenital anomaly/birth defect; or required or prolonged hospitalization. (NCT02159352)
Timeframe: From start of study treatment (Day 1) to study discharge for AEs (up to 15 days); Day 1 to 30 days after last dose of study treatment for SAEs (up to 44 days)

,,,
Interventionparticipants (Number)
SAEDiscontinued due to AEsDeath
Group 1: Daclatasvir (30 mg) + Darunavir/Ritonavir030
Group 1: Daclatasvir (60 mg)000
Group 2: Daclatasvir (30 mg) + Lopinavir/Ritonavir010
Group 2: Daclatasvir (60 mg)000

[back to top]

Dose-normalized Area Under the Concentration-Time Curve in 1 Dosing Interval (AUC[TAU]/D) of Daclatasvir

AUC(TAU)/D was obtained from concentration-time plot of daclatasvir by using noncompartmental method by a validated pharmacokinetic analysis program. (NCT02159352)
Timeframe: Predose (0 hour) on Day 2, 3 and 0, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24 hours on Day 4 (Period 1); Predose (0 hour) on Day 12, 13 and 0, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24 hour on Day 14 (Period 2)

Intervention(ng*h/mL)/mg (Geometric Mean)
Group 1: Daclatasvir (60 mg)211
Group 1: Daclatasvir (30 mg) + Darunavir/Ritonavir276
Group 2: Daclatasvir (60 mg)230
Group 2: Daclatasvir (30 mg) + Lopinavir/Ritonavir262

[back to top]

Maximum Observed Plasma Concentration (Cmax) for Daclatasvir

Cmax was obtained from concentration-time plot using a noncompartmental method and a validated pharmacokinetic analysis program. (NCT02159352)
Timeframe: Predose (0 hour) on Day 2, 3 and 0, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24 hours on Day 4 (Period 1); Predose (0 hour) on Day 12, 13 and 0, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24 hour on Day 14 (Period 2)

Interventionng/mL (Geometric Mean)
Group 1: Daclatasvir (60 mg)1335
Group 1: Daclatasvir (30 mg) + Darunavir/Ritonavir493
Group 2: Daclatasvir (60 mg)1412
Group 2: Daclatasvir (30 mg) + Lopinavir/Ritonavir476

[back to top]

Number of Participants With Abnormalities in Vital Sign Measurements

Criteria for abnormalities in vital sign measurements: Diastolic blood pressure: Value >90 and change from baseline > 0 or value < 55 and change from baseline <-10. Systolic blood pressure: Value >140 and change from baseline >20 or value <90 and change from baseline <-20. Heart rate: Value >100 and change from baseline >30 or value <55 and change from baseline <-15. Respiration: Value >16 or change from baseline >10. Temperature: Value >38.3°C or change from baseline >1.6°C. (NCT02159352)
Timeframe: From start of study treatment (Day 1) to study discharge (up to 15 days)

Interventionparticipants (Number)
Group 1: Daclatasvir + Darunavir/Ritonavir3
Group 2: Daclatasvir + Lopinavir/Ritonavir2

[back to top]

Number of Participants With Marked Abnormalities in Hematology Laboratory Test Results

Criteria for marked abnormalities in test results: Platelet count >1.5*upper limits of normal (ULN) value, >1.5*ULN if pretreatment (PreRx) value is missing, <0.85*lower limit of normal (LLN) if PreRx ≥LLN, <0.85*LLN if PreRx is missing, <0.85*PreRx if PreRx 1.2*ULN if LLN ≤PreRx ≤ULN, >1.2*ULN if PreRx is missing, >1.5*PreRx if PreRx >ULN, >ULN if PreRx ULN. Lymphocytes >7.5*10^3 c/uL and <0.75*10^3 c/uL. Neutrophils <0.85*PreRx if PreRx <1.5*ULN, <1.5*ULN if PreRx ≥1.5*ULN and <1.5*ULN if PreRx is missing. (NCT02159352)
Timeframe: From start of study treatment (Day 1) to study discharge (up to 15 days)

,,,
Interventionparticipants (Number)
Platelet CountLeukocytesNeutrophils (absolute)Lymphocytes (absolute)
Group 1: Daclatasvir (30 mg) + Darunavir/Ritonavir0121
Group 1: Daclatasvir (60 mg)1000
Group 2: Daclatasvir (30 mg) + Lopinavir/Ritonavir0100
Group 2: Daclatasvir (60 mg)0110

[back to top]

Area Under the Concentration-Time Curve in 1 Dosing Interval (AUC[TAU]) for Daclatasvir

AUC(TAU) was the area under the curve from time zero to end of dosing interval. AUC(TAU) was obtained from concentration-time plot of daclatasvir using noncompartmental method and a validated pharmacokinetic analysis program. (NCT02159352)
Timeframe: Predose (0 hour) on Day 2, 3 and 0, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24 hours on Day 4 (Period 1); Predose (0 hour) on Day 12, 13 and 0, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24 hour on Day 14 (Period 2)

Interventionng*h/mL (Geometric Mean)
Daclatasvir (60 mg) Days 1-412677
Daclatasvir (30 mg) + Darunavir/Ritonavir Days 5-148295
Daclatasvir (60 mg) Days 5-1413799
Daclatasvir (30 mg) + Lopinavir/Ritonavir Days 5-147855

[back to top]

Number of Participants With Abnormalities in Urinalysis and Other Chemistry Testing Results

Criteria for marked abnormalities on laboratory test results: urinary dipstick blood: ≥2 if pretreatment (PreRx) <1, ≥2 if PreRx is missing or ≥2*PreRx if PreRx ≥1. Urinary microscopic red blood cell (RBC): ≥2 if PreRx <2, ≥2 if PreRx is missing or ≥4 if PreRx ≥2. Urinary microscopic white blood cell (WBC): ≥2 if PreRx <2, ≥2 if PreRx is missing or ≥4 if PreRx ≥2. Lactate dehydrogenase >1.25*upper limit of normal (ULN) if PreRx ≤ULN, >1.25*ULN if PreRx is missing and >1.5*PreRx if PreRx >ULN. (NCT02159352)
Timeframe: From start of study treatment (Day 1) to study discharge (up to 15 days)

,,,
Interventionparticipants (Number)
Blood, urineRBC, urineWBC, urineLactate dehydrogenase
Group 1: Daclatasvir (30 mg) + Darunavir/Ritonavir1132
Group 1: Daclatasvir (60 mg)2112
Group 2: Daclatasvir (30 mg) + Lopinavir/Ritonavir0001
Group 2: Daclatasvir (60 mg)1001

[back to top]

Mean Change From Baseline in Short Form 36 Version 2.0 (SF-36 V2) Mental Component Summary (MCS) Score at Post-Treatment Week 12 and Post-Treatment Week 24

The SF-36v2 is a non-disease specific Health Related Quality of Life (HRQoL) instrument with extensive use in multiple disease states. The SF-36v2 instrument comprises 36 total items (questions) targeting a subject's functional health and well-being in 8 domains (physical functioning, role physical, bodily pain, general health, vitality, social functioning, role emotional and mental health) with a recall period of 4 weeks. Domain scores are aggregated into a Physical Component Summary (PCS) score and a Mental Component Summary (MCS) score. SF-36v2 scores for each domain and PCS/MCS range from 0-100: higher scores indicate a better state of health. Positive numbers indicate improvement from baseline. (NCT02167945)
Timeframe: From Baseline to Post-Treatment Week 12 and Post-Treatment Week 24

,,,
Interventionunits on a scale (Mean)
At Post-treatment Week 12At Post-treatment Week 24
Baseline Fibrosis Stage F0-F12.73.2
Baseline Fibrosis Stage F21.01.9
Baseline Fibrosis Stage F30.91.5
Baseline Fibrosis Stage F40.30.4

[back to top]

Mean Change From Baseline in Short Form 36 Version 2.0 (SF-36 V2) Physical Component Summary (PCS) Score at Post-Treatment Week 12 and Post-Treatment Week 24

The SF-36v2 is a non-disease specific Health Related Quality of Life (HRQoL) instrument with extensive use in multiple disease states. The SF-36v2 instrument comprises 36 total items (questions) targeting a subject's functional health and well-being in 8 domains (physical functioning, role physical, bodily pain, general health, vitality, social functioning, role emotional and mental health) with a recall period of 4 weeks. Domain scores are aggregated into a Physical Component Summary (PCS) score and a Mental Component Summary (MCS) score. SF-36v2 scores for each domain and PCS/MCS range from 0-100: higher scores indicate a better state of health. Positive numbers indicate improvement from baseline. (NCT02167945)
Timeframe: From Baseline to Post-Treatment Week 12 and Post-Treatment Week 24

,,,
Interventionunits on a scale (Mean)
At Post-treatment Week 12At Post-treatment Week 24
Baseline Fibrosis Stage F0-F11.21.1
Baseline Fibrosis Stage F20.00.5
Baseline Fibrosis Stage F31.82.3
Baseline Fibrosis Stage F41.41.8

[back to top]

Liver Decompensation: Time to Event

Time to liver decompensation was defined as number of days from the 1st day of study drug dosing for the participant to the date of liver decompensation. All liver decompensation was to be included, regardless of whether it occurred while the participant was still taking study drug or had previously discontinued study drug. If the participant didn't experience the event of interest nor had died (all-cause death), their data was to be censored at the date of their last available assessment of clinical outcomes. For participants with no post-baseline assessment, their data was to be censored on the 1st day of study drug dosing. All-cause death was a censoring event for liver decompensation. The event-free survival rates were estimated using Kaplan-Meier methodology and incidence estimates are presented with 95% confidence intervals. The pre-specified analysis of liver decompensation included pooled data from TOPAZ-II (this study) and the companion study TOPAZ-I (M14-423; NCT02219490). (NCT02167945)
Timeframe: At Post-Treatment Weeks 52, 104, 156, 208, and 260

,
Interventionpercentage of participants (Number)
Kaplan-Meier estimate at PT Week 52Kaplan-Meier estimate at PT Week 104Kaplan-Meier estimate at PT Week 156Kaplan-Meier estimate at PT Week 208Kaplan-Meier estimate at PT Week 260
Participants in Studies M14-222 and M14-423 Who Achieved SVR120.20.20.30.30.5
Participants in Studies M14-222 and M14-423 Who Did Not Achieve SVR124.54.54.54.54.5

[back to top]

Liver Transplantation: Time to Event

Time to liver transplantation was defined as number of days from the 1st day of study drug dosing for the participant to date of liver transplantation. All liver transplantation was to be included, regardless of whether it occurred while the participant was still taking study drug or had previously discontinued study drug. If the participant didn't experience the event of interest nor had died (all-cause death), their data was to be censored at the date of their last available assessment of clinical outcomes. For participants with no post-baseline assessment, their data was to be censored on the 1st day of study drug dosing. All-cause death was a censoring event for liver transplantation. The event-free survival rates were estimated using Kaplan-Meier methodology and incidence estimates are presented with 95% confidence intervals. The pre-specified analysis of liver transplantation included pooled data from TOPAZ-II (this study) and the companion study TOPAZ-I (M14-423; NCT02219490). (NCT02167945)
Timeframe: At Post-Treatment Weeks 52, 104, 156, 208, and 260

,
Interventionpercentage of participants (Number)
Kaplan-Meier estimate at PT Week 52Kaplan-Meier estimate at PT Week 104Kaplan-Meier estimate at PT Week 156Kaplan-Meier estimate at PT Week 208Kaplan-Meier estimate at PT Week 260
Participants in Studies M14-222 and M14-423 Who Achieved SVR12000.10.10.2
Participants in Studies M14-222 and M14-423 Who Did Not Achieve SVR1200000

[back to top] [back to top]

All-Cause Death: Time to Event

Time to all-cause death was defined as the number of days from the first day of study drug dosing for the participant to the date of death. All deaths were to be included, regardless of whether the death occurred while the participant was still taking study drug or had previously discontinued study drug. If the participant did not die, their data was to be censored at the date of their last available assessment of clinical outcomes. For participants with no post-baseline assessment, the participant's data was to be censored on the first day of study drug dosing. The event-free survival rates were estimated using Kaplan-Meier methodology and incidence estimates are presented with 95% confidence intervals. The pre-specified analysis of all-cause death included pooled data from TOPAZ-II (this study) and the companion study TOPAZ-I (M14-423; NCT02219490). (NCT02167945)
Timeframe: At Post-Treatment Weeks 52, 104, 156, 208, and 260

,
Interventionpercentage of participants (Number)
Kaplan-Meier estimate at PT Week 52Kaplan-Meier estimate at PT Week 104Kaplan-Meier estimate at PT Week 156Kaplan-Meier estimate at PT Week 208Kaplan-Meier estimate at PT Week 260
Participants in Studies M14-222 and M14-423 Who Achieved SVR120.10.71.21.52.0
Participants in Studies M14-222 and M14-423 Who Did Not Achieve SVR128.38.38.38.38.3

[back to top]

Hepatocellular Carcinoma: Time to Event

Time to hepatocellular carcinoma was defined as number of days from the 1st day of study drug dosing for the participant to date of hepatocellular carcinoma. All hepatocellular carcinoma was to be included, whether it occurred while the participant was still taking study drug or had previously discontinued study drug. If the participant didn't experience the event of interest nor had died (all-cause death), their data was to be censored at the date of their last available assessment of clinical outcomes. For those with no post-baseline assessment, their data was to be censored on the 1st day of study drug dosing. All-cause death was a censoring event for hepatocellular carcinoma. The event-free survival rates were estimated using Kaplan-Meier methodology and incidence estimates are presented with 95% confidence intervals. The pre-specified analysis of hepatocellular carcinoma included pooled data from TOPAZ-II (this study) and the companion study TOPAZ-I (M14-423; NCT02219490). (NCT02167945)
Timeframe: At Post-Treatment Weeks 52, 104, 156, 208, and 260

,
Interventionpercentage of participants (Number)
Kaplan-Meier estimate at PT Week 52Kaplan-Meier estimate at PT Week 104Kaplan-Meier estimate at PT Week 156Kaplan-Meier estimate at PT Week 208Kaplan-Meier estimate at PT Week 260
Participants in Studies M14-222 and M14-423 Who Achieved SVR120.20.40.50.60.9
Participants in Studies M14-222 and M14-423 Who Did Not Achieve SVR1200000

[back to top]

Treatment Compliance: Percentage of Tablets Taken Relative to the Total Tablets

Treatment compliance was calculated as the percentage of tablets taken (presumed as [tablets dispensed-tablets returned]) relative to the total tablets, respectively, expected to be taken. Study drug interruptions due to an adverse event or other planned interruptions recorded on the electronic case report form (eCRF) were to be subtracted from the duration. For compliance to ribavirin (RBV), RBV dose modifications due to adverse events, toxicity management, or weight changes as recorded on the RBV Dose Modifications eCRF were to be used to modify the total number of tablets that should have been taken. A participant is considered to be compliant if the percentage is between 80% and 120%. (NCT02167945)
Timeframe: Up to Treatment Week 24

Interventionpercentage of tablets taken (Mean)
ABT-450/r/ABT-267 Plus ABT-333100.22
Weight-based Ribavirin (RBV)99.86

[back to top]

Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

SVR12 is defined as hepatitis C virus ribonucleic acid (HCV RNA) less than the lower limit of quantification (LLOQ) 12 weeks after the last actual dose of study drug. (NCT02167945)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
ABT-450/r/ABT-267 Plus ABT-333 With or Without Ribavirin (RBV)95.3

[back to top]

Mean Change From Baseline in Functional Assessment of Chronic Illness Therapy - Fatigue (FACIT-F) Total Score at Post-Treatment Week 12 and Post-Treatment Week 24

The FACIT-F is a symptom specific instrument with a focus on measuring fatigue in a variety of chronic diseases or health conditions. It was originally developed from interviews with oncology patients and clinical experts to assess anemia-associated fatigue. Its 13-item-version assesses peripheral, central, or mixed fatigue with a recall period of 7 days and yields a summed total score ranging between 0 and 52. Higher FACIT-F scores indicate a lesser degree of fatigue. Positive numbers indicate improvement from baseline. (NCT02167945)
Timeframe: From Baseline to Post-Treatment Week 12 and Post-Treatment Week 24

,,,
Interventionunits on a scale (Mean)
At Post-treatment Week 12At Post-treatment Week 24
Baseline Fibrosis Stage F0-F13.93.9
Baseline Fibrosis Stage F22.03.1
Baseline Fibrosis Stage F32.02.3
Baseline Fibrosis Stage F43.02.6

[back to top] [back to top]

Number of Participants With Selected HIV-1 Resistance Mutations Among Participants Who Experience HIV-1 Virologic Failure (VF)

Participants with one or more genotype mutations in protease conferring major resistance to any HIV-1 Protease Inhibitors (PI) antiretroviral drug, from a plasma sample drawn following confirmed HIV-1 VF outcome. (NCT02194998)
Timeframe: At confirmation of HIV-1 virologic failure.

InterventionParticipants (Count of Participants)
Cohort A [INI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]0
Cohort B [INI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]0
Cohort C [PI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]0
Cohort D [PI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]0

[back to top]

Percentage of Participants With Sustained Virologic Response at 12 Weeks After HCV Treatment Discontinuation (SVR12)

"SVR12 was defined as HCV RNA less than the assay LLOQ (<15 IU/mL) at 12 weeks post HCV treatment discontinuation.~A two-sided 90% confidence interval was calculated for the percentage of participants with SVR12 response using Clopper-Pearson method.~For those whose HCV early responses prior to SVR12 evaluation met the guidelines for HCV Virologic Failure (VF), their SVR12 outcome was defined as non-response. Those missing a HCV RNA result from the week 12 post HCV treatment discontinuation visit (and missing all subsequent evaluations) were considered non-responders. However, if HCV RNA evaluations subsequent to week 12 post treatment discontinuation were non-missing, then the first HCV RNA subsequent to week 12 post treatment discontinuation was instead used to define the primary outcome." (NCT02194998)
Timeframe: At 12 weeks after last dose of HCV study treatment. The duration for study treatment for Cohorts A and C and Cohorts B and D was 24 and 12 weeks, respectively.

Interventionpercentage of participants (Number)
Cohort A [INI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]95.2
Cohort B [INI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]60
Cohort C [PI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]100
Cohort D [PI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]100

[back to top]

Percentage of Participants With Sustained Virologic Response at 24 Weeks After HCV Treatment Discontinuation (SVR24)

"SVR24 was defined as HCV RNA less than the assay LLOQ (<15 IU/mL) at 24 weeks post treatment discontinuation.~A two-sided 90% confidence interval was calculated for the percentage of SVR24 response using method of Clopper-Pearson.~For those whose HCV early responses prior to SVR24 evaluation met the guidelines for HCV VF, their SVR24 outcome was defined as non-response. Those missing a HCV RNA result from the week 24 post HCV treatment discontinuation visit (and missing all subsequent evaluations) were considered non-responders. However, if HCV RNA evaluations subsequent to week 24 post treatment discontinuation were non-missing, then the first HCV RNA subsequent to week 24 post treatment discontinuation was instead used to define the primary outcome." (NCT02194998)
Timeframe: At 24 weeks after the date of last dose of HCV study treatment. The duration for study treatment for Cohorts A and C and Cohorts B and D was 24 and 12 weeks, respectively.

Interventionpercentage of participants (Number)
Cohort A [INI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]90.5
Cohort B [INI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]60
Cohort C [PI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]93.3
Cohort D [PI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]100

[back to top]

Change in IP-10 Concentration.

Absolute change from baseline in IP-10 (Interferon gamma-induced protein 10) concentration in plasma, calculated as value at the later time point minus baseline value. Baseline was defined as the date of first treatment dose. (NCT02194998)
Timeframe: At baseline, end of HCV treatment (EOT), and 12 weeks post EOT. The EOT for Cohorts A and C and Cohorts B and D was 24 and 12 weeks, respectively.

,,,
Interventionpg/mL (Median)
Change from baseline to EOTChange from baseline to 12 weeks post EOT
Cohort A [INI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]-244.4-127
Cohort B [INI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]-22.2-29.1
Cohort C [PI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]-116-83.1
Cohort D [PI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]-61.1-114.9

[back to top]

Change in Soluble CD14 (sCD14)

Absolute change from baseline in sCD14 levels in plasma calculated as value at the later time point minus baseline value. Baseline was defined as the date of first treatment dose. (NCT02194998)
Timeframe: At baseline, end of HCV treatment (EOT), and 12 weeks post EOT. The EOT for Cohorts A and C and Cohorts B and D was 24 and 12 weeks, respectively.

,,,
Interventionng/mL (Median)
Change from baseline to EOTChange from baseline to 12 weeks post EOT
Cohort A [INI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]307.6145.2
Cohort B [INI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]-1,063.2-894.2
Cohort C [PI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]-380.3-22.6
Cohort D [PI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]318.2-987.0

[back to top]

Levels of IP-10 Concentration.

Levels of IP-10 (Interferon gamma-induced protein 10) concentration in plasma. Baseline was defined as the date of first treatment dose. (NCT02194998)
Timeframe: At baseline, end of HCV treatment (EOT), and 12 weeks post EOT. The EOT for Cohorts A and C and Cohorts B and D was 24 and 12 weeks, respectively.

,,,
Interventionpg/mL (Median)
IP-10 at baselineIP-10 at EOTIP-10 at 12 weeks post EOT
Cohort A [INI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]379100.994.6
Cohort B [INI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]120.260.485.5
Cohort C [PI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]225.576.682.5
Cohort D [PI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]196.4182.6159.5

[back to top]

Levels of Soluble CD14 (sCD14)

Levels of sCD14. Baseline was defined as the date of first treatment dose. (NCT02194998)
Timeframe: At baseline, end of HCV treatment (EOT), and 12 weeks post EOT. The EOT for Cohorts A and C and Cohorts B and D was 24 and 12 weeks, respectively.

,,,
Interventionng/mL (Median)
sCD14 at BaselinesCD14 at EOTsCD14 at 12 weeks post EOT
Cohort A [INI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]1,832.02,126.51,977.3
Cohort B [INI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]2,226.81,132.81,367.4
Cohort C [PI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]3,157.02,421.13,424.5
Cohort D [PI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]3,092.02,801.32,608.3

[back to top]

Number of Participants Who Experienced HIV-1 Virologic Failure (VF)

HIV-1 VF was defined as two consecutive HIV-1 RNA results ≥ 200 copies/mL. (NCT02194998)
Timeframe: From treatment initiation to 4 weeks after last dose of HCV study treatment. HIV-1 RNA was measured at weeks 2, 4, 6, 8, 10, 12, 16, 20, 24, and 28. The durations for HCV study treatment for Cohorts A/C and Cohorts B/D were 24 and 12 weeks, respectively.

InterventionParticipants (Count of Participants)
Cohort A [INI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]0
Cohort B [INI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]0
Cohort C [PI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]0
Cohort D [PI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]1

[back to top]

Number of Participants Who Prematurely Discontinued HCV Study Treatment for Any Reason Other Than HCV Virologic Failure (VF)

HCV VF was defined as follows: confirmed increase from nadir in HCV RNA (defined as two consecutive HCV RNA measurements of >1 log10 IU/mL above nadir) at any time point; failure to achieve HCV RNA NCT02194998)
Timeframe: From treatment initiation to either 24 weeks (for Cohort A and C) or 12 weeks (for Cohort B and D). The duration for HCV study treatment for Cohorts A and C and Cohorts B and D was 24 and 12 weeks, respectively.

InterventionParticipants (Count of Participants)
Cohort A [INI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]1
Cohort B [INI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]1
Cohort C [PI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]0
Cohort D [PI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]0

[back to top]

Number of Participants With an Occurrence of Diagnoses Leading to Premature HCV Study Treatment or HIV-1 Antiretroviral (ARV) Discontinuation.

Participants who had diagnoses leading to premature HCV study treatment or HIV-1 ARV discontinuation post treatment initiation. (NCT02194998)
Timeframe: From treatment initiation to end of study follow-up at 48 weeks.The duration for HCV study treatment for Cohorts A and C and Cohorts B and D was 24 and 12 weeks, respectively. The duration for ARV treatment for all cohorts was 48 weeks.

InterventionParticipants (Count of Participants)
Cohort A [INI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]0
Cohort B [INI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]1
Cohort C [PI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]0
Cohort D [PI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]0

[back to top]

Number of Participants With an Occurrence of Laboratory Abnormality Grade 3 or Higher.

"Participants with an observed laboratory abnormalities grade 3 or higher post treatment initiation.~If entry (pre-treatment) lab result was grade 3, then a grade 4 result was required to meet this outcome.~Severity grading was based on DAIDS AE Grading Table, Version 1.0." (NCT02194998)
Timeframe: From treatment initiation to 30 days post date of last dose of HCV study treatment (whether planned or premature discontinuation). The duration for HCV study treatment for Cohorts A and C and Cohorts B and D was 24 and 12 weeks, respectively.

InterventionParticipants (Count of Participants)
Cohort A [INI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]5
Cohort B [INI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]3
Cohort C [PI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]3
Cohort D [PI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]0

[back to top]

Number of Participants With an Occurrence of Serious Adverse Events (SAEs) as Defined by International Conference on Harmonisation (ICH) Criteria

Participants who experienced at least one observed SAEs as defined by ICH after initiating HCV study treatment through 30 days post date of last dose of HCV study treatment. (NCT02194998)
Timeframe: From treatment initiation to 30 days post date of last dose of HCV study treatment (whether planned or premature discontinuation). The duration for HCV study treatment for Cohorts A and C and Cohorts B and D was 24 and 12 weeks, respectively.

InterventionParticipants (Count of Participants)
Cohort A [INI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]1
Cohort B [INI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]1
Cohort C [PI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]2
Cohort D [PI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]0

[back to top]

Number of Participants With an Occurrence of Signs/Symptoms Grade 3 or Higher

"Participants with signs/symptoms of grade 3 or higher post treatment initiation.~Participants with grade 3 sign/symptom prior to treatment initiation must have had one grade higher than pre-treatment to meet this outcome.~Severity grading was based on DAIDS AE Grading Table, Version 1.0." (NCT02194998)
Timeframe: From treatment initiation to 30 days post date of last dose of HCV study treatment (whether planned or premature discontinuation). The duration for HCV study treatment for Cohorts A and C and Cohorts B and D was 24 and 12 weeks, respectively.

InterventionParticipants (Count of Participants)
Cohort A [INI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]0
Cohort B [INI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]2
Cohort C [PI-based ART + PTV/r/OBT + DSV +/- RBV 24 Weeks]3
Cohort D [PI-based ART + PTV/r/OBT + DSV +/- RBV 12 Weeks]0

[back to top]

Percentage of Participants With Sustained Virologic Response 12 (SVR12) Weeks Post-treatment

SVR12 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification (NCT02207088)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
3-DAA ± RBV94.1

[back to top]

Percentage of Participants With On-treatment Virologic Failure

On-treatment virologic failure was defined as confirmed HCV RNA ≥ LLOQ after < LLOQ during treatment, confirmed increase of > 1 log (subscript)10(subscript) IU/mL above the lowest value post-baseline HCV RNA during treatment, or HCV RNA ≥ LLOQ persistently during treatment with at least 6 weeks of treatment. (NCT02207088)
Timeframe: Up to 24 weeks

Interventionpercentage of participants (Number)
3-DAA ± RBV0

[back to top]

Percentage of Participants With Post-Treatment Relapse

Post-treatment relapse was defined as confirmed HCV RNA ≥ LLOQ between end of treatment and 12 weeks after the last dose of study drug among participants completing treatment and with HCV RNA < LLOQ at the end of treatment. (NCT02207088)
Timeframe: Within 12 weeks after the last dose of study drug

Interventionpercentage of participants (Number)
3-DAA ± RBV1.5

[back to top]

Percentage of Participants With Sustained Virologic Response 12 Weeks (SVR12) Post-treatment

Sustained Virologic Response 12 (SVR12) is defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) less than the lower limit of quantification (< LLOQ; < 25 IU/mL) 12 weeks after the last dose of study drug. Participants with missing data were imputed as failures. (NCT02216422)
Timeframe: Post-treatment Day 1 to Post-treatment Week 12

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/Ritonavir Plus Dasabuvir With RBV100

[back to top]

Percentage of Participants With Post-Treatment Relapse

Post- Treatment Relapse is defined as confirmed HCV RNA >= LLOQ between end of treatment and 12 weeks after last actual dose of active study drug [up to and including the SVR12 assessment time point] for a participant with HCV RNA < LLOQ at Final Treatment Visit who completes treatment. (NCT02216422)
Timeframe: Post-treatment Day 1 to Post-treatment Week 12

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/Ritonavir Plus Dasabuvir With RBV0

[back to top]

Percentage of Participants With On-Treatment Virologic Failure

On-Treatment Virologic Failure is defined as confirmed HCV RNA >= LLOQ after HCV RNA < LLOQ during treatment, or confirmed increase from nadir (local minimum value) in HCV RNA [2 consecutive HCV RNA measurements > 1 log10 IU/mL above nadir] at any time point during treatment, or failure to suppress during treatment [all on-treatment values of HCV RNA >= LLOQ] with at least 6 weeks [defined as active study drug duration ≥ 36 days] of treatment. (NCT02216422)
Timeframe: Day 1 through Week 12

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/Ritonavir Plus Dasabuvir With RBV0

[back to top]

Percentage of Participants With Improvement From Baseline to Post-Treatment Week 12 in Hepatic Function Tests

"Improvement was defined as:~increase of more than 0.2 g/L from baseline to post-treatment Week 12 in albumin~decrease of more than 0.3 µmol/L from baseline to post-treatment Week 12 in bilirubin~decrease of more than 5 ng/mL from baseline to post-treatment Week 12 in alpha-fetoprotein~increase of more than 15*10^9/L from baseline to post-treatment Week 12 in platelet count~decrease of more than 0.2 from baseline to post-treatment Week 12 in international normalized ratio." (NCT02219477)
Timeframe: Up to post-treatment Week 12

Interventionpercentage of participants (Number)
AlbuminBilirubinAlpha-fetoproteinPlatelet countInternational normalized ratio
Group 1: GT1B77.866.733.322.20

[back to top]

Percentage of Participants With Improvement From Baseline to Post-Treatment Week 12 in Hepatic Function Tests

"Improvement was defined as:~increase of more than 0.2 g/L from baseline to post-treatment Week 12 in albumin~decrease of more than 0.3 µmol/L from baseline to post-treatment Week 12 in bilirubin~decrease of more than 5 ng/mL from baseline to post-treatment Week 12 in alpha-fetoprotein~increase of more than 15*10^9/L from baseline to post-treatment Week 12 in platelet count~decrease of more than 0.2 from baseline to post-treatment Week 12 in international normalized ratio." (NCT02219477)
Timeframe: Up to post-treatment Week 12

,
Interventionpercentage of participants (Number)
AlbuminBilirubinPlatelet countInternational normalized ratio
Group 2: GT1 Non-B77.372.714.30
Group 3: GT450.010000

[back to top]

Percentage of Participants With SVR12 in Group 3

SVR12, defined as HCV RNA < LLOQ in the SVR12 window (12 weeks after the last actual dose of study drug) without any confirmed quantifiable (≥ LLOQ) post-treatment value before or during that SVR window. Flanking imputation: for participants with missing HCV RNA at a visit, who have an undetectable HCV RNA or unquantifiable HCV RNA at the preceding visit and the succeeding visit, the missing value was imputed as undetectable or unquantifiable. For SVR analyses, if there was no value in the window after the flanking imputation but there was an HCV RNA value after the window, then it was imputed into the SVR window. After above imputations were applied, if there was still no value in the window but there was an HCV RNA value from a local laboratory present, then it was imputed into the SVR window. Otherwise, participants with missing data were counted as failures. (NCT02219477)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Group 3: GT466.7

[back to top]

Percentage of Participants With Improvement From Baseline to Post-Treatment Week 12 in FibroTest

The FibroTest score is used to assess liver fibrosis. Scores range from 0.00 to 1.00, with higher scores indicating a greater degree of fibrosis. Improvement was defined as a decrease of more than 0.2 from baseline to post-treatment Week 12. (NCT02219477)
Timeframe: Up to post-treatment Week 12

Interventionpercentage of participants (Number)
Group 1: GT1B0
Group 2: GT1 Non-B9.1
Group 3: GT450.0

[back to top]

Percentage of Participants With Improvement From Baseline to Post-Treatment Week 12 in Model for End-Stage Liver Disease (MELD) Score

MELD is a scoring system for assessing the severity of chronic liver disease. Scores range from 6 to 40, with higher scores indicating more severity. Improvement was defined as a decrease of 1 or more from baseline to post-treatment Week 12. (NCT02219477)
Timeframe: Up to post-treatment Week 12

Interventionpercentage of participants (Number)
Group 1: GT1B87.5
Group 2: GT1 Non-B61.9
Group 3: GT4100

[back to top]

Percentage of Participants With SVR12 Non-Response Due to Experiencing On-Treatment Virologic Failure

On-treatment virologic failure was defined as: confirmed HCV RNA ≥ LLOQ after HCV RNA < LLOQ during treatment; confirmed increase from nadir in HCV RNA (two consecutive HCV RNA measurements > 1 log˅10 IU/mL above nadir) at any time point during treatment; or HCV RNA ≥ LLOQ persistently during treatment with at least 6 weeks (≥ 36 days) of treatment. The 95% confidence interval was calculated using Wilson score method. SVR12 was defined as HCV RNA < LLOQ in the SVR12 window (12 weeks after the last actual dose of study drug) without any confirmed quantifiable (≥ LLOQ) post-treatment value before or during that SVR window. (NCT02219477)
Timeframe: Up to 24 weeks during treatment

Interventionpercentage of participants (Number)
Group 1: GT1B0
Group 2: GT1 Non-B0
Group 3: GT40

[back to top]

Percentage of Participants With SVR12 Non-Response Due to Experiencing Relapse˅12

Relapse˅12 was defined as confirmed HCV RNA ≥ LLOQ between end of treatment and 12 weeks after last actual dose of active study drug (up to and including the SVR12 window) for a participant with HCV RNA < LLOQ at final treatment visit who completes treatment and has post-treatment HCV RNA data. Completion of treatment was defined as a study drug duration ≥ 77 days for participants assigned to 12 weeks of treatment or ≥ 154 days for participants assigned to 24 weeks of treatment. SVR12 was defined as HCV RNA < LLOQ in the SVR12 window (12 weeks after the last actual dose of study drug) without any confirmed quantifiable (≥ LLOQ) post-treatment value before or during that SVR window. The 95% confidence interval was calculated using the Wilson score method. (NCT02219477)
Timeframe: Up to 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Group 1: GT1B0
Group 2: GT1 Non-B0
Group 3: GT40

[back to top]

Percentages of Participants With Sustained Virologic Response 12 Weeks Post-Treatment (SVR12) in Group 1 and in Group 2

SVR12, defined as HCV RNA < lower limit of quantification (LLOQ) in the SVR12 window (12 weeks after the last actual dose of study drug) without any confirmed quantifiable (≥ LLOQ) post-treatment value before or during that SVR window. Flanking imputation: for participants with missing HCV RNA at a visit who have an undetectable HCV RNA or unquantifiable HCV RNA at the preceding visit and the succeeding visit, the missing value was imputed as undetectable or unquantifiable. For SVR analyses, if there was no value in the window after the flanking imputation but there was an HCV RNA value after the window, then it was imputed into the SVR window. After above imputations were applied, if there was still no value in the window but there was an HCV RNA value from a local laboratory present, then it was imputed into the SVR window. Otherwise, participants with missing data were counted as failures. The 95% confidence interval was calculated using the Wilson score method. (NCT02219477)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Group 1: GT1B100
Group 2: GT1 Non-B95.8

[back to top]

Percentage of Participants With Improvement From Baseline to Post-Treatment Week 12 in Chld-Pugh Score

The The Child-Pugh score uses five clinical measures of liver disease (3 laboratory parameters and 2 clinical assessments) to measure severity of cirrhosis. Scores range from 5 to 15, with higher scores indicating more severity. Improvement was defined as a decrease of 1 or more from baseline to post-treatment Week 12. (NCT02219477)
Timeframe: Up to post-treatment Week 12

Interventionpercentage of participants (Number)
Group 1: GT1B66.7
Group 2: GT1 Non-B54.5
Group 3: GT450.0

[back to top]

Liver Decompensation: Time to Event

Time to liver decompensation was defined as number of days from the 1st day of study drug dosing for the participant to the date of liver decompensation. All liver decompensation was to be included, regardless of whether it occurred while the participant was still taking study drug or had previously discontinued study drug. If the participant didn't experience the event of interest nor had died (all-cause death), their data was to be censored at the date of their last available assessment of clinical outcomes. For participants with no post-baseline assessment, their data was to be censored on the 1st day of study drug dosing. All-cause death was a censoring event for liver decompensation. The event-free survival rates were estimated using Kaplan-Meier methodology and incidence estimates are presented with 95% confidence intervals. The pre-specified analysis of liver decompensation included pooled data from TOPAZ-I (this study) and the companion study TOPAZ-II (M14-222; NCT02167945). (NCT02219490)
Timeframe: At Post-Treatment Weeks 52, 104, 156, 208, and 260

,
Interventionpercentage of participants (Number)
Kaplan-Meier estimate at PT Week 52Kaplan-Meier estimate at PT Week 104Kaplan-Meier estimate at PT Week 156Kaplan-Meier estimate at PT Week 208Kaplan-Meier estimate at PT Week 260
Participants in Studies M14-222 and M14-423 Who Achieved SVR120.20.20.30.30.5
Participants in Studies M14-222 and M14-423 Who Did Not Achieve SVR124.54.54.54.54.5

[back to top] [back to top]

Liver Transplantation: Time to Event

Time to liver transplantation was defined as days from 1st day of study drug dosing for subject to date of liver transplantation. All liver transplantation was to be included, whether it occurred while the subject was still taking study drug or had previously discontinued study drug. If the subject didn't experience event of interest nor had died (all-cause death), their data was to be censored at the date of their last available assessment. For those with no post-baseline assessment, data was to be censored on 1st day of study drug dosing. All-cause death was a censoring event for liver transplantation. The event-free survival rates were estimated using Kaplan-Meier methodology and incidence estimates are presented with 95% confidence intervals. The pre-specified analysis of liver transplantation included pooled data from TOPAZ-I (this study) and the companion study TOPAZ-II (M14-222; NCT02167945). (NCT02219490)
Timeframe: At Post-Treatment Weeks 52, 104, 156, 208, and 260

,
Interventionpercentage of participants (Number)
Kaplan-Meier estimate at PT Week 52Kaplan-Meier estimate at PT Week 104Kaplan-Meier estimate at PT Week 156Kaplan-Meier estimate at PT Week 208Kaplan-Meier estimate at PT Week 260
Participants in Studies M14-222 and M14-423 Who Achieved SVR12000.10.10.2
Participants in Studies M14-222 and M14-423 Who Did Not Achieve SVR1200000

[back to top]

Hepatocellular Carcinoma: Time to Event

Time to hepatocellular carcinoma (HCC) was defined as number of days from 1st day of study drug dosing for subject to date of hepatocellular carcinoma. All HCC was to be included, whether it occurred while subject was still taking study drug or had previously discontinued study drug. If the subject didn't experience the event of interest nor had died (all-cause death), their data was to be censored at the date of their last available assessment. For those with no post-baseline assessment, their data was to be censored on the 1st day of study drug dosing. All-cause death was a censoring event for HCC. The event-free survival rates were estimated using Kaplan-Meier methodology and incidence estimates are presented with 95% confidence intervals. The pre-specified analysis of hepatocellular carcinoma included pooled data from TOPAZ-I (this study) and the companion study TOPAZ-II (M14-222; NCT02167945). (NCT02219490)
Timeframe: At Post-Treatment Weeks 52, 104, 156, 208, and 260

,
Interventionpercentage of participants (Number)
Kaplan-Meier estimate at PT Week 52Kaplan-Meier estimate at PT Week 104Kaplan-Meier estimate at PT Week 156Kaplan-Meier estimate at PT Week 208Kaplan-Meier estimate at PT Week 260
Participants in Studies M14-222 and M14-423 Who Achieved SVR120.20.40.50.60.9
Participants in Studies M14-222 and M14-423 Who Did Not Achieve SVR1200000

[back to top] [back to top]

Change From Baseline in FibroScan Score by SVR12 Status

The FibroScan test is a validated non-invasive test used to assess liver fibrosis in participants with chronic liver disease, and it was performed at study sites where it was available. For participants with Hepatitis C infection, a FibroScan score of 2-7 kPa indicates no liver scarring or mild scarring; a score of 8 or 9 is associated with moderate liver scarring; 9-14 indicates severe liver scarring; and 14 or higher is indicative of advanced liver scarring, cirrhosis. Negative changes from baseline indicate improvement in liver fibrosis. (NCT02219490)
Timeframe: At the final treatment visit and Post-Treatment Weeks 12, 24, 52, 104, 156, 208, and 260

,
InterventionkPa (Mean)
At the final treatment visitPost-Treatment Week 12Post-Treatment Week 24Post-Treatment Week 52Post-Treatment Week 104Post-Treatment Week 156Post-Treatment Week 208Post-Treatment Week 260
Participants in Study M14-423 Who Achieved SVR12-1.41-1.76-1.98-2.46-2.80-2.92-3.08-3.08
Participants in Study M14-423 Who Did Not Achieve SVR12-2.55-1.81-1.26-0.30-0.35-0.37-0.88-1.31

[back to top]

All-Cause Death: Time to Event

Time to all-cause death was defined as the number of days from the first day of study drug dosing for the participant to the date of death. All deaths were to be included, regardless of whether the death occurred while the participant was still taking study drug or had previously discontinued study drug. If the participant did not die, their data was to be censored at the date of their last available assessment of clinical outcomes. For participants with no post-baseline assessment, the participant's data was to be censored on the first day of study drug dosing. The event-free survival rates were estimated using Kaplan-Meier methodology and incidence estimates are presented with 95% confidence intervals. The pre-specified analysis of all-cause death included pooled data from TOPAZ-I (this study) and the companion study TOPAZ-II (M14-222; NCT02167945). (NCT02219490)
Timeframe: At Post-Treatment Weeks 52, 104, 156, 208, and 260

,
Interventionpercentage of participants (Number)
Kaplan-Meier estimate at PT Week 52Kaplan-Meier estimate at PT Week 104Kaplan-Meier estimate at PT Week 156Kaplan-Meier estimate at PT Week 208Kaplan-Meier estimate at PT Week 260
Participants in Studies M14-222 and M14-423 Who Achieved SVR120.10.71.21.52.0
Participants in Studies M14-222 and M14-423 Who Did Not Achieve SVR128.38.38.38.38.3

[back to top]

Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

SVR12 is defined as hepatitis C virus ribonucleic acid (HCV RNA) less than the lower limit of quantification (LLOQ) 12 weeks after the last actual dose of study drug. Flanking imputation, where applicable, was used to impute missing data. After applying flanking imputation, if there was no value in the window but there was an HCV RNA value from a local laboratory present, then it was to be imputed into the SVR window. Otherwise, participants with missing data were counted as failures. (NCT02219490)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
ABT-450/r/ABT-267 Plus ABT-333 With or Without Ribavirin (RBV)97.0

[back to top]

Percentage of Participants With Sustained Virologic Response 12 Weeks (SVR12) Post-treatment

"Sustained Virologic Response 12 (SVR12) is defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) less than the lower limit of quantification (< LLOQ; < 25 IU/mL) 12 weeks after the last dose of study drug.~The primary efficacy endpoints were non-inferiority and superiority of the percentage of participants who achieved sustained virologic response 12 weeks after treatment in each treatment arm compared with the historical threshold for sofosbuvir and peginterferon (pegIFN)/RBV for the treatment of subjects with HCV GT1b infection and cirrhosis." (NCT02219503)
Timeframe: Post-treatment Day 1 to Post-treatment Week 12

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/Ritonavir Plus Dasabuvir100

[back to top]

Percentage of Participants With On-Treatment Virologic Failure

On-Treatment Virologic Failure is defined as confirmed HCV RNA >= LLOQ after HCV RNA < LLOQ during treatment, or confirmed increase from nadir (local minimum value) in HCV RNA [2 consecutive HCV RNA measurements > 1 log10 IU/mL above nadir] at any time point during treatment, or failure to suppress during treatment [all on-treatment values of HCV RNA >= LLOQ] with at least 6 weeks [defined as active study drug duration ≥ 36 days] of treatment. (NCT02219503)
Timeframe: Day 1 through Week 12

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/Ritonavir Plus Dasabuvir0

[back to top]

Percentage of Participants With Post-Treatment Relapse

Post- Treatment Relapse is defined as confirmed HCV RNA >= LLOQ between end of treatment and 12 weeks after last actual dose of active study drug [up to and including the SVR12 assessment time point] for a participant with HCV RNA < LLOQ at Final Treatment Visit who completes treatment. (NCT02219503)
Timeframe: Post-treatment Day 1 to Post-treatment Week 12

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/Ritonavir Plus Dasabuvir0

[back to top]

Half-Life (t1/2) of ASP2151

(NCT02223351)
Timeframe: Blood samples were taken at pre-dose of Day 1 and 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36, 48 and 72h after post doses in first or second intervention

Interventionh (Geometric Mean)
400 mg ASP2151 Alone7.375
400mg ASP2151 With Ritonavir12.575
1200 mg ASP2151 Alone7.178
1200mg ASP2151 With Ritonavir12.498

[back to top]

Apparent Total Body Clearance (CL/F) From Plasma of ASP1955888-00

ASP1955888-00 is a metabolite of the study drug (ASP2151) (NCT02223351)
Timeframe: Blood samples were taken at pre-dose of Day 1 and 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36, 48 and 72h after post doses in first or second intervention

InterventionL/h (Mean)
400 mg ASP2151 Alone156.72
400mg ASP2151 With Ritonavir540.93
1200 mg ASP2151 Alone207.45
1200mg ASP2151 With Ritonavir775.35

[back to top]

Apparent Total Body Clearance (CL/F) of ASP2151 From Plasma

(NCT02223351)
Timeframe: Blood samples were taken at pre-dose of Day 1 and 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36, 48 and 72h after post doses in first or second intervention

InterventionL/h (Mean)
400 mg ASP2151 Alone18.314
400mg ASP2151 With Ritonavir6.815
1200 mg ASP2151 Alone25.735
1200mg ASP2151 With Ritonavir7.63

[back to top]

Apparent Volume of Distribution (Vd/F) of ASP1955888-00

ASP1955888-00 is a metabolite of the study drug (ASP2151) (NCT02223351)
Timeframe: Blood samples were taken at pre-dose of Day 1 and 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36, 48 and 72h after post doses in first or second intervention

InterventionL (Mean)
400 mg ASP2151 Alone1840.8
400mg ASP2151 With Ritonavir21474
1200 mg ASP2151 Alone2340.7
1200mg ASP2151 With Ritonavir27624.2

[back to top]

Area Under the Curve (AUC) of ASP1955888-00

ASP1955888-00 is a metabolite of the study drug (ASP2151) (NCT02223351)
Timeframe: Blood samples were taken at pre-dose of Day 1 and 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36, 48 and 72h after post doses in first or second intervention

Interventionh*ng/mL (Geometric Mean)
400 mg ASP2151 Alone2636.2
400mg ASP2151 With Ritonavir744.4
1200 mg ASP2151 Alone6023.1
1200mg ASP2151 With Ritonavir1646.1

[back to top]

Area Under the Curve (AUC) of ASP2151

(NCT02223351)
Timeframe: Blood samples were taken at pre-dose of Day 1 and 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36, 48 and 72h after post doses in first or second intervention

Interventionh*ng/mL (Geometric Mean)
400 mg ASP2151 Alone23162.4
400mg ASP2151 With Ritonavir60225.3
1200 mg ASP2151 Alone48532.3
1200mg ASP2151 With Ritonavir162131.6

[back to top]

Half-life (t1/2) of ASP1955888-00

ASP1955888-00 is a metabolite of the study drug (ASP2151) (NCT02223351)
Timeframe: Blood samples were taken at pre-dose of Day 1 and 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36, 48 and 72h after post doses in first or second intervention

Interventionh (Geometric Mean)
400 mg ASP2151 Alone8.063
400mg ASP2151 With Ritonavir40.213
1200 mg ASP2151 Alone7.617
1200mg ASP2151 With Ritonavir42.197

[back to top]

Peak Plasma Concentration (Cmax) of ASP1955888-00

ASP1955888-00 is a metabolite of the study drug (ASP2151) (NCT02223351)
Timeframe: Blood samples were taken at pre-dose of Day 1 and 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36, 48 and 72h after post doses in first or second intervention

Interventionng/mL (Geometric Mean)
400 mg ASP2151 Alone189.9
400mg ASP2151 With Ritonavir21.6
1200 mg ASP2151 Alone421.5
1200mg ASP2151 With Ritonavir56.2

[back to top]

Peak Plasma Concentration (Cmax) of ASP2151

(NCT02223351)
Timeframe: Blood samples were taken at pre-dose of Day 1 and 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36, 48 and 72h after post doses in first or second intervention

Interventionng/mL (Geometric Mean)
400 mg ASP2151 Alone1845.7
400mg ASP2151 With Ritonavir2518.7
1200 mg ASP2151 Alone3804
1200mg ASP2151 With Ritonavir6211.6

[back to top]

Time of Peak Concentration (Tmax) of ASP1955888-00

ASP1955888-00 is a metabolite of the study drug (ASP2151) (NCT02223351)
Timeframe: Blood samples were taken at pre-dose of Day 1 and 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36, 48 and 72h after post doses in first or second intervention

Interventionh (Median)
400 mg ASP2151 Alone3
400mg ASP2151 With Ritonavir1.5
1200 mg ASP2151 Alone3
1200mg ASP2151 With Ritonavir1.01

[back to top]

Time of Peak Concentration (Tmax) of ASP2151

(NCT02223351)
Timeframe: Blood samples were taken at pre-dose of Day 1 and 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36, 48 and 72h after post doses in first or second intervention

Interventionh (Median)
400 mg ASP2151 Alone3
400mg ASP2151 With Ritonavir4
1200 mg ASP2151 Alone3.025
1200mg ASP2151 With Ritonavir4

[back to top]

Number of Participants With Serious and Non-Serious Adverse Events

Refer to the result of adverse event. (NCT02223351)
Timeframe: Up to 31 days

,,,
Interventionparticipants (Number)
Non-serious adverse eventserious adverse event
1200 mg ASP2151 Alone150
1200 mg ASP2151 Withritonavir130
400 mg ASP2151 Alone80
400 mg ASP2151 With Ritonavir170

[back to top]

Apparent Volume of Distribution (Vd/F) of ASP2151

(NCT02223351)
Timeframe: Blood samples were taken at pre-dose of Day 1 and 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36, 48 and 72h after post doses in first or second intervention

InterventionL (Mean)
400 mg ASP2151 Alone190.1
400mg ASP2151 With Ritonavir123.17
1200 mg ASP2151 Alone266.33
1200mg ASP2151 With Ritonavir139.33

[back to top]

Percentage of Participants in Arms A, B and C With Post-treatment Relapse

Post-treatment relapse was defined as confirmed HCV RNA ≥ LLOQ between the end of treatment and 12 weeks after the last dose of study drug among participants with HCV RNA levels < LLOQ at the end of treatment. (NCT02265237)
Timeframe: From the end of treatment through 12 weeks after the last dose of study drug

Interventionpercentage of participants (Number)
Arm A0
Arm B0
Arm C0

[back to top]

Percentage of Participants in Arms A, B and C With On-treatment Virologic Failure

On-treatment virologic failure was defined as confirmed HCV RNA ≥ LLOQ after HCV RNA < LLOQ during treatment; confirmed increase of > 1 log(subscript)10(subscript) IU/mL above the lowest value post-baseline in HCV RNA during treatment; or all on-treatment values of HCV RNA >= LLOQ with at least 6 weeks of treatment. (NCT02265237)
Timeframe: Up to Treatment Week 24 (end of treatment) or premature discontinuation from treatment

Interventionpercentage of participants (Number)
Arm A1.7
Arm B0
Arm C0

[back to top]

Percentage of Participants With SVR12 in Participants Receiving 16 Weeks (Arm B) of Treatment Compared to Participants Receiving 24 Weeks of Treatment (Arm C)

SVR12 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification [NCT02265237)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Arm B100
Arm C93.4

[back to top]

Percentage of Participants With SVR12 in Participants Receiving 12 Weeks (Arm A) of Treatment Compared to Participants Receiving 16 Weeks of Treatment (Arm B)

SVR12 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification [NCT02265237)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Arm A96.6
Arm B100

[back to top]

Percentage of Participants in Arms A, B and C With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

SVR12 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification (NCT02265237)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Arm A96.6
Arm B100.0
Arm C93.4

[back to top]

Change From Baseline in Mean CD4+ T-cell Count at Week 48

CD4+ T-cell counts were quantified by a central laboratory using a commercially available assay. (NCT02275780)
Timeframe: Baseline and Week 48

InterventionCells/mm^3 (Mean)
Doravirine 100 mg192.7
Daurunavir 800 mg + Ritonavir 100 mg185.6

[back to top]

Mean Change From Baseline in Fasting Low Density Lipoprotein Cholesterol (LDL-C) at Week 48

Serum LDL-C was determined after an overnight fast. Change from Baseline was analyzed using ANCOVA models with terms for Baseline lipid level and treatment group. The Last Observation Carry Forward (LOCF) approach was applied for missing data or data collected after modifying lipid-lowering therapy. (NCT02275780)
Timeframe: Baseline and Week 48

,
Interventionmg/dL (Mean)
BaselineChange from Baseline
Daurunavir 800 mg + Ritonavir 100 mg91.769.92
Doravirine 100 mg91.10-4.51

[back to top]

Mean Change From Baseline in Fasting Triglyceride at Week 48

Serum triglyceride was determined after an overnight fast. Change from Baseline was analyzed using ANCOVA models with terms for Baseline lipid level and treatment group. The LOCF approach was applied for missing data or data collected after modifying lipid-lowering therapy. (NCT02275780)
Timeframe: Baseline and Week 48

,
Interventionmg/dL (Mean)
BaselineChange from Baseline
Daurunavir 800 mg + Ritonavir 100 mg117.0221.97
Doravirine 100 mg111.16-3.14

[back to top]

Mean Change From Baseline in Fasting Total Cholesterol at Week 48

Serum total cholesterol was determined after an overnight fast. Change from Baseline was analyzed using ANCOVA models with terms for Baseline lipid level and treatment group. The LOCF approach was applied for missing data or data collected after modifying lipid-lowering therapy. (NCT02275780)
Timeframe: Baseline and Week 48

,
Interventionmg/dL (Mean)
BaselineChange from Baseline
Daurunavir 800 mg + Ritonavir 100 mg157.7117.90
Doravirine 100 mg156.92-1.37

[back to top]

Percentage of Participants Achieving Plasma HIV-1 RNA <40 Copies/mL at Week 96

"The percentage of participants in each arm achieving HIV-1 RNA levels <40 copies/mL at Week 96 was determined. Plasma HIV-1 RNA levels were quantified with the Abbott RealTime HIV-1 Assay. Data were handled according to the US Food and Drug Administration (FDA) snapshot approach and all missing data were considered treatment failures, regardless of the reason." (NCT02275780)
Timeframe: Week 96

InterventionPercentage of participants (Number)
Doravirine 100 mg72.0
Daurunavir 800 mg + Ritonavir 100 mg64.4

[back to top]

Percentage of Participants Achieving Plasma HIV-1 RNA <40 Copies/mL at Week 48

"The percentage of participants in each arm achieving HIV-1 RNA levels <40 copies/mL at Week 48 was determined. Plasma HIV-1 RNA levels were quantified with the Abbott RealTime HIV-1 Assay. Data were handled according to the US Food and Drug Administration (FDA) snapshot approach and all missing data were considered treatment failures, regardless of the reason." (NCT02275780)
Timeframe: Week 48

InterventionPercentage of participants (Number)
Doravirine 100 mg83.3
Daurunavir 800 mg + Ritonavir 100 mg79.1

[back to top]

Mean Change From Baseline in Fasting Non-High Density Lipoprotein Cholesterol (Non-HDL-C) at Week 48

Serum non-HDL-C was determined after an overnight fast. Change from Baseline was analyzed using ANCOVA models with terms for Baseline lipid level and treatment group. The LOCF approach was applied for missing data or data collected after modifying lipid-lowering therapy. (NCT02275780)
Timeframe: Baseline and Week 48

,
Interventionmg/dL (Mean)
BaselineChange from Baseline
Daurunavir 800 mg + Ritonavir 100 mg114.4413.75
Doravirine 100 mg113.34-5.30

[back to top]

Percentage of Participants Achieving Plasma HIV-1 RNA <50 Copies/mL at Week 48

"The percentage of participants in each arm achieving HIV-1 RNA levels <50 copies/mL at Week 48 was determined. Plasma HIV-1 RNA levels were quantified with the Abbott RealTime HIV-1 Assay. Data were handled according to the US Food and Drug Administration (FDA) snapshot approach and all missing data were considered treatment failures, regardless of the reason." (NCT02275780)
Timeframe: Week 48

InterventionPercentage of participants (Number)
Doravirine 100 mg83.8
Daurunavir 800 mg + Ritonavir 100 mg79.9

[back to top]

Change From Baseline in Mean CD4+ T-cell Count at Week 96

CD4+ T-cell counts were quantified by a central laboratory using a commercially available assay. (NCT02275780)
Timeframe: Baseline and Week 96

InterventionCells/mm^3 (Mean)
Doravirine 100 mg224.1
Daurunavir 800 mg + Ritonavir 100 mg206.7

[back to top]

Mean Change From Baseline in Fasting High Density Lipoprotein Cholesterol (HDL-C) at Week 48

Serum HDL-C was determined after an overnight fast. Change from Baseline was analyzed using ANCOVA models with terms for Baseline lipid level and treatment group. The LOCF approach was applied for missing data or data collected after modifying lipid-lowering therapy. (NCT02275780)
Timeframe: Baseline and Week 48

,
Interventionmg/dL (Mean)
BaselineChange from Baseline
Daurunavir 800 mg + Ritonavir 100 mg43.274.15
Doravirine 100 mg43.583.94

[back to top]

Percentage of Participants With Any Serious Adverse Event

A serious adverse event is an AE that results in death, is life threatening, results in persistent or significant disability or incapacity, results in or prolongs a hospitalization, is a congenital anomaly or birth defect, is a cancer, is associated with an overdose, or is another important medical event. The percentage of participants with any SAE was assessed. (NCT02275780)
Timeframe: Up to 98 weeks

InterventionPercentage of Participants (Number)
Doravirine 100 mg7.0
Daurunavir 800 mg + Ritonavir 100 mg8.6

[back to top] [back to top] [back to top]

Percentage of Participants With Any Adverse Event

An adverse event (AE) is defined as any untoward medical occurrence in a study participant and which does not necessarily have to have a causal relationship to treatment. An adverse event can therefore be any unfavourable and unintended sign, symptom, or disease temporally associated with the study treatment or protocol-specified procedure, whether or not considered related to study treatment or protocol-specified procedure. Any worsening of a preexisting condition that is temporally associated with the study treatment is also an AE. The percentage of participants with any AE was assessed. (NCT02275780)
Timeframe: Up to 98 weeks

InterventionPercentage of Participants (Number)
Doravirine 100 mg84.6
Daurunavir 800 mg + Ritonavir 100 mg82.8

[back to top]

Percentage of Participants Who Discontinued Study Treatment Due to an Adverse Event

The percentage of participants who discontinued study treatment due to an AE was assessed. (NCT02275780)
Timeframe: Up to 96 weeks

InterventionPercentage of Participants (Number)
Doravirine 100 mg1.6
Daurunavir 800 mg + Ritonavir 100 mg3.4

[back to top]

Percentage of Participants Achieving Plasma HIV-1 RNA <50 Copies/mL at Week 96

"The percentage of participants in each arm achieving HIV-1 RNA levels <50 copies/mL at Week 96 was determined. Plasma HIV-1 RNA levels were quantified with the Abbott RealTime HIV-1 Assay. Data were handled according to the US Food and Drug Administration (FDA) snapshot approach and all missing data were considered treatment failures, regardless of the reason." (NCT02275780)
Timeframe: Week 96

InterventionPercentage of participants (Number)
Doravirine 100 mg73.1
Daurunavir 800 mg + Ritonavir 100 mg66.0

[back to top]

Percentage of Participants With Post-treatment Relapse

Post-treatment relapse was defined as confirmed HCV RNA ≥ LLOQ between the end of treatment and 12 weeks after the last dose of study drug among participants who completed treatment with HCV RNA levels < LLOQ at the end of treatment. (NCT02292719)
Timeframe: Up to 12 weeks after the last actual dose of active study drug

Interventionpercentage of participants (Number)
Arm A (Genotype [GT]3, Noncirrhotic)0
Arm B (GT3, Noncirrhotic)0
Arm C (GT2, Noncirrhotic)10.0
Arm D (GT2, Noncirrhotic)55.6
Arm E (GT3, Cirrhotic)0
Arm F (GT3, Noncirrhotic)0

[back to top]

Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

SVR12 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification [NCT02292719)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Arm A (Genotype [GT]3, Noncirrhotic)100
Arm B (GT3, Noncirrhotic)90.9
Arm C (GT2, Noncirrhotic)90.0
Arm D (GT2, Noncirrhotic)44.4
Arm E (GT3, Cirrhotic)100
Arm F (GT3, Noncirrhotic)100

[back to top]

Percentage of Participants With On-treatment Virologic Failure

On-treatment virologic failure was defined as confirmed increase of > 1 log(subscript)10(subscript) IU/mL above the lowest value post-baseline HCV RNA during treatment; confirmed HCV RNA ≥ LLOQ after HCV RNA < LLOQ during treatment, or HCV RNA ≥ LLOQ at end of treatment with at least 6 weeks of treatment for 12-week and 8-week treatment or at least 26 days of treatments for 6-week treatment. (NCT02292719)
Timeframe: Up to Week 12

Interventionpercentage of participants (Number)
Arm A (Genotype [GT]3, Noncirrhotic)0
Arm B (GT3, Noncirrhotic)0
Arm C (GT2, Noncirrhotic)0
Arm D (GT2, Noncirrhotic)0
Arm E (GT3, Cirrhotic)0
Arm F (GT3, Noncirrhotic)0

[back to top]

Percentage of Participants With Post-Treatment Relapse

Post-treatment relapse was defined as confirmed HCV RNA ≥ LLOQ between end of treatment and 12 weeks after the last dose of study drug among participants completing treatment and with HCV RNA < LLOQ at the end of treatment. (NCT02356562)
Timeframe: Within 12 weeks after the last actual dose of active study drug

Interventionpercentage of participants (Number)
Part 1, 3-DAA With SOF With or Without RBV4.8
Part 2, 3-DAA With RBV0.0

[back to top]

Percentage of Participants With On-treatment Virologic Failure

On-treatment virologic failure was defined as confirmed HCV RNA ≥ LLOQ after < LLOQ during treatment, confirmed increase of > 1 log (subscript)10(subscript) IU/mL above the lowest post-baseline HCV RNA during treatment, or HCV RNA ≥ LLOQ persistently during treatment with at least 6 weeks of treatment. (NCT02356562)
Timeframe: Up to week 24

Interventionpercentage of participants (Number)
Part 1, 3-DAA With SOF With or Without RBV0.0
Part 2, 3-DAA With RBV14.3

[back to top]

Percentage of Part 2 Participants With Sustained Virologic Response 12 (SVR12) Weeks Post-treatment

SVR12 was defined as plasma HCV RNA level NCT02356562)
Timeframe: 12 weeks after the last dose of active drug

Interventionpercentage of participants (Number)
Part 2, 3-DAA With RBV85.7

[back to top]

Percentage of Part 1 Participants With Sustained Virologic Response 12 (SVR12) Weeks Posttreatment

SVR12 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification [NCT02356562)
Timeframe: 12 weeks after the last dose of active drug

Interventionpercentage of participants (Number)
Part 1, 3-DAA With SOF With or Without RBV95.5

[back to top]

Percentage of Participants With Plasma HIV-1 RNA <40 c/mL at Weeks 48 and 96-Stage 1

Blood samples were collected for quantitative analysis of plasma HIV-1 RNA. Response was assessed using the last plasma HIV-1 RNA value in the predefined visit window to classify a participant's response status. The percentage of responders with HIV-1 RNA <40 c/mL at Weeks 48 and 96 using mITT Population (observed) which consisted of participants in the mITT Population excluding participants who had no HIV-1 RNA result data in the assessment visit windows due to discontinuation and who discontinued on or after the date of site notification of study termination by the sponsor (October 10, 2016) is presented. The study was terminated early during the primary end point analysis of Stage 1; hence, data was not collected for Week 96 analysis. (NCT02386098)
Timeframe: Weeks 48 and 96

,
InterventionPercentage of participants (Number)
Week 48; n=8, 9
Stage 1: BMS-955176 120 mg QD+ATV/r 300/100 mg QD+DTG 50 mg QD75.0
Stage 1: TDF 300 mg QD+ATV/r 300/100 mg QD+DTG 50 mg QD66.7

[back to top]

Percentage of Participants With HIV-1 RNA <200 c/mL at Weeks 24, 48 and 96-Stage 1

Blood samples were collected for quantitative analysis of plasma HIV-1 RNA. Response was assessed using the last plasma HIV-1 RNA value in the predefined visit window to classify a participant's response status. The percentage of responders with HIV-1 RNA <200 c/mL at Weeks 24, 48 and 96 using mITT Population (observed) which consisted of participants in the mITT Population excluding participants who had no HIV-1 RNA result data in the assessment visit windows due to discontinuation and who discontinued on or after the date of site notification of study termination by the sponsor (October 10, 2016) is presented. The study was terminated early during the primary end point analysis of Stage 1; hence, data was not collected for Week 96 analysis. (NCT02386098)
Timeframe: Weeks 24, 48 and 96

,
InterventionPercentage of participants (Number)
Week 24; n=32, 29Week 48; n=8, 9
Stage 1: BMS-955176 120 mg QD+ATV/r 300/100 mg QD+DTG 50 mg QD93.8100
Stage 1: TDF 300 mg QD+ATV/r 300/100 mg QD+DTG 50 mg QD89.7100

[back to top]

Change From Baseline in Percentage of CD4+ Cells Over Time-Stage 1

The percentage of CD4+ cells was assessed using flow cytometry. Baseline is the last value on or before the start of study treatment. Change from Baseline was calculated as the value at specified visit minus the Baseline value. NA indicates data was not available. The standard deviation could not be calculated as a single participant was analyzed at the specified time point. (NCT02386098)
Timeframe: Baseline and up to Week 72

,
InterventionPercentage of CD4+ cells (Mean)
Week 4; n=37, 33Week 8; n=36, 30Week 12; n=35, 32Week 16; n=34, 31Week 24; n=31, 28Week 32; n=23, 22Week 40; n=20, 15Week 48; n=7, 9Week 60; n=4, 2Week 72; n=1, 1
Stage 1: BMS-955176 120 mg QD+ATV/r 300/100 mg QD+DTG 50 mg QD1.941.803.413.144.714.135.387.097.9512.50
Stage 1: TDF 300 mg QD+ATV/r 300/100 mg QD+DTG 50 mg QD1.821.692.883.664.724.815.387.1610.0510.70

[back to top]

Change From Baseline in Logarithm to the Base 10 (log10) HIV-1 RNA Over Time-Stage 1

Blood samples were collected for analysis of HIV-1 RNA. Baseline is the last value on or before the start of study treatment. Change from Baseline was calculated as the value at specified visit minus the Baseline value. Change from Baseline in plasma HIV-1 RNA (log10) is summarized over time for the mITT Population using observed values, which excluded participants without HIV-1 RNA result data in the assessment visit windows due to discontinuation and who discontinued on or after the date of site notification of study termination by the sponsor (10 October 2016). NA indicates data was not available. The standard deviation could not be calculated as a single participant was analyzed at the specified time point. (NCT02386098)
Timeframe: Baseline and up to Week 72

,
Interventionlog10 c/mL (Mean)
Week 2; n=10, 5Week 4; n=37, 33Week 8; n=37, 30Week 12; n=36, 32Week 16; n=34, 32Week 24; n=32, 29Week 32; n=23, 23Week 40; n=21, 15Week 48; n=8, 9Week 60; n=4, 3Week 72; n=1, 1
Stage 1: BMS-955176 120 mg QD+ATV/r 300/100 mg QD+DTG 50 mg QD-4.232-4.400-4.103-4.394-4.402-4.220-4.381-4.366-4.508-5.037-3.326
Stage 1: TDF 300 mg QD+ATV/r 300/100 mg QD+DTG 50 mg QD-4.050-3.922-4.145-4.113-4.074-4.079-3.364-4.400-4.680-4.977-5.713

[back to top]

Change From Baseline in Cluster of Differentiation 4+ (CD4+) Cell Count Over Time-Stage 1

The CD4+ cell count was assessed using flow cytometry. Baseline is the last value on or before the start of study treatment. Change from Baseline was calculated as the value at specified visit minus the Baseline value. NA indicates data was not available. The standard deviation could not be calculated as a single participant was analyzed at the specified time point. (NCT02386098)
Timeframe: Baseline and up to Week 72

,
InterventionCells per microliter (Mean)
Week 4; n=37, 33Week 8; n=36, 30Week 12; n=35, 32Week 16; n=34, 31Week 24; n=31, 28Week 32; n=23, 22Week 40; n=20, 15Week 48; n=7, 9Week 60; n=4, 2Week 72; n=1, 1
Stage 1: BMS-955176 120 mg QD+ATV/r 300/100 mg QD+DTG 50 mg QD57.677.690.483.2127.290.0139.5125.0127.00.0
Stage 1: TDF 300 mg QD+ATV/r 300/100 mg QD+DTG 50 mg QD26.753.7115.193.8109.5122.1137.1175.1158.5171.0

[back to top]

Number of Participants With Serious Adverse Events (SAEs) and Adverse Events (AEs) Leading to Discontinuation (AELD)-Stage 1

An SAE is any untoward medical occurrence that at any dose: results in death; is life-threatening; requires inpatient hospitalization or causes prolongation of existing hospitalization; results in persistent or significant disability/incapacity; is a congenital anomaly/birth defect; or medical events that may jeopardize the participant or require intervention (medical or surgical) to prevent one of the outcomes mentioned before. The number of participants with SAEs and AELDs are presented. (NCT02386098)
Timeframe: Up to Week 96

,
InterventionParticipants (Count of Participants)
SAEsAELD
Stage 1: BMS-955176 120 mg QD+ATV/r 300/100 mg QD+DTG 50 mg QD42
Stage 1: TDF 300 mg QD+ATV/r 300/100 mg QD+DTG 50 mg QD31

[back to top]

Number of Participants With Occurrence of New Acquired Immunodeficiency Syndrome (AIDS) Defining Events-Stage 1

The occurrence of new AIDS defining events that is, Centers for Disease Control (CDC) Class C events in participants is presented. (NCT02386098)
Timeframe: Up to Week 96

InterventionParticipants (Count of Participants)
Stage 1: BMS-955176 120 mg QD+ATV/r 300/100 mg QD+DTG 50 mg QD1
Stage 1: TDF 300 mg QD+ATV/r 300/100 mg QD+DTG 50 mg QD0

[back to top]

Percentage of Participants With Plasma Human Immunodeficiency Virus-1 (HIV-1) Ribonucleic Acid (RNA) <40 Copies Per Milliliter (c/mL) at Week 24-Stage 1

Blood samples were collected for quantitative analysis of plasma HIV-1 RNA. Percentage of participants with plasma HIV-1 RNA <40 c/mL at Week 24 was assessed using the Food and Drug Administration (FDA) snapshot algorithm which used the last on-treatment plasma HIV-1 RNA measurement, within an FDA-specified visit window (18 to 30 weeks), to determine response. Analysis was performed on the modified intent to treat (mITT) Population which comprised of all randomized participants who received atleast one dose of BMS-955176 or TDF. (NCT02386098)
Timeframe: Week 24

InterventionPercentage of participants (Number)
Stage 1: BMS-955176 120 mg QD+ATV/r 300/100 mg QD+DTG 50 mg QD73.7
Stage 1: TDF 300 mg QD+ATV/r 300/100 mg QD+DTG 50 mg QD60.0

[back to top]

Mean Change From Baseline in Cluster of Differentiation (CD4) Cell Counts

The mean change from baseline in CD4 cell counts was assessed using the Observed Failure (OF) approach. With the OF approach, baseline values were carried forward for participants who discontinued due to lack of efficacy. Cell counts were measured and expressed as cells/mm^3, and percent change was then calculated. CD4 cell counts were quantified by a central laboratory using a commercially available assay. (NCT02397096)
Timeframe: Immediate Switch to MK-1439A arm: Baseline and Week 48; Delayed Switch to MK-1439A arm: Baseline and Week 24

,
Interventioncells/mm^3 (Mean)
BaselineChange from Baseline
Delayed Switch Group (DSG)655.618.0
Immediate Switch Group (ISG)660.513.9

[back to top]

Mean Change From Baseline in Fasting Low-density Lipoprotein Cholesterol (LDL-C)

To evaluate the effect on fasting LDL-C of an immediate switch to DOR/3TC/TDF on Study Day 1 compared with continuation of a ritonavir-boosted, PI-based regimen, as measured by mean change from baseline in each treatment group. The Last Observation Carry Forward (LOCF) approach was applied to missing data and data collected after a participant initiated lipid-modifying therapy. (NCT02397096)
Timeframe: Baseline and Week 24

,
Interventionmg/dL (Mean)
BaselineChange from Baseline
Delayed Switch (Ritonavir-boosted, PI-based) Group (DSG)109.00-1.94
Immediate Switch (Ritonavir--boosted, PI-based) Group (ISG)108.82-16.54

[back to top]

Mean Change From Baseline in Fasting Non-high-density Lipoprotein Cholesterol (Non-HDL-C)

Serum non-HDL-C was determined after an overnight fast. Change from Baseline was analyzed using ANCOVA models with terms for Baseline lipid level and treatment group. The Last Observation Carry Forward (LOCF) approach was applied for missing data or data collected after modifying lipid lowering therapy. (NCT02397096)
Timeframe: Baseline and Week 24

,
Interventionmg/dL (Mean)
BaselineChange from Baseline
Delayed Switch (Ritonavir-boosted, PI-based) Group (DSG)137.99-1.31
Immediate Switch (Ritonavir--boosted, PI-based) Group (ISG)139.14-24.74

[back to top]

Mean Change From Baseline in Cluster of Differentiation (CD4) Cell Counts

The mean change from baseline in CD4 cell counts at Week 48 was assessed using the Observed Failure (OF) approach. With the OF approach, baseline values were carried forward for participants who discontinued due to lack of efficacy. Cell counts were measured and expressed as cells/mm^3, and percent change was then calculated. CD4 cell counts were quantified by a central laboratory using a commercially available assay. (NCT02397096)
Timeframe: Baseline and Week 24

,
Interventioncells/mm^3 (Geometric Mean)
BaselineChange from Baseline
Delayed Switch Group (DSG)655.618.0
Immediate Switch Group (ISG)664.55.1

[back to top]

Percentage of Participants Discontinuing From Study Medication Due to an AE(s)

An AE is defined as any untoward medical occurrence in a participant administered a pharmaceutical product and which does not necessarily have to have a causal relationship with this treatment. (NCT02397096)
Timeframe: Up to Week 24

InterventionPercentage of Participants (Number)
Immediate Switch Group (ISG)2.5
Delayed Switch Group (DSG)0.4

[back to top]

Percentage of Participants Experiencing ≥1 Adverse Event (AE)

An AE is defined as any untoward medical occurrence in a participant administered a pharmaceutical product and which does not necessarily have to have a causal relationship with this treatment. (NCT02397096)
Timeframe: Up to week 24

InterventionPercentage of Participants (Number)
Immediate Switch Group (ISG)68.9
Delayed Switch Group (DSG)52.5

[back to top]

Percentage of Participants Experiencing ≥1 Serious Adverse Event (SAE)

A serious adverse event is an AE that results in death, is life threatening, results in persistent or significant disability or incapacity, results in or prolongs a hospitalization, is a congenital anomaly or birth defect, is a cancer, is associated with an overdose, or is another important medical event. The percentage of participants with any SAE was assessed. (NCT02397096)
Timeframe: Up to 24 weeks

InterventionPercentage of Participants (Number)
Immediate Switch Group (ISG)2.9
Delayed Switch Group (DSG)3.6

[back to top]

Percentage of Participants Maintaining HIV-1 RNA <40 Copies/mL

"The percentage of participants in each arm achieving HIV-1 RNA levels <40 copies/mL was determined. Plasma HIV-1 RNA levels were quantified with the Abbott RealTime HIV-1 Assay. Data were handled according to the US Food and Drug Administration (FDA) snapshot approach and all missing data were considered treatment failures, regardless of the reason." (NCT02397096)
Timeframe: Immediate Switch to MK-1439A arm: Week 48; Delayed Switch to MK-1439A arm: Week 24

InterventionPercentage of Participants (Number)
Immediate Switch Group (ISG)89.7
Delayed Switch Group (DSG)93.3

[back to top]

Percentage of Participants Maintaining HIV-1 RNA <40 Copies/mL

To evaluate the immunological effect of an immediate switch to MK -1439A on Study Day 1 compared with continuation of a ritonavir boosted, PI-based regimen, as measured by the proportion of subjects maintaining HIV-1 RNA below the limit of quantification (BLoQ) by the Abbott RealTime HIV-1 Assay (<40 copies/mL) in both treatment groups. (NCT02397096)
Timeframe: Immediate Switch to MK-1439A arm: Week 24; Delayed Switch to MK-1439A arm: Week 24

InterventionPercentage of Participants (Number)
Immediate Switch Group (ISG)92.8
Delayed Switch Group (DSG)93.3

[back to top]

Percentage of Participants Maintaining HIV-1 RNA <50 Copies/mL

"The percentage of participants in each arm achieving HIV-1 RNA levels <50 copies/mL was determined. Plasma HIV-1 RNA levels were quantified with the Abbott RealTime HIV-1 Assay. Data were handled according to the US Food and Drug Administration (FDA) snapshot approach and all missing data were considered treatment failures, regardless of the reason." (NCT02397096)
Timeframe: Week 24

InterventionPercentage of Participants (Number)
Immediate Switch Group (ISG)93.7
Delayed Switch Group (DSG)94.6

[back to top]

Percentage of Participants Maintaining Human Immunodeficiency Virus-1 Ribonucleic Acid (HIV-1 RNA) <50 Copies/mL

"The percentage of participants in each arm achieving HIV-1 RNA levels <50 copies/mL was determined. Plasma HIV-1 RNA levels were quantified with the Abbott RealTime HIV-1 Assay. Data were handled according to the US Food and Drug Administration (FDA) snapshot approach and all missing data were considered treatment failures, regardless of the reason." (NCT02397096)
Timeframe: Immediate Switch to MK-1439A arm: Week 48; Delayed Switch to MK-1439A arm: Week 24

InterventionPercentage of Participants (Number)
Immediate Switch Group (ISG)90.8
Delayed Switch Group (DSG)94.6

[back to top]

Percentage of Participants With HIV-1 RNA >=50 Copies/mL

"The percentage of participants in each arm achieving HIV-1 RNA levels >=50 copies/mL was determined. Plasma HIV-1 RNA levels were quantified with the Abbott RealTime HIV-1 Assay. Data were handled according to the US Food and Drug Administration (FDA) snapshot approach." (NCT02397096)
Timeframe: Immediate Switch to MK-1439A arm: Week 48; Delayed Switch to MK-1439A arm: Week 24

InterventionPercentage of Participants (Number)
Immediate Switch Group (ISG)1.6
Delayed Switch Group (DSG)1.8

[back to top]

Percentage of Participants With Sustained Virologic Response 12 Weeks (SVR12) Post-treatment

The percentage of participants with sustained virologic response (plasma Hepatitis C virus ribonucleic acid [HCV RNA] level less than the lower limit of quantitation [< LLOQ]) 12 weeks after the last dose of study drug. The LLOQ for the assay was 25 IU/mL. (NCT02399345)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/r, Dasabuvir, and SOF Plus RBV80

[back to top]

Percentage of Subjects With On-treatment Virologic Failure

Virologic failure during treatment was defined as confirmed HCV RNA ≥ LLOQ after HCV RNA < LLOQ during treatment; confirmed increase from nadir in HCV RNA (defined as 2 consecutive HCV RNA measurements > 1 log10 IU/mL above nadir) during treatment; or failure to suppress during treatment (defined as all values of HCV RNA ≥ LLOQ during treatment). (NCT02399345)
Timeframe: 6 weeks

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/r, Dasabuvir, and SOF Plus RBV0

[back to top]

Percentage of Subjects With Post-treatment Relapse

Percentage of subjects with HCV RNA less than the lower limit of quantification at the end of treatment with confirmed HCV RNA greater than or equal to the lower limit of quantification through 12 weeks post treatment (NCT02399345)
Timeframe: Up to 12 weeks after last actual dose of active study drug

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/r, Dasabuvir, and SOF Plus RBV20

[back to top]

Improvement in Quantitative Serum HDV RNA Levels After 4-12 Weeks of Lonafarnib-based Therapy

log HDV RNA decline from baseline to end of treatment (4-12 weeks of lonafarnib-based therapy) (NCT02430181)
Timeframe: 4-12 weeks

Interventionlog IU/mL (Mean)
Lonafarnib 200 mg BID0.03
Lonafarnib 300 mg BID-1.78
Lonafarnib 100 mg TID-1.3
100 mg BID Lonafarnib/PEG IFN-a-1.85
200 mg BID Lonafarnib/PEG IFN-a0
300 mg BID Lonafarnib/PEG IFN-a0
Lonafarnib/Ritonavir-1.2

[back to top]

ALT Normalization at End of Treatment

Proportion of intent to treat population who normalize ALT at end of treatment (NCT02430194)
Timeframe: 12-48 weeks

InterventionParticipants (Count of Participants)
Lonafarnib/Ritonavir - I3
Lonafarnib/Ritonavir - II1
Lonafarnib/Ritonavir - III1
Lonafarnib/Ritonavir - IV2
Lonafarnib/Ritonavir - V0
Lonafarnib/Ritonavir - VI2
Lonafarnib/Ritonavir - VII6
Lonafarnib/Ritonavir/PEG IFN-a - VIII0
Lonafarnib/Ritonavir/PEG IFN-a - IX5
Lonafarnib/Ritonavir/PEG IFN-a - X2

[back to top]

Mean HDV RNA Decline

mean HDV RNA decline of intent to treat population from baseline to end of treatment (NCT02430194)
Timeframe: 12-48 weeks

Interventionlog HDV RNA IU/mL (Mean)
Lonafarnib/Ritonavir - I-1.39
Lonafarnib/Ritonavir - II0.33
Lonafarnib/Ritonavir - III-1.11
Lonafarnib/Ritonavir - IV-0.67
Lonafarnib/Ritonavir/PEG IFN-a - V-1.97
Lonafarnib/Ritonavir - VI-0.31
Lonafarnib/Ritonavir - VII-1.94
Lonafarnib/Ritonavir/PEG IFN-a - VIII-2.85
Lonafarnib/Ritonavir/PEG IFN-a - IX-2.69
Lonafarnib/Ritonavir/PEG IFN-a - X-3.81

[back to top]

≥2 log10 Decline of HDV RNA From Baseline at End of Treatment (EOT)

Proportion of intent to treat patients with ≥2 log10 decline of HDV RNA from baseline at end of treatment (EOT) (NCT02430194)
Timeframe: 12-48 weeks

InterventionParticipants (Count of Participants)
Lonafarnib/Ritonavir - I1
Lonafarnib/Ritonavir - II0
Lonafarnib/Ritonavir - III1
Lonafarnib/Ritonavir - IV0
Lonafarnib/Ritonavir/PEG IFN-a - V1
Lonafarnib/Ritonavir - VI1
Lonafarnib/Ritonavir - VII5
Lonafarnib/Ritonavir/PEG IFN-a - VIII4
Lonafarnib/Ritonavir/PEG IFN-a - IX3
Lonafarnib/Ritonavir/PEG IFN-a - X4

[back to top]

< LLOQ in HDV RNA at End of Treatment (EOT)

Proportion of intent to treat patients with HDV RNA below the limit of quantitation at end of treatment (NCT02430194)
Timeframe: 12-48 weeks

InterventionParticipants (Count of Participants)
Lonafarnib/Ritonavir - I0
Lonafarnib/Ritonavir - II1
Lonafarnib/Ritonavir - III1
Lonafarnib/Ritonavir - IV0
Lonafarnib/Ritonavir/PEG IFN-a - V1
Lonafarnib/Ritonavir - VI0
Lonafarnib/Ritonavir - VII6
Lonafarnib/Ritonavir/PEG IFN-a - VIII1
Lonafarnib/Ritonavir/PEG IFN-a - IX3
Lonafarnib/Ritonavir/PEG IFN-a - X2

[back to top]

Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

SVR12 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification [NCT02442271)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
3-DAA ± RBV96.4

[back to top]

Hepatitis C Virus Patient-Reported Outcomes Instrument (HCV-PRO) Total Score: Change From Baseline to 12 Weeks After the Last Dose of Study Drug

The HCV-PRO has been developed to capture the function and well-being impact of HCV conditions and treatment and contains 16 items important to HCV-infected patients; items were totaled to a summary score. Scores range from 0 to 100. A higher HCV-PRO score indicates a better state of health and a decrease from baseline represents worsening. If a participant answered at least 12 of the 16 items, the missing items were imputed with the mean score of the answered items; if a participant did not answer at least 12 of the items, the total score was considered missing. (NCT02442271)
Timeframe: Day 1 (Baseline), 12 weeks after the last actual dose of the study drug

,
Interventionunits on a scale (Mean)
SVR12 Not AchievedSVR12 Achieved
Fibrosis Stage F30.53.8
Fibrosis Stage F40.84.2

[back to top]

Percentage of Participants With SVR12 by Fibrosis Stage

SVR12 was defined as plasma HCV RNA level NCT02442271)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Fibrosis Stage F396.6
Fibrosis Stage F496.2

[back to top]

Percentage of Participants With SVR12 by Participant Eligibility for Treatment With Interferon (IFN) at Screening

SVR12 was defined as HCV RNA level NCT02442271)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Interferon (IFN)-Ineligible, Treatment-Naive90.0
Interferon (IFN)-Eligible, Treatment-Naive96.7
Interferon (IFN)-Ineligible, Treatment-Experienced85.7
Interferon (IFN)-Eligible, Treatment-Experienced97.3

[back to top]

Percentage of Participants With SVR12 by Participant Prior HCV Treatment Experience

SVR12 was defined as HCV RNA level NCT02442271)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Treatment-Naive96.1
Pegylated Interferon (PegIFN)/RBV Null Responders95.5
Pegylated Interferon (PegIFN)//RBV Partial Responders100
Pegylated Interferon (PegIFN)/RBV Non-Responders100
Pegylated Interferon (PegIFN)/RBV Relapser97.1
Pegylated Interferon (PegIFN)/RBV Breakthrough100
IFN Interolerant85.7
Other91.7

[back to top]

(SF-36v2) Mental Component Summary (MCS) Scores: Change From Baseline to 12 Weeks After the Last Dose of Study Drug

The SF-36v2 is a non-disease specific Health Related Quality of Life (HRQoL) instrument. The SF-36v2 comprises 36 total items (questions) targeting a subject's functional health and well-being in 8 domains (physical functioning, role physical, bodily pain, general health, vitality, social functioning, role emotional and mental health) with a recall period of four weeks. Domain scores are aggregated into a PCS score and a MCS score. Scores SF-36v2 scores range from 1-100: higher scores indicate a better state of health and a decrease from baseline represents worsening. If a participant answered at least 50% of the items in a multi-item scale of the SF-36v2, the missing items were imputed with the average score of the answered items in the same domain. In cases where the participant did not answer at least 50% of the items, the score for that domain was considered missing. The SF-36v2 MCS and PCS scores were not computed if any domain was missing. (NCT02442271)
Timeframe: Day 1 (Baseline), 12 weeks after the last actual dose of the study drug

,
Interventionunits on a scale (Mean)
SVR12 Not AchievedSVR12 Achieved
Fibrosis Stage F32.42.4
Fibrosis Stage F4-0.62.5

[back to top]

Short-Form 36 Version 2 Health Survey (SF-36v2) Physical Component Summary (PCS) Scores: Change From Baseline to 12 Weeks After the Last Dose of Study Drug

The SF-36v2 is a non-disease specific Health Related Quality of Life (HRQoL) instrument. The SF-36v2 comprises 36 total items (questions) targeting a subject's functional health and well-being in 8 domains (physical functioning, role physical, bodily pain, general health, vitality, social functioning, role emotional and mental health) with a recall period of four weeks. Domain scores are aggregated into a Physical Component Summary (PCS) score and a Mental Component Summary (MCS) score. SF-36v2 scores range from 1-100: higher scores indicate a better state of health and a decrease from baseline represents worsening. If a participant answered at least 50% of the items in a multi-item scale of the SF-36v2, the missing items were imputed with the average score of the answered items in the same domain. In cases where the participant did not answer at least 50% of the items, the score for that domain was considered missing. The SF-36v2 MCS and PCS scores were not computed if any domain (NCT02442271)
Timeframe: Day 1 (Baseline), 12 weeks after the last actual dose of the study drug

,
Interventionunits on a scale (Mean)
SVR12 Not AchievedSVR12 Achieved
Fibrosis Stage F3-0.50.1
Fibrosis Stage F41.32.1

[back to top]

Percentage of Participants With Post-treatment Relapse

Post-treatment relapse was defined as confirmed HCV RNA ≥ LLOQ between the end of treatment and 12 weeks after the last dose of study drug among participants who completed treatment with HCV RNA levels < LLOQ at the end of treatment. (NCT02442284)
Timeframe: From the end of treatment through 12 weeks after the last dose of study drug

Interventionpercentage of participants (Number)
3-DAA ± RBV for 12 or 24 Weeks2.2

[back to top]

Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

SVR12 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification [NCT02442284)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
3-DAA ± RBV for 12 or 24 Weeks93.9

[back to top]

Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12) Among Participants With Ongoing Psychiatric Disorders

SVR12 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification [NCT02442284)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
3-DAA ± RBV for 12 or 24 Weeks95.8

[back to top]

Percentage of Participants With Virologic Failure During Treatment

On-treatment virologic failure was defined as confirmed HCV RNA ≥ LLOQ after HCV RNA < LLOQ during treatment, or HCV RNA ≥ LLOQ at end of treatment. (NCT02442284)
Timeframe: up to 12 weeks (for 12-week treatment group) or up to 24 weeks (for 24-week treatment group

Interventionpercentage of participants (Number)
3-DAA ± RBV for 12 or 24 Weeks1.0

[back to top]

Sustained Virological Response (SVR) at Week 12

Percentage of study participants achieving sustained virological response (SVR) at Week 12 per protocol among study participants who completed 12-week course of treatment. (NCT02461745)
Timeframe: 12 weeks

InterventionParticipants (Count of Participants)
Genotype 1a116
Genotype 1b66

[back to top]

Sustained Virological Response (SVR) at Week 4

Percentage of study participants achieving sustained virological response (SVR) at Week 4 per protocol among subjects who completed 12-week course of treatment in the study. (NCT02461745)
Timeframe: 4 weeks

InterventionParticipants (Count of Participants)
Genotype 1a116
Genotype 1b65

[back to top]

Change in Interferon (IFN)-Stimulated Genes (ISG) Expression in Peripheral Blood Mononucleated Cells (PBMCs) for Participants Achieving SVR12

The changes from week 0 to post-treatment (PT) week 12 in key ISG expression in PBMCs for participants achieving sustained virologic response 12 weeks PT (SVR12) where SVR12 was defined as hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification (NCT02476617)
Timeframe: Week 0 to Post-Treatment Week 12

InterventionFold change (Mean)
CXCL10ISG15CXCL11MX1IFI27OAS2DDX60IFIT3IFI44STAT1CXCL9IFIT1HERC5IRF7OAS1PLSCR1IRF9IFIH1TRIM14IRF1IRF3STAT2IFNGIFNL1IFNL2IFNL3IFNL4IFNGR1ADARALG10BCHECCL2CCL4CD163CXCL5CXCR3DDIT4DDX58DPP4EIF2AK2GBP1GCKRGUCY1B3HELZ2HERC6IFI35IFI44LIFI6IFIT2IFIT5IFITM1IFITM2IFITM3IFNAIFNBIL10IL18IRF2ISG20MAP3K14MOV10MS4A4AMX2MYST1NOS2ANT5C3OASLPPP3CBRNASELRSAD2SLC27A2SOCS1SOCS3USP18
Ombitasvir/Paritaprevir/Ritonavir + Dasabuvir + RBV-0.464-0.148-0.611-0.399-0.462-0.168-0.016-0.543-0.467-0.301-0.552-0.440-0.358-0.1710.284-0.255-0.205-0.3210.022-0.1300.044-0.0510.516-0.361-0.199-0.205-0.228-0.046-0.212-0.083-0.1010.2000.6380.5561.0850.3080.0290.3880.869-0.317-0.2880.0090.350-0.232-0.074-0.023-0.554-0.464-0.365-0.291-0.0660.158-0.423-0.202-0.1670.202-0.0780.3110.156-0.072-0.185-0.005-0.101-0.0570.2490.017-0.2330.0110.395-0.4060.0221.3130.025-0.288

[back to top]

Part 1: Maximum Plasma Concentration (Cmax) of Ombitasvir (OBV)

Cmax is the peak concentration that a drug or drug metabolite achieves in a specified compartment after the drug has been administrated and before administration of a second dose. (NCT02486406)
Timeframe: At Week 2

Interventionng/mL (Geometric Mean)
15 - 29 kg Body Weight99.6
30 - 44 kg Body Weight116
≥ 45 kg Body Weight83.7

[back to top]

Parts 1 and 2: Percentage of Participants With Alanine Aminotransferase (ALT) Normalization During Treatment by Formulation, Age and Weight Group, Across All Subjects, and Across All Subjects on the Adult Formulations

Alanine aminotransferase (ALT) normalization during treatment is defined as ALT ≤ the upper limit of normal (ULN) at the final treatment visit for participants with ALT > ULN at baseline. (NCT02486406)
Timeframe: 12 or 24 weeks after starting study drug, depending on treatment duration

Interventionpercentage of participants (Number)
Adult Tablet,12-17 YR, ≥ 45 kg, ALT Normalization87.5
Mini-tablet, 9-11 YR, 15 to 29 kg, ALT Normalization100
Mini-tablet, 9-11 YR, 30 to 44 kg, ALT Normalization100
Mini-tablet, 3-8 YR, 15 to 29 kg, ALT Normalization80.0
Mini-tablet Total, ALT Normalization87.5
Participants in Parts 1 and 2 of the Study, ALT Normalization87.5

[back to top]

Parts 1 & 2: Percentage of Participants With Sustained Virologic Response 24 Weeks After the Last Actual Dose of Study Drug (SVR24), Summarized by Formulation, Age and Weight Group, Across All Subjects, and Across All Subjects on the Adult Formulations

SVR24 is defined as hepatitis C virus ribonucleic acid (HCV RNA) < lower limit of quantification (LLOQ) 24 weeks after the last actual dose of study drug. (NCT02486406)
Timeframe: 24 weeks after last dose of study drug (Week 36 or 48 depending on treatment duration)

Interventionpercentage of participants (Number)
Participants in Parts 1 and 2 of the Study96.9
Adult Tablet, 12-17 YR, ≥ 45 kg100
Mini-tablet, 9-11 YR, 15 to 29 kg100.0
Mini-tablet, 9-11 YR, 30 to 44 kg88.9
Mini-tablet, 9-11 YR, ≥ 45 kg100.0
Mini-tablet, 3-8 YR, 15 to 29 kg92.9
Mini-tablet Total92.3

[back to top]

Part 1: Concentration of Drug in Blood Plasma Against Time [Area Under the Curve (AUC)] of Dasabuvir (DSV)

AUC is a measure of how long and how much drug is present in the body after dosing. The amount of dasabuvir present was measured up to 12 hours after dosing. For two subjects in the 15-29 kg group, the 24 h concentration was used as the 12 h concentration due to the significant sampling time deviation. For one subject in the 30-44 kg group, the 24 h concentration was used as the 12 h concentration due to the significant sampling time deviation. (NCT02486406)
Timeframe: At Week 2

Interventionng•h/mL (Geometric Mean)
15 - 29 kg Body Weight3960
30 - 44 kg Body Weight5960
≥ 45 kg Body Weight4630

[back to top]

Part 1: Maximum Plasma Concentration (Cmax) of Ritonavir (RTV)

Cmax is the peak concentration that a drug or drug metabolite achieves in a specified compartment after the drug has been administrated and before administration of a second dose. (NCT02486406)
Timeframe: At Week 2

Interventionng/mL (Geometric Mean)
15 - 29 kg Body Weight1090
30 - 44 kg Body Weight1830
≥ 45 kg Body Weight1180

[back to top]

Part 1: Concentration of Drug in Blood Plasma Over Time [Area Under the Curve (AUC)] of Ritonavir (RTV)

AUC is a measure of how long and how much drug is present in the body after dosing. The amount of ritonavir present was measured up to 24 hours after dosing. (NCT02486406)
Timeframe: At Week 2

Interventionng•h/mL (Geometric Mean)
15 - 29 kg Body Weight6570
30 - 44 kg Body Weight14100
≥ 45 kg Body Weight8900

[back to top]

Parts 1 and 2: Percentage of Participants With Sustained Virologic Response 12 Weeks After the Last Actual Dose of Study Drug (SVR12)

SVR12 is defined as hepatitis C virus ribonucleic acid (HCV RNA) < lower limit of quantification (LLOQ) 12 weeks after the last actual dose of study drug. (NCT02486406)
Timeframe: 12 weeks after last dose of study drug (Week 24 or 36 depending on treatment duration)

Interventionpercentage of participants (Number)
All Participants, Total98.4

[back to top]

Part 1: Lowest Plasma Concentration (Ctrough) of Dasabuvir (DSV)

Minimum plasma concentration (Ctrough; measured in ng/mL) was directly determined from the concentration-time data. (NCT02486406)
Timeframe: At Weeks 2 and 8

,,
Interventionng/mL (Geometric Mean)
Week 2Week 8
≥ 45 kg Body Weight165191
15 - 29 kg Body Weight110168
30 - 44 kg Body Weight215264

[back to top]

Part 1: Lowest Plasma Concentration (Ctrough) of Ombitasvir (OBV)

Minimum plasma concentration (Ctrough; measured in ng/mL) was directly determined from the concentration-time data. (NCT02486406)
Timeframe: At Weeks 2 and 8

,,
Interventionng/mL (Geometric Mean)
Week 2Week 8
≥ 45 kg Body Weight21.820.9
15 - 29 kg Body Weight24.729.6
30 - 44 kg Body Weight28.230.4

[back to top]

Part 1: Lowest Plasma Concentration (Ctrough) of Paritaprevir (PTV)

Minimum plasma concentration (Ctrough; measured in ng/mL) was directly determined from the concentration-time data. (NCT02486406)
Timeframe: At Weeks 2 and 8

,,
Interventionng/mL (Geometric Mean)
Week 2Week 8
≥ 45 kg Body Weight18.023.5
15 - 29 kg Body Weight9.8617.3
30 - 44 kg Body Weight16.118.4

[back to top]

Part 1: Lowest Plasma Concentration (Ctrough) of Ritonavir (RTV)

Minimum plasma concentration (Ctrough; measured in ng/mL) was directly determined from the concentration-time data. (NCT02486406)
Timeframe: At Weeks 2 and 8

,,
Interventionng/mL (Geometric Mean)
Week 2Week 8
≥ 45 kg Body Weight29.858.2
15 - 29 kg Body Weight16.191.8
30 - 44 kg Body Weight32.138.1

[back to top]

Part 1: Maximum Plasma Concentration (Cmax) of Paritaprevir (PTV)

Cmax is the peak concentration that a drug or drug metabolite achieves in a specified compartment after the drug has been administrated and before administration of a second dose. (NCT02486406)
Timeframe: At Week 2

Interventionng/mL (Geometric Mean)
15 - 29 kg Body Weight294
30 - 44 kg Body Weight1540
≥ 45 kg Body Weight870

[back to top]

Parts 1 and 2: Percentage of Participants With Sustained Virologic Response 12 Weeks After the Last Actual Dose of Study Drug (SVR12) Summarized by Formulation, Age and Weight Group, and Across All Subjects on the Adult Formulations

SVR12 is defined as hepatitis C virus ribonucleic acid (HCV RNA) < lower limit of quantification (LLOQ) 12 weeks after the last actual dose of study drug. (NCT02486406)
Timeframe: 12 weeks after last dose of study drug (Week 24 or 36 depending on treatment duration)

Interventionpercentage of participants (Number)
Adult Tablet, 12-17 YR, ≥ 45 kg100
Mini-tablet, 9-11 YR, 15 to 29 kg100
Mini-tablet, 9-11 YR, 30 to 44 kg100
Mini-tablet, 9-11 YR, ≥ 45 kg100
Mini-tablet, 3-8 YR, 15 to 29 kg92.9
Mini-tablet Total96.2

[back to top]

Part 1: Maximum Plasma Concentration (Cmax) of Dasabuvir (DSV)

Cmax is the peak concentration that a drug or drug metabolite achieves in a specified compartment after the drug has been administrated and before administration of a second dose. (NCT02486406)
Timeframe: At Week 2

Interventionng/mL (Geometric Mean)
15 - 29 kg Body Weight579
30 - 44 kg Body Weight830
≥ 45 kg Body Weight671

[back to top]

Part 1: Concentration of Drug in Blood Plasma Over Time [Area Under the Curve (AUC)] of Paritaprevir (PTV)

AUC is a measure of how long and how much drug is present in the body after dosing. The amount of paritaprevir present was measured up to 24 hours after dosing. (NCT02486406)
Timeframe: At Week 2

Interventionng•h/mL (Geometric Mean)
15 - 29 kg Body Weight2180
30 - 44 kg Body Weight8640
≥ 45 kg Body Weight5770

[back to top]

Part 1: Concentration of Drug in Blood Plasma Over Time [Area Under the Curve (AUC)] of Ombitasvir (OBV)

AUC is a measure of how long and how much drug is present in the body after dosing. The amount of ombitasvir present was measured up to 24 hours after dosing. (NCT02486406)
Timeframe: At Week 2

Interventionng•h/mL (Geometric Mean)
15 - 29 kg Body Weight1270
30 - 44 kg Body Weight1490
≥ 45 kg Body Weight1060

[back to top]

Percentage of Participants With On-treatment Virologic Failure

On-treatment virologic failure was defined as confirmed HCV RNA ≥ LLOQ after HCV RNA < LLOQ during treatment or confirmed increase of > 1 log(subscript)10(subscript) IU/mL above the lowest value post-baseline in HCV RNA during treatment with at least 6 weeks of treatment. (NCT02487199)
Timeframe: 12 weeks

Interventionpercentage of participants (Number)
HCV GT1a (3-DAA)0.0
HCV GT4 (2-DAA)0.0

[back to top]

Percentage of Participants With Post-treatment Relapse

Post-treatment relapse was defined as confirmed HCV RNA ≥ LLOQ between the end of treatment and 12 weeks after the last dose of study drug among participants who completed treatment with HCV RNA levels < LLOQ at the end of treatment. (NCT02487199)
Timeframe: From the end of treatment through 12 weeks after the last dose of study drug

Interventionpercentage of participants (Number)
HCV GT1a (3-DAA)0.0
HCV GT4 (2-DAA)0.0

[back to top]

Number of Participants With Adverse Events

An adverse event (AE) is defined as any untoward medical occurrence in a patient or clinical investigation subject administered a pharmaceutical product and which does not necessarily have a causal relationship with this treatment. The investigator assessed the relationship of each event to the use of study drug as either reasonable possibility or no reasonable possibility. A serious adverse event (SAE) is an event that results in death, is life-threatening, requires or prolongs hospitalization, results in a congenital anomaly, persistent or significant disability/incapacity or is an important medical event that, based on medical judgment, may jeopardize the subject and may require medical or surgical intervention to prevent any of the outcomes listed above. Treatment-emergent events (TEAEs/TESAEs) are defined as any event that began or worsened in severity from first dose of study drug until 30 days after the last dose. For more details on AEs please see the Adverse Event section. (NCT02487199)
Timeframe: Treatment-emergent adverse events (TEAEs) and serious adverse events (TESAEs) were collected from first dose of study drug until 30 days after the last dose of study drug (up to 16 weeks)

,
InterventionParticipants (Count of Participants)
Any TEAEAny TESAE
HCV GT1a (3-DAA)133
HCV GT4 (2-DAA)51

[back to top]

Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

SVR12 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification (NCT02487199)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
HCV GT1a (3-DAA)100
HCV GT4 (2-DAA)80.0

[back to top]

Slope of the Second Phase Decline in Plasma HCV Ribonucleic Acid (RNA) Levels During Treatment

HCV viral kinetics in plasma during therapy were modeled through non-linear mixed effect models, including a rapid first phase of initial decline and a slower second phase decline. The slope of the second phase decline was estimated for each treatment arm. (NCT02493855)
Timeframe: From Week 0 to Week 2

Intervention1/day (Median)
Arm A: Ribavirin Full Dose for Last 10 Weeks0.0036
Arm B: Ribavirin Full Dose for 12 Weeks0.0046
Arm C: Ribavirin Low-dose for 12 Weeks0.0051

[back to top]

Number of Participants With Decline of HDV RNA Quantitative Measurement of > 2 Logs From Baseline at 24 Weeks of Treatment

The number of participants who experienced a > 2 log IU/mL decline in serum HDV RNA levels at 24 weeks of treatment (NCT02511431)
Timeframe: 24 weeks

InterventionParticipants (Count of Participants)
50mg/100mg Lonafarnib/Ritonavir1
75mg/100mg Lonafarnib/Ritonavir1
100mg/100mg Lonafarnib/Ritonavir0
Placebo Then 50 mg/100 mg Lonafarnib/Ritonavir0
Placebo Then 75mg/100mg Lonafarnib/Ritonavir0
Placebo Then 100mg/100mg Lonafarnib/Ritonavir0

[back to top]

Number of Participants With Decline of Hepatitis Delta Virus (HDV) RNA Quantitative Measurements of >2 Logs From Baseline at 12 Weeks of Treatment

The number of participants who experienced a > 2 log IU/mL decline in serum HDV RNA at 12 weeks of treatment (NCT02511431)
Timeframe: 12 weeks

InterventionParticipants (Count of Participants)
50mg/100mg Lonafarnib/Ritonavir1
75mg/100mg Lonafarnib/Ritonavir1
100mg/100mg Lonafarnib/Ritonavir1
Placebo Then 50 mg/100 mg Lonafarnib/Ritonavir0
Placebo Then 75mg/100mg Lonafarnib/Ritonavir1
Placebo Then 100mg/100mg Lonafarnib/Ritonavir0

[back to top]

Percentage of Participants With Post-treatment Relapse by Post-treatment Week 12

Post-treatment relapse was defined as confirmed HCV RNA ≥ LLOQ between the end of treatment and 12 weeks after the last dose of active study drug among participants who completed treatment with HCV RNA levels < LLOQ at the end of treatment. (NCT02517515)
Timeframe: From the end of treatment through 12 weeks after the last dose of active study drug

Interventionpercentage of participants (Number)
Double-blind 3-DAA (Treatment-Naïve)0
Double-blind 3-DAA (Treatment-Experienced)0

[back to top]

Percentage of Participants With Post-treatment Relapse by Post-treatment Week 24

Post-treatment relapse was defined as confirmed HCV RNA ≥ LLOQ between the end of treatment and 24 weeks after the last dose of active study drug among participants who completed treatment with HCV RNA levels < LLOQ at the end of treatment. (NCT02517515)
Timeframe: From the end of treatment through 24 weeks after the last dose of active study drug

Interventionpercentage of participants (Number)
Double-blind 3-DAA (Treatment-Naïve)0
Double-blind 3-DAA (Treatment-Experienced)0

[back to top]

Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

SVR12 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification [NCT02517515)
Timeframe: 12 weeks after the last actual dose of active study drug

Interventionpercentage of participants (Number)
Double-blind 3-DAA (Treatment-Naïve)99.5
Double-blind 3-DAA (Treatment-Experienced)100

[back to top]

Percentage of Participants With Sustained Virologic Response 24 Weeks Post-treatment (SVR24)

SVR24 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification [NCT02517515)
Timeframe: 24 weeks after the last actual dose of active study drug

Interventionpercentage of participants (Number)
Double-blind 3-DAA (Treatment-Naïve)99.5
Double-blind 3-DAA (Treatment-Experienced)100

[back to top]

Percentage of Participants With On-treatment Virologic Failure

On-treatment virologic failure was defined as confirmed HCV RNA ≥ LLOQ after HCV RNA < LLOQ during active treatment; confirmed increase of > 1 log(subscript)10(subscript) IU/mL above the lowest value post-baseline in HCV RNA during active treatment; or all on-treatment values of HCV RNA ≥ LLOQ with at least 6 weeks of active treatment. (NCT02517515)
Timeframe: up to 12 weeks

Interventionpercentage of participants (Number)
Double-blind 3-DAA (Treatment-Naïve)1.1
Double-blind 3-DAA (Treatment-Experienced)0

[back to top]

Percentage of Participants Achieving Sustained Virologic Response 24 Weeks Post-Treatment (SVR24)

SVR24 is defined as HCV RNA less than the LLOQ at 24 weeks following therapy. 95% CI is calculated using Wilson's score method. The lower bound of the 95% CI for the percentage of participants with SVR12 must exceed 67% to achieve superiority. SVR24 is primary outcome measure only for China. (NCT02517528)
Timeframe: 24 weeks after last dose of study drug

Interventionpercentage of participants (Number)
ABT-450/r/ABT-267 + ABT-333 + Ribavirin100

[back to top]

Percentage of Participants With On Treatment Virologic Failure

On treatment virologic failure is defined as confirmed HCV RNA greater than or equal to the LLOQ at any point during treatment after HCV RNA less than LLOQ, confirmed increase from the lowest value post-baseline in HCV RNA (two consecutive HCV RNA measurements greater than 1 log10 IU/mL above the lowest value post-baseline) at any time point during treatment, or HCV RNA greater than or equal to LLOQ persistently during treatment with at least 6 weeks (greater than or equal to 36 days) of treatment. 95% CI is calculated using Wilson's score method. (NCT02517528)
Timeframe: Within 12 weeks after first dose of study drug

Interventionpercentage of participants (Number)
ABT-450/r/ABT-267 + ABT-333 + Ribavirin0

[back to top]

Percentage of Participants With Virologic Relapse

Virologic relapse is defined as confirmed HCV RNA greater than or equal to LLOQ between end of treatment and 12 weeks after the last dose of study drugs among participants completing treatment and with HCV RNA less than LLOQ at the end of treatment. Completion of treatment is defined as a study drug duration greater than or equal to 77 days. 95% CI is calculated using Wilson's score method. (NCT02517528)
Timeframe: Within 12 weeks after the last dose of study drug

Interventionpercentage of participants (Number)
ABT-450/r/ABT-267 + ABT-333 + Ribavirin0

[back to top]

Percentage of Participants Achieving Sustained Virologic Response 12 Weeks Post-Treatment (SVR12)

SVR12 is defined as hepatitis C virus ribonucleic acid (HCV RNA) less than the lower limit of quantification (LLOQ) at 12 weeks following therapy. 95% confidence interval (CI) is calculated using Wilson's score method. The lower bound of the 95% CI for the percentage of participants with SVR12 must exceed 67% to achieve superiority. (NCT02517528)
Timeframe: 12 weeks after last dose of study drug

Interventionpercentage of participants (Number)
ABT-450/r/ABT-267 + ABT-333 + Ribavirin100

[back to top]

Percentage of Participants With Virologic Relapse by Post-Treatment Week 24

Virologic relapse is defined as confirmed HCV RNA greater than or equal to LLOQ between end of treatment and 24 weeks after the last dose of study drugs among participants completing treatment and with HCV RNA less than LLOQ at the end of treatment. Completion of treatment is defined as a study drug duration greater than or equal to 77 days. 95% CI is calculated using Wilson's score method. (NCT02517528)
Timeframe: Within 24 weeks after the last dose of study drug

Interventionpercentage of participants (Number)
ABT-450/r/ABT-267 + ABT-333 + Ribavirin0

[back to top]

Number of Patients With 1 Log Reduction From Baseline by Timepoint

Number of patients with at least 1 log reduction in HDV RNA from baseline by dose level and timepoint (NCT02527707)
Timeframe: Baseline and Week 1, Week 2, Week 4, Week 6, Week 8, Week 12, Week 16, Week 20, or Week 24

InterventionParticipants (Count of Participants)
Week 1Week 2Week 4Week 6Week 8Week 12Week 16Week 20Week 24
Lonafarnib/Ritonavir761011101212109

[back to top]

Change From Baseline to Week 24 in Mean Hepatitis D Virus (HDV) Ribonucleic Acid (RNA) Titer

Change from baseline to Week 24 in mean HDV RNA titer following dose escalating from lonafarnib 50 mg BID to 75 mg BID and to 100 mg BID, all boosted with ritonavir 100 mg BID. (NCT02527707)
Timeframe: Baseline and Week 24 (6 months)

Interventionlog IU/mL (Mean)
Lonafarnib/Ritonavir-1.62

[back to top]

Percentage of Participants With Relapse

Relapse was defined as hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL at the end of treatment followed by HCV RNA level greater than or equal to 50 IU/mL. (NCT02581163)
Timeframe: Up to 48 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 42.3

[back to top]

Percentage of Participants Achieving Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

"SVR12 was defined as hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL 12 weeks after the last actual dose of study drug. The core population (CP) consisted of participants who met all inclusion criteria and were adequately treated according to the standard of care and within local label recommendations for their specific disease characteristics (cirrhotic status, genotype). The core population with sufficient follow-up data 12 weeks after the last actual dose of study drug (CPSFU12) was defined as all CP participants who fulfilled one of the following criteria:~evaluable HCV RNA data ≥70 days after the last actual dose of the ABBVIE REGIMEN~an HCV RNA value ≥50 IU/mL at the last measurement post-baseline~HCV RNA <50 IU/mL at the last measurement post-baseline, but no HCV RNA measurement ≥70 days after the last actual dose of the ABBVIE REGIMEN due to reasons related to safety (e.g. dropped out due to AE) or virologic failure" (NCT02581163)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Core population (CP)CPSFU12
Participants With HCV Genotype 1 or 487.793.8

[back to top]

Percentage of Participants With Virologic Response at End of Treatment (EoT)

Virologic response was defined as hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL at the end of treatment. (NCT02581163)
Timeframe: Up to 24 weeks

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 492.9

[back to top]

Percentage of Participants With On-treatment Virologic Failure

On-treatment virologic failure was defined as breakthrough (at least 1 documented hepatitis C virus ribonucleic acid (HCV RNA) less than 50 IU/mL followed by HCV RNA greater than or equal to 50 IU/mL during treatment) or failure to suppress (each measured on-treatment HCV RNA value greater than or equal to 50 IU/mL). (NCT02581163)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 41.6

[back to top]

Percentage of Participants With Viral Breakthrough

Viral breakthrough was defined as at least 1 documented hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL followed by HCV RNA level greater than or equal to 50 IU/mL during treatment. (NCT02581163)
Timeframe: Up to 24 weeks

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 40.3

[back to top]

Percentage of Participants With Missing Sustained Virologic Response 12 Weeks Post-Treatment (SVR12) Data and/or Nonresponders Who Did Not Meet Specific SVR12 Nonresponder Criteria

The number of participants with missing SVR12 data or SVR12 nonresponder participants who did not meet criteria for on-treatment virologic failure, relapse, premature treatment discontinuation, and who did not have an Insufficient virological response reported was documented. (NCT02581163)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 45.8

[back to top]

Percentage of Participants Meeting Relapse Criteria

Relapse was defined as hepatitis C virus ribonucleic acid (HCV RNA) less than 50 IU/mL at end of treatment or at the last on-treatment HCV RNA measurement followed by HCV RNA greater than or equal to 50 IU/mL post-treatment. (NCT02581163)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 41.9

[back to top]

Percentage of Participants Meeting Premature Study Drug Discontinuation Criteria

Premature study drug discontinuation was defined as participants who prematurely discontinued study drug with no on-treatment virologic failure. (NCT02581163)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 41.6

[back to top]

Percentage of Participants Meeting Premature Study Drug Discontinuation Criteria

Premature study drug discontinuation was defined as participants who prematurely discontinued study drug with no on-treatment virologic failure. (NCT02581189)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 42.1

[back to top]

Percentage of Participants With On-treatment Virologic Failure

On-treatment virologic failure was defined as breakthrough (at least 1 documented hepatitis C virus ribonucleic acid (HCV RNA) less than 50 IU/mL followed by HCV RNA greater than or equal to 50 IU/mL during treatment) or failure to suppress (each measured on-treatment HCV RNA value greater than or equal to 50 IU/mL). (NCT02581189)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 41.3

[back to top]

Percentage of Participants With Relapse

Relapse was defined as hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL at the end of treatment followed by HCV RNA level greater than or equal to 50 IU/mL. (NCT02581189)
Timeframe: Up to 48 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 42.6

[back to top]

Percentage of Participants With Viral Breakthrough

Viral breakthrough was defined as at least 1 documented hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL followed by HCV RNA level greater than or equal to 50 IU/mL during treatment. (NCT02581189)
Timeframe: Up to 24 weeks

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 40.9

[back to top]

Percentage of Participants With Virologic Response at End of Treatment (EoT)

Virologic response was defined as hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL at the end of treatment. (NCT02581189)
Timeframe: Up to 24 weeks

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 493.8

[back to top]

Percentage of Participants Achieving Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

"SVR12 was defined as hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL 12 weeks after the last actual dose of study drug. The core population (CP) consisted of participants who met all inclusion criteria and were adequately treated according to the standard of care and within local label recommendations for their specific disease characteristics (cirrhotic status, genotype). The core population with sufficient follow-up data 12 weeks after the last actual dose of study drug (CPSFU12) was defined as all CP participants who fulfilled one of the following criteria:~evaluable HCV RNA data ≥70 days after the last actual dose of the ABBVIE REGIMEN~an HCV RNA value ≥50 IU/mL at the last measurement post-baseline~HCV RNA <50 IU/mL at the last measurement post-baseline, but no HCV RNA measurement ≥70 days after the last actual dose of the ABBVIE REGIMEN due to reasons related to safety (e.g. dropped out due to AE) or virologic failure" (NCT02581189)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Core population (CP)CPSFU12
Participants With HCV Genotype 1 or 486.694.1

[back to top]

Percentage of Participants Meeting Relapse Criteria

Relapse was defined as hepatitis C virus ribonucleic acid (HCV RNA) less than 50 IU/mL at end of treatment or at the last on-treatment HCV RNA measurement followed by HCV RNA greater than or equal to 50 IU/mL post-treatment. (NCT02581189)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 42.1

[back to top]

Percentage of Participants With Missing Sustained Virologic Response 12 Weeks Post-Treatment (SVR12) Data and/or Nonresponders Who Did Not Meet Specific SVR12 Nonresponder Criteria

The number of participants with missing SVR12 data or SVR12 nonresponder participants who did not meet criteria for on-treatment virologic failure, relapse, premature treatment discontinuation, and who did not have an Insufficient virological response reported was documented. (NCT02581189)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 46.6

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: Glucose

(NCT02581202)
Timeframe: Baseline, Week 24

Interventionmmol/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants4.88-0.10

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: HDL

(NCT02581202)
Timeframe: Baseline, Week 24

Interventionmmol/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants1.530.07

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: Insulin

(NCT02581202)
Timeframe: Baseline, Week 24

InterventionµEq/mL (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants10.000.68

[back to top]

Number of Participants With Adverse Events

Adverse events were not recorded in a solicited manner as in an interventional trial, but were recorded by spontaneous reporting in line with the requirements described in European Medicines Agency (EMA) Guideline on Good Pharmacovigilance Practices (GVP) module VI and local pharmacovigilance practice of the Russian Federation. (NCT02581202)
Timeframe: up to Week 48

InterventionParticipants (Count of Participants)
HIV-1 Infected Participants3

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: Serum Creatinine

(NCT02581202)
Timeframe: Baseline, Week 24

Interventionµmol/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants81.502.36

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: Total Cholesterol

(NCT02581202)
Timeframe: Baseline, Week 24

Interventionmmol/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants5.110.07

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: Triglycerides

(NCT02581202)
Timeframe: Baseline, Week 24

Interventionmmol/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants1.72-0.70

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: LDL

(NCT02581202)
Timeframe: Baseline, Week 24

Interventionmmol/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants2.66-0.07

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: Glucose

(NCT02581202)
Timeframe: Baseline, Week 48

Interventionmmol/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants4.83-0.16

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: HDL

(NCT02581202)
Timeframe: Baseline, Week 48

Interventionmmol/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants1.43-0.00

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: Insulin

(NCT02581202)
Timeframe: Baseline, Week 48

InterventionµEq/mL (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants10.000.69

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: LDL

(NCT02581202)
Timeframe: Baseline, Week 48

Interventionmmol/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants2.920.14

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: Serum Creatinine

(NCT02581202)
Timeframe: Baseline, Week 48

Interventionµmol/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants82.132.75

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: Total Cholesterol

(NCT02581202)
Timeframe: Baseline, Week 48

Interventionmmol/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants5.120.11

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: Triglycerides

(NCT02581202)
Timeframe: Baseline, Week 48

Interventionmmol/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants1.61-0.06

[back to top]

Number of Participants Who Developed Resistance to Nucleoside Reverse Transcriptase Inhibitors (NRTIs), Non-nucleoside Reverse Transcriptase Inhibitors (NNRTIs), and Protease Inhibitors (PIs)

(NCT02581202)
Timeframe: Week 48

InterventionParticipants (Count of Participants)
NRTINNRTIPI
HIV-1 Infected Participants110

[back to top]

Absolute Values and Change From Baseline in CD4+ T-cell Counts (Cells/mm^3) at Week 24

(NCT02581202)
Timeframe: Baseline, Week 24

Interventioncells/mm^3 (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants703.4364.70

[back to top]

Percentage of Participants on Dual Therapy (LPV/r + 3TC) With Undetectable HIV-1 RNA Level at Week 24

(NCT02581202)
Timeframe: Week 24

Interventionpercentage of participants (Number)
HIV-1 Infected Participants99.5

[back to top]

Percentage of Participants on Dual Therapy (LPV/r + 3TC) With Undetectable HIV-1 RNA Level at Week 48

(NCT02581202)
Timeframe: Week 48

Interventionpercentage of participants (Number)
HIV-1 Infected Participants100

[back to top]

Absolute Values and Change From Baseline (BL) in HIV-1- RNA Viral Load at Week 24 (Base-10 Logarithm Transformed Data)

(NCT02581202)
Timeframe: Baseline, Week 24

Interventionlog10 copies/mL (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants3.220.01

[back to top]

Absolute Values and Change From Baseline (BL) in HIV-1- RNA Viral Load at Week 24 (Untransformed Data)

(NCT02581202)
Timeframe: Baseline, Week 24

Interventioncopies/mL (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants25.220.22

[back to top]

Absolute Values and Change From Baseline in Anthropometric Measurements At Week 24

(NCT02581202)
Timeframe: Baseline, Week 24

Interventioncm (Mean)
Absolute values: arm circumferenceChange from BL: arm circumferenceAbsolute values: hip circumferenceChange from BL: hip circumferenceAbsolute values: waist circumferenceChange from BL: waist circumference
HIV-1 Infected Participants29.82-0.3854.620.5783.290.31

[back to top]

Absolute Values and Change From Baseline in Anthropometric Measurements At Week 48

(NCT02581202)
Timeframe: Baseline, Week 48

Interventioncm (Mean)
Absolute values: arm circumferenceChange from BL: arm circumferenceAbsolute values: hip circumferenceChange from BL: hip circumferenceAbsolute values: waist circumferenceChange from BL: waist circumference
HIV-1 Infected Participants30.05-0.2055.251.1483.640.63

[back to top]

Absolute Values and Change From Baseline in CD4+ T-cell Counts (%) at Week 24

(NCT02581202)
Timeframe: Baseline, Week 24

Interventionpercentage of lymphocytes that are CD4+ (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants32.500.96

[back to top]

Absolute Values and Change From Baseline in CD4+ T-cell Counts (%) at Week 48

(NCT02581202)
Timeframe: Baseline, Week 48

Interventionpercentage of lymphocytes that are CD4+ (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants33.111.43

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: Alanine Aminotransferase (µmol/L)

(NCT02581202)
Timeframe: Baseline, Week 24

Interventionµmol/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants39.8119.06

[back to top]

Absolute Values and Change From Baseline in CD4+ T-cell Counts (Cells/mm^3) at Week 48

(NCT02581202)
Timeframe: Baseline, Week 48

Interventioncells/mm^3 (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants752.44111.75

[back to top]

Absolute Values and Change From Baseline in HIV-1- RNA Viral Load at Week 48 (Base-10 Logarithm Transformed Data)

(NCT02581202)
Timeframe: Baseline, Week 48

Interventionlog10 copies/mL (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants3.220.000

[back to top]

Absolute Values and Change From Baseline in HIV-1- RNA Viral Load at Week 48 (Untransformed Data)

Statistics for absolute HIV-1 RNA viral load, where all participants with undetectable HIV-1-RNA viral load data were included into calculations with half of the lower indication limit (50/2 copies/mL, or 25.00 copies/mL). (NCT02581202)
Timeframe: Baseline, Week 48

Interventioncopies/mL (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants25.000.00

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: Alanine Aminotransferase (U/L)

(NCT02581202)
Timeframe: Baseline, Week 24

InterventionU/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants29.37-1.31

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: Alanine Aminotransferase (U/L)

(NCT02581202)
Timeframe: Baseline, Week 48

InterventionU/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants28.35-2.14

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: Alanine Aminotransferase (µmol/L)

(NCT02581202)
Timeframe: Baseline, Week 48

Interventionµmol/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants27.566.81

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: Aspartate Aminotransferase (U/L)

(NCT02581202)
Timeframe: Baseline, Week 48

InterventionU/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants26.92-0.49

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 48: Aspartate Aminotransferase (µmol/L)

(NCT02581202)
Timeframe: Baseline, Week 48

Interventionµmol/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants27.195.63

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: Aspartate Aminotransferase (U/L)

(NCT02581202)
Timeframe: Baseline, Week 24

InterventionU/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants26.90-0.38

[back to top]

Absolute Values and Change From Baseline in Metabolic Parameters at Week 24: Aspartate Aminotransferase (µmol/L)

(NCT02581202)
Timeframe: Baseline, Week 24

Interventionµmol/L (Mean)
Absolute valuesChange from BL
HIV-1 Infected Participants46.5625.00

[back to top]

Percentage of Female Participants Responding With SVR12

SVR12 is defined as HCV RNA < LLOQ 12 weeks after the last dose of study drugs without any confirmed quantifiable (≥ LLOQ) post-treatment value before or during that SVR window. Confidence interval calculated using the normal approximation to the binomial distribution. (NCT02582632)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir97.9

[back to top]

Percentage of Female Participants Responding With SVR12: mITT-GT Population

SVR12 is defined as HCV RNA < LLOQ 12 weeks after the last dose of study drugs without any confirmed quantifiable (≥ LLOQ) post-treatment value before or during that SVR window. Confidence interval calculated using the normal approximation to the binomial distribution. (NCT02582632)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir97.8

[back to top]

Percentage of Participants Who Achieve Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

SVR12 is defined as hepatitis C virus (HCV) ribonucleic acid (RNA) < lower limit of quantification (LLOQ) 12 weeks after the last dose of study drugs without any confirmed quantifiable (≥ LLOQ) post-treatment value before or during that SVR window. Confidence interval calculated using the normal approximation to the binomial distribution. (NCT02582632)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir97.6

[back to top]

Percentage of Participants With Post-Treatment Relapse12: mITT-GT Population

Relapse12 is defined as confirmed HCV RNA ≥ LLOQ between end of treatment and 12 weeks after last actual dose of active study drug (up to and including the SVR12 window) excluding reinfection among participants with HCV RNA < LLOQ at final treatment visit who complete treatment and have post-treatment HCV RNA data. Completion of treatment is defined as a study drug duration ≥ 51 days for participants who receive 8 weeks of treatment. HCV reinfection is defined as confirmed HCV RNA ≥ LLOQ after the end of treatment in a participant who had HCV RNA < LLOQ at final treatment visit, along with the post treatment detection of a different HCV genotype, subtype, or clade compared with baseline, as determined by phylogenetic analysis of the NS3 or NS5A, and/or NS5B gene sequences. Confidence interval calculated using the normal approximation to the binomial distribution. (NCT02582632)
Timeframe: Up to 12 weeks after last dose of study drug

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir1.2

[back to top]

Percentage of Participants With Post-Treatment Relapse12

Relapse12 is defined as confirmed HCV RNA ≥ LLOQ between end of treatment and 12 weeks after last actual dose of active study drug (up to and including the SVR12 window) excluding reinfection among participants with HCV RNA < LLOQ at final treatment visit who complete treatment and have post-treatment HCV RNA data. Completion of treatment is defined as a study drug duration ≥ 51 days for participants who receive 8 weeks of treatment. HCV reinfection is defined as confirmed HCV RNA ≥ LLOQ after the end of treatment in a participant who had HCV RNA < LLOQ at final treatment visit, along with the post treatment detection of a different HCV genotype, subtype, or clade compared with baseline, as determined by phylogenetic analysis of the NS3 or NS5A, and/or NS5B gene sequences. Confidence interval calculated using the normal approximation to the binomial distribution. (NCT02582632)
Timeframe: Up to 12 weeks after last dose of study drug

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir1.2

[back to top]

Percentage of Participants With On-Treatment Virologic Failure During Treatment Period: mITT-GT Population

On-treatment virologic failure is defined as breakthrough (confirmed HCV RNA ≥ LLOQ after HCV RNA < LLOQ during treatment, or confirmed increase from nadir in HCV RNA (two consecutive HCV rna measurements > 1 log^10 IU/mL above nadir) at any time point during treatment) or failure to suppress during treatment (all on-treatment values of HCV RNA ≥ LLOQ) with at least 6 weeks (defined as study drug duration ≥ 36 days) of treatment. Confidence interval calculated using the Wilson score method. (NCT02582632)
Timeframe: Up to 8 weeks while on treatment

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir0

[back to top]

Percentage of Participants With On-Treatment Virologic Failure During Treatment Period

On-treatment virologic failure is defined as breakthrough (confirmed HCV RNA ≥ LLOQ after HCV RNA < LLOQ during treatment, or confirmed increase from nadir in HCV RNA (two consecutive HCV rna measurements > 1 log^10 IU/mL above nadir) at any time point during treatment) or failure to suppress during treatment (all on-treatment values of HCV RNA ≥ LLOQ) with at least 6 weeks (defined as study drug duration ≥ 36 days) of treatment. Confidence interval calculated using the normal approximation to the binomial distribution. (NCT02582632)
Timeframe: Up to 8 weeks while on treatment

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir0.6

[back to top]

Percentage of Participants With Baseline HCV RNA < 6,000,000 IU/mL Responding With SVR12: mITT-GT Population

SVR12 is defined as HCV RNA < LLOQ 12 weeks after the last dose of study drugs without any confirmed quantifiable (≥ LLOQ) post-treatment value before or during that SVR window. Confidence interval calculated using the normal approximation to the binomial distribution. (NCT02582632)
Timeframe: Baseline and 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir98.7

[back to top]

Percentage of Participants With Baseline HCV RNA < 6,000,000 IU/mL Responding With SVR12

SVR12 is defined as HCV RNA < LLOQ 12 weeks after the last dose of study drugs without any confirmed quantifiable (≥ LLOQ) post-treatment value before or during that SVR window. Confidence interval calculated using the normal approximation to the binomial distribution. (NCT02582632)
Timeframe: Baseline and 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir98.1

[back to top]

Percentage of Participants Who Achieve SVR12: mITT-GT Population

SVR12 is defined as HCV RNA < LLOQ 12 weeks after the last dose of study drugs without any confirmed quantifiable (≥ LLOQ) post-treatment value before or during that SVR window. Confidence interval calculated using the normal approximation to the binomial distribution. (NCT02582632)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir98.2

[back to top]

Percentage of Participants Achieving Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

"SVR12 was defined as hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL 12 weeks after the last actual dose of study drug. The core population (CP) consisted of participants who met all inclusion criteria and were adequately treated according to the standard of care and within local label recommendations for their specific disease characteristics (cirrhotic status, genotype). The core population with sufficient follow-up data 12 weeks after the last actual dose of study drug (CPSFU12) was defined as all CP participants who fulfilled one of the following criteria:~evaluable HCV RNA data ≥70 days after the last actual dose of the ABBVIE REGIMEN~an HCV RNA value ≥50 IU/mL at the last measurement post-baseline~HCV RNA <50 IU/mL at the last measurement post-baseline, but no HCV RNA measurement ≥70 days after the last actual dose of the ABBVIE REGIMEN due to reasons related to safety (e.g. dropped out due to AE) or virologic failure" (NCT02582671)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Core population (CP)CPSFU12
Participants With HCV Genotype 197.098.0

[back to top]

Percentage of Participants With Virologic Response at End of Treatment (EOT)

Virologic response was defined as hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL at the end of treatment. (NCT02582671)
Timeframe: Up to 24 weeks

InterventionPercentage of participants (Number)
Participants With HCV Genotype 197.0

[back to top]

Percentage of Participants With Relapse

Relapse was defined as hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL at the end of treatment followed by HCV RNA level greater than or equal to 50 IU/mL. (NCT02582671)
Timeframe: From the end of treatment through the end of study (maximum of 48 weeks post-treatment)

Interventionpercentage of participants (Number)
Participants With HCV Genotype 10

[back to top]

Percentage of Participants With On-treatment Virologic Failure

On-treatment virologic failure was defined as breakthrough (at least 1 documented hepatitis C virus ribonucleic acid (HCV RNA) less than 50 IU/mL followed by HCV RNA greater than or equal to 50 IU/mL during treatment) or failure to suppress (each measured on-treatment HCV RNA value greater than or equal to 50 IU/mL). (NCT02582671)
Timeframe: Up to 24 weeks

Interventionpercentage of participants (Number)
Participants With HCV Genotype 10

[back to top]

Percentage of Participants With Missing Sustained Virologic Response 12 Weeks Post-Treatment (SVR12) Data and/or Nonresponders Who Did Not Meet Specific SVR12 Nonresponder Criteria

The number of participants with missing SVR12 data or SVR12 nonresponder participants who did not meet criteria for on-treatment virologic failure, relapse, premature treatment discontinuation, and who did not have an Insufficient virological response reported was documented. (NCT02582671)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 11.0

[back to top]

Percentage of Participants Meeting Relapse Criteria

Relapse was defined as hepatitis C virus ribonucleic acid (HCV RNA) less than 50 IU/mL at end of treatment or at the last on treatment HCV RNA measurement followed by HCV RNA greater than or equal to 50 IU/mL post-treatment. (NCT02582671)
Timeframe: Up to 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 10

[back to top]

Percentage of Participants Meeting Premature Study Drug Discontinuation Criteria

Premature study drug discontinuation was defined as participants who prematurely discontinued study drug with no on-treatment virologic failure. (NCT02582671)
Timeframe: Up to 24 weeks

Interventionpercentage of participants (Number)
Participants With HCV Genotype 12.0

[back to top]

Percentage of Participants With Viral Breakthrough

Viral breakthrough was defined as at least 1 documented hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL followed by HCV RNA level greater than or equal to 50 IU/mL during treatment. (NCT02582671)
Timeframe: Up to 24 weeks

Interventionpercentage of participants (Number)
Participants With HCV Genotype 10

[back to top]

Percentage of Participants With Hemoglobin < 10 g/dL During Treatment

The percentage of participants with hemoglobin <10 g/dL during treatment is provided. (NCT02609659)
Timeframe: up to 12 weeks

Interventionpercentage of participants (Number)
3-DAA + RBV 600 mg0

[back to top]

Percentage of Participants With On-treatment Virologic Failure

On-treatment virologic failure was defined as confirmed HCV RNA ≥ LLOQ after HCV RNA < LLOQ during treatment; confirmed increase of > 1 log(subscript)10(subscript) IU/mL above the lowest value post-baseline in HCV RNA during treatment; or all on-treatment values of HCV RNA ≥ LLOQ with at least 6 weeks of treatment. (NCT02609659)
Timeframe: Up to 12 weeks

Interventionpercentage of participants (Number)
3-DAA + RBV 600 mg1.0

[back to top]

Percentage of Participants With Post-treatment Relapse

Post-treatment relapse was defined as confirmed HCV RNA ≥ LLOQ between the end of treatment and 12 weeks after the last dose of study drug, excluding reinfection, among participants who completed treatment with HCV RNA levels < LLOQ at the end of treatment. (NCT02609659)
Timeframe: From the end of treatment through 12 weeks after the last dose of study drug

Interventionpercentage of participants (Number)
3-DAA + RBV 600 mg4.1

[back to top]

Mean Change in Hemoglobin Values From Baseline to End of Treatment

The mean change in hemoglobin (g/L) from baseline to each study visit and to the final treatment visit (up to 12 weeks) is provided. (NCT02609659)
Timeframe: Baseline (Day 1) to Weeks 2, 4, 8, and 12, and the Final Treatment Visit (up to 12 weeks)

Interventiong/L (Mean)
Week 2Week 4Week 8Week 12Final Treatment Visit
3-DAA + RBV 600 mg-6.4-8.9-11.2-12.4-12.1

[back to top]

Percentage of Participants With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

SVR12 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification [NCT02609659)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
3-DAA + RBV 600 mg89.5

[back to top]

Percentage of Participants in Arm A With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

SVR12 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification [NCT02707952)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Arm A99.2

[back to top]

Percentage of Participants in Arms A and B With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

SVR12 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification [NCT02707952)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Arm A99.1
Arm B100

[back to top]

Percentage of Participants With On-treatment Virologic Failure

On-treatment virologic failure was defined as confirmed increase of > 1 log(subscript)10(subscript) IU/mL above the lowest value post-baseline HCV RNA during treatment; confirmed HCV RNA ≥ 100 IU/mL after HCV RNA < LLOQ during treatment, or HCV RNA ≥ LLOQ at end of treatment with at least 6 weeks of treatment. 95% CI was calculated using the Wilson score method. (NCT02707952)
Timeframe: Treatment Weeks 1, 2, 4, 8 (end of treatment for 8-week treatment arm), and 12 (end of treatment for 12-week treatment arm) or premature discontinuation from treatment

Interventionpercentage of participants (Number)
Arm A0
Arm B0
Arm C1.0
Arm D0

[back to top]

Percentage of Participants With Post-Treatment Relapse

Post-treatment relapse was defined as confirmed HCV RNA ≥ LLOQ between the end of treatment and 12 weeks after the last dose of study drug among participants with HCV RNA levels < LLOQ at the end of treatment, excluding participants who were been shown to be re-infected. The confidence interval was calculated using the Wilson score method. (NCT02707952)
Timeframe: From the end of treatment through 12 weeks after the last dose of study drug

Interventionpercentage of participants (Number)
Arm A0
Arm B0
Arm C3.0
Arm D0

[back to top]

Percentage of Participants for Each Sub-Population in Arms C and D With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

SVR12 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification [NCT02707952)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With Severe RI and without Cirrhosis
Arm D100

[back to top]

Percentage of Participants for Each Sub-Population in Arms C and D With Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

SVR12 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than the lower limit of quantification [NCT02707952)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
HCV GT1 Participants with Compensated CirrhosisHCV GT2 Participants with Compensated CirrhosisDAA Experienced ParticipantsHCV GT3,4,5 and 6 ParticipantsParticipants With Severe RI and with Cirrhosis
Arm C10010093.983.3100.0

[back to top]

Percentage of Participants With On-treatment Virologic Failure

On-treatment virologic failure was defined as breakthrough (at least 1 documented plasma hepatitis C virus ribonucleic acid (HCV RNA) less than 50 IU/mL followed by HCV RNA greater than or equal to 50 IU/mL during treatment) or failure to suppress (each measured on-treatment HCV RNA value greater than or equal to 50 IU/mL). (NCT02725866)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 40.5

[back to top]

Percentage of Participants With Viral Breakthrough

Viral breakthrough was defined as at least 1 documented plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL followed by HCV RNA level greater than or equal to 50 IU/mL during treatment. (NCT02725866)
Timeframe: Up to 24 weeks

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 40.0

[back to top]

Percentage of Participants With Missing Sustained Virologic Response 12 Weeks Post-Treatment (SVR12) Data and/or Nonresponders Who Did Not Meet Specific SVR12 Nonresponder Criteria

The number of participants with missing SVR12 data or SVR12 nonresponder participants who did not meet criteria for on-treatment virologic failure, relapse, premature treatment discontinuation, and who did not have an Insufficient virological response was documented. (NCT02725866)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 43.8

[back to top]

Percentage of Participants Achieving Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

"SVR12 was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL 12 weeks after the last actual dose of study drug. The core population (CP) consisted of participants who met all inclusion criteria and were adequately treated according to the standard of care and within local label recommendations for their specific disease characteristics (cirrhotic status, genotype). The core population with sufficient follow-up data 12 weeks after the last actual dose of study drug (CPSFU12) was defined as all CP participants who fulfilled one of the following criteria:~evaluable HCV RNA data ≥70 days after the last actual dose of the ABBVIE REGIMEN~an HCV RNA value ≥50 IU/mL at the last measurement post-baseline~HCV RNA <50 IU/mL at the last measurement post-baseline, but no HCV RNA measurement ≥70 days after the last actual dose of the ABBVIE REGIMEN due to reasons related to safety (e.g. dropped out due to AE) or virologic failure" (NCT02725866)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Core population (CP)CPSFU12
Participants With HCV Genotype 1 or 491.195.6

[back to top]

Percentage of Participants With Relapse

Relapse was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL at the end of treatment followed by HCV RNA level greater than or equal to 50 IU/mL. (NCT02725866)
Timeframe: Up to 24 weeks

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 40.9

[back to top]

Percentage of Participants Meeting Relapse Criteria

Relapse was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) less than 50 IU/mL at end of treatment or at the last on-treatment HCV RNA measurement followed by HCV RNA greater than or equal to 50 IU/mL post-treatment. (NCT02725866)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 40.9

[back to top]

Percentage of Participants Meeting Premature Study Drug Discontinuation Criteria

Premature study drug discontinuation was defined as participants who prematurely discontinued study drug with no on-treatment virologic failure. (NCT02725866)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 42.3

[back to top]

Percentage of Participants With Virologic Response at End of Treatment (EoT)

Virologic response was defined as plasma hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL at the end of treatment. (NCT02725866)
Timeframe: Up to 24 weeks

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 493.9

[back to top]

Mean Change in Fatigue PRO Score -Phase 1

Fatigue severity collected from validated, Patient Reported Outcomes survey (PROs), 'PROMIS Fatigue Short Form'. PROMIS® T-scores were computated to compare difference between baseline value of PROMIS score to the highest (worst) score during treatment. Results presented as computated t-score from baseline to average of on Treatment Scores. A positive (+) score suggests improvements in functional well-being. A negative (-) PRO change score is suggestive of symptom improvement or lack of drug side effect. PROMIS Fatigue Score scale per question: 1=Never, 2=Rarely, 3=Sometimes, 4=Often, 5=Always with 7 questions for a total maximum score of 35. (NCT02786537)
Timeframe: Baseline to On-treatment

Interventionunits on a scale (Mean)
EBR/GZR With RBV1.5
EBR/GZR-1.2
SOF/LDV With RBV-7.2
SOF/LDV-2.0
PrOD With RBV-1.9
PrOD-3.0

[back to top]

Mean Change in HCV- PRO- Phase 1

"HCV-PRO, a survey for patients with HCV that measures physical, emotional, and social functioning, productivity, intimacy, and perception of quality of life, was used to assess 'overall functioning and well-being'. In general, lower score is worst outcome and higher scores indicate greater well-being and functioning. However, for ease of interpretation, HCV-PRO scale has been transformed by using '100 - HCV-PRO' ultimately revising the score to mean 0 (lowest score) is best to 100 (worst outcome). A positive change (End of treatment to baseline) suggests improvements in functional well-being.~Total score = (SUM-N)/(4*N)*100, where N is the number of questions answered." (NCT02786537)
Timeframe: Baseline to End of Treatment

Interventionunits on a scale (Mean)
EBR/GZR With RBV-0.9
EBR/GZR5.6
SOF/LDV With RBV2.5
SOF/LDV6.9
PrOD With RBV3.2
PrOD9.9

[back to top]

Mean Change in HCV-PRO (Functional Well-being) EBR/GZR vs. SOF/LDV Score-Phase 2

"HCV-PRO, a survey designed to assess functional status of patients with HCV and measures physical, emotional, and social functioning, productivity, intimacy, and perception of quality of life, was used to assess functional well-being. In general, lower score is worst outcome and higher scores indicate greater well-being and functioning. However, for ease of interpretation, HCV-PRO scale has been transformed by using '100 - HCV-PRO' ultimately revising the score to mean 0 (lowest score) is best to 100 (worst outcome). A positive change (End of treatment to baseline) suggests improvements in functional well-being.~Total score = (SUM-N)/(4*N)*100, where N is the number of questions answered. End of Treatment -Baseline were used to calculate CHANGE in outcome. Number of subjects reflects participants from both Phase 1 and 2." (NCT02786537)
Timeframe: End of Treatment - Baseline

Interventionscore on a scale (Mean)
EBR/GZR With RBV3.2
EBR/GZR6.1
SOF/LDV With RBV6.3
SOF/LDV6.8

[back to top]

Mean Change in Headache-EBR/GZR and SOF/LDV

Headache was evaluated by the HIT-6 score, a validated, Patient Reported Outcomes survey (PROs) 'PROMIS Headache Impact Test (HIT)' with scores ranging from 36 to 78 with higher score reflecting greater impact. Mean change in headache side effect was evaluated using difference between baseline value of HIT-6 score to the highest (worst) score during treatment. Estimates of mean change and differences obtained from a constrained longitudinal linear mixed-effects model that treated baseline score as one of outcomes. Negative values for mean change represent improvement, while negative values for 'difference' indicate LDV/SOF performed better than PrOD (NCT02786537)
Timeframe: Baseline to On-Treatment

Interventionunits on a scale (Mean)
EBR/GZR With RBV-0.8
EBR/GZR-0.7
SOF/LDV With RBV0.4
SOF/LDV-0.8

[back to top]

Mean Change in Headache-PRO Scores -Phase 1

Headache was evaluated by the HIT-6 score, a validated, Patient Reported Outcomes survey (PROs) 'PROMIS Headache Impact Test (HIT)' with scores ranging from 36 to 78 with higher score reflecting greater impact. Mean change in headache side effect was evaluated using difference between baseline value of HIT-6 score to the highest (worst) score during treatment. Estimates of mean change and differences obtained from a constrained longitudinal linear mixed-effects model that treated baseline score as one of outcomes. Negative values for mean change represent improvement in symptom. (NCT02786537)
Timeframe: Baseline to On-Treatment

Interventionunits on a scale (Mean)
EBR/GZR With Ribavirin (RBV)0.0
EBR/GZR Regimen-0.8
SOF/LDV With RBV-0.7
SOF/LDV-0.5
PrOD With RBV Regimen-0.2
PrOD-2.2

[back to top]

Mean Change in Nausea/Vomiting PRO Score -Phase 1

"Patients completed the PROMIS® Nausea Short Form at Baseline (T1) and on-treatment. PROMIS raw scores from each of the completed questionnaires were converted to standardized T-scores. Change was calculated as the difference between baseline and on-treatment score. T-scores for the PROMIS Nausea and Vomiting 4a scale range from 45.0 - 80.1. Higher scores indicate worse nausea. Negative values for mean change represent improvement.~The estimates of mean change and differences were obtained from a constrained longitudinal linear mixed-effects model that treated the baseline score as one of the outcomes. The model expressed mean score as a function of DAA regimen, cirrhosis status, HCV genotype, sex, age, race, and previous treatment status." (NCT02786537)
Timeframe: Baseline to On-Treatment

Interventionunits on a scale (Mean)
EBR/GZR With RBV1.3
EBR/GZR-1.4
SOF/LDV With RBV-3.9
SOF/LDV-0.7
PrOD With RBV2.5
PrOD0.7

[back to top]

Mean Change in Nausea/Vomiting PROMIS Score -EBR/GZR vs. LDV/SOF

"Participants completed the Patient Reported Outcomes surveys (PROs) 'PROMIS Nausea/Vomiting -4 Short Form' at baseline and during treatment. Raw scores are converted to standardized T-scores with a range of 45.0-80.1. Higher scores indicate worse nausea/vomiting.~Results presented as mean difference from baseline to average of on Treatment Scores (highest/worst) score during treatment.~A negative (-) PROMIS change score is suggestive of symptom improvement or lack of drug side effect. Estimates of mean change were obtained from a constrained longitudinal linear mixed-effects model that treated the baseline score as one of the outcomes." (NCT02786537)
Timeframe: Baseline and Average On-Treatment Score

Interventionunits on a scale (Mean)
EBR/GZR With RBV-0.3
EBR/GZR-0.6
SOF/LDV With RBV-1.6
SOF/LDV-0.4

[back to top]

Median Change in Fatigue -Phase 1

Fatigue severity collected from validated, Patient Reported Outcomes survey (PROs), 'PROMIS Fatigue Short Form'. PROMIS® T-scores were computated to compare difference between baseline value of PROMIS score to the highest (worst) score during treatment. Results presented as computated t-score from baseline to average of on Treatment Scores. A positive (+) score suggests improvements in functional well-being. A negative (-) PRO change score is suggestive of symptom improvement or lack of drug side effect. PROMIS Fatigue Score scale per question: 1=Never, 2=Rarely, 3=Sometimes, 4=Often, 5=Always with 7 questions for a total maximum score of 35. (NCT02786537)
Timeframe: Baseline to End of Treatment

Interventionunits on a scale (Median)
EBR/GZR With RBV2.2
EBR/GZR Regimen-0.9
SOF/LDV With RBV-10.2
SOF/LDV-3.4
PrOD Regimen With RBV-0.2
PrOD-4.1

[back to top]

Median Change in Fatigue-Phase 2

Fatigue severity collected from validated, Patient Reported Outcomes survey (PROs), 'PROMIS Fatigue Short Form'. PROMIS® T-scores were computated to compare difference between baseline value of PROMIS score to the highest (worst) score during treatment. Results presented as computated t-score from baseline to average of on Treatment Scores. A positive (+) score suggests improvements in functional well-being. A negative (-) PRO change score is suggestive of symptom improvement or lack of drug side effect. PROMIS Fatigue Score scale per question: 1=Never, 2=Rarely, 3=Sometimes, 4=Often, 5=Always with 7 questions for a total maximum score of 35. (NCT02786537)
Timeframe: Baseline-On Treatment (up to 16 weeks)

Interventionunits on a scale (Median)
EBR/GZR With RBV-1.3
EBR/GZR-1.2
SOF/LDV With RBV-2.4
SOF/LDV-1.4

[back to top]

Median Change in HCV-PRO (Overall Well Being) -Phase 1

"HCV-PRO, a survey for patients with HCV that measures physical, emotional, and social functioning, productivity, intimacy, and perception of quality of life, was used to assess 'Overall Functioning and Well-being'. In general, lower score is worst outcome and higher scores indicate greater well-being and functioning. However, for ease of interpretation, HCV-PRO scale has been transformed by using '100 - HCV-PRO' ultimately revising the score to mean 0 (lowest score) is best to 100 (worst outcome). A positive change (End of treatment to baseline) suggests improvements in functional well-being.~Total score = (SUM-N)/(4*N)*100, where N is the number of questions answered." (NCT02786537)
Timeframe: Baseline to End of Treatment

Interventionscore on a scale (Median)
EBR/GZR With RBV0.0
EBR/GZR4.3
SOF/LDV With RBV4.7
SOF/LDV4.7
PrOD With RBV3.1
PrOD8.6

[back to top]

Median Change in Headache -Phase 1

Headache was evaluated by the HIT-6 score, a validated, Patient Reported Outcomes survey (PROs) 'PROMIS Headache Impact Test (HIT)' with scores ranging from 36 to 78 with higher score reflecting greater impact. Median change in headache side effect was evaluated using difference between baseline value of HIT-6 score to the highest (worst) score during treatment. Estimates of mean change and differences obtained from a constrained longitudinal linear mixed-effects model that treated baseline score as one of outcomes. Negative values for change represent improvement, while negative values for 'difference' indicate LDV/SOF performed better than PrOD (NCT02786537)
Timeframe: 12 weeks (Baseline and Average On-treatment Score)

Interventionunits on a scale (Median)
EBR/GZR (Elbasvir/Grazoprevir) With RBV0.0
EBR/GZR (Elbasvir/Grazoprevir)0.0
SOF/LDV (Sofosbuvir/Ledipasvir) With RBV-2.0
SOF/LDV (Sofosbuvir/Ledipasvir)-1.0
PrOD (Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir) With RBV (Phase 1 Only)0.0
PrOD (Ombitasvir/Paritaprevir/Ritonavir and Dasabuvir)-1.0

[back to top]

Median Change in Headache-Phase 2

Headache was evaluated by the HIT-6 score, a validated, Patient Reported Outcomes survey (PROs) 'PROMIS Headache Impact Test (HIT)' with scores ranging from 36 to 78 with higher score reflecting greater impact. Median change in headache side effect was evaluated using difference between baseline value of HIT-6 score to the highest (worst) score during treatment. Estimates of median change and differences obtained from a constrained longitudinal linear mixed-effects model that treated baseline score as one of outcomes. Negative values for mean change represent improvement, while negative values for 'difference' indicate LDV/SOF performed better than PrOD (NCT02786537)
Timeframe: Baseline -on Treatment (12-16 weeks)

Interventionunits on a scale (Median)
EBR/GZR With RBV-1.0
EBR/GZR0.0
SOF/LDV With RBV0.5
SOF/LDV-0.5

[back to top]

Median Change in Nausea PRO Score -Phase 1

"Patients completed the PROMIS® Nausea Short Form at Baseline (T1) and on-treatment. PROMIS raw scores from each of the completed questionnaires were converted to standardized T-scores. Change was calculated as the difference between baseline and on-treatment score. T-scores for the PROMIS Nausea and Vomiting 4a scale range from 45.0 - 80.1. Higher scores indicate worse nausea. Negative values for mean change represent improvement.~The estimates of change and differences were obtained from a constrained longitudinal linear mixed-effects model that treated the baseline score as one of the outcomes." (NCT02786537)
Timeframe: Baseline to end of treatment

Interventionunits on a scale (Median)
EBR/GZR With RBV0.4
EBR/GZR0.0
SOF/LDV With RBV-6.1
SOF/LDV0.0
PrOD With RBV0.0
PrOD0.0

[back to top]

Median Change in Nausea PROMIS Score-EBR/GZR SOF/LDV

"Patients completed the PROMIS® Nausea Short Form at Baseline (T1) and on-treatment. PROMIS raw scores from each of the completed questionnaires were converted to standardized T-scores. Change was calculated as the difference between baseline and on-treatment score. T-scores for the PROMIS Nausea and Vomiting 4a scale range from 45.0 - 80.1. Higher scores indicate worse nausea. Negative values for change represent improvement.~The estimates of change and differences were obtained from a constrained longitudinal linear mixed-effects model that treated the baseline score as one of the outcomes." (NCT02786537)
Timeframe: Baseline- On Treatment (up to 16 weeks)

Interventionscore on a scale (Median)
EBR/GZR With RBV0.0
EBR/GZR0.0
SOF/LDV With RBV0.0
SOF/LDV0.0

[back to top]

Number of Participants With Adverse Events That Caused Treatment Discontinuation-EBR/GZR vs. LDV/SOF

The number of participants with adverse events that led to early treatment discontinuation (defined as duration less than originally prescribed treatment regimen) (NCT02786537)
Timeframe: Treatment start date through treatment completion (up to 24 weeks)

InterventionParticipants (Count of Participants)
EBR/GZR Regimen12
SOF/LDV Regimen4

[back to top]

Phase 1 Number of Participants With Sustained Virologic Response (SVR12-mITT Without Imputation)

"SVR (Sustained Virologic Response) 12 will be defined as undetectable hepatitis C virus (HCV) RNA at 12 week follow-up visit (12 -24 weeks after HCV treatment discontinuation as dictated by standard of care at each individual site).~Number of subjects reflects participants randomized during Phase 1 only." (NCT02786537)
Timeframe: 12 -24 weeks post-treatment

InterventionParticipants (Count of Participants)
EBR/GZR With RBV9
EBR/GZR108
SOF/LDV With RBV6
SOF/LDV88
PrOD With RBV77
PrOD (Phase 1 Only)42

[back to top]

Phase 1-Sustained Virologic Response (SVR12) mITT With Imputation

"SVR (Sustained Virologic Response) 12 will be defined as patients who have undetectable hepatitis C virus (HCV) RNA at 12 week follow-up visit (12 -24 weeks after HCV treatment discontinuation as dictated by standard of care at each individual site).~mITT with imputation (missing=failure). Total number of subjects reflects participants from Phase 1 only." (NCT02786537)
Timeframe: 12 weeks post-treatment

InterventionParticipants (Count of Participants)
EBR/GZR With RBV9
EBR/GZR108
SOF/LDV With RBV6
SOF/LDV88
PrOD With RBV77
PrOD42

[back to top]

Phase 1/2 Number of Participants With Sustained Virologic Response (SVR12-mITT Without Imputation)

"SVR (Sustained Virologic Response) 12 will be defined as undetectable hepatitis C virus (HCV) RNA at 12 week follow-up visit (12 -24 weeks after HCV treatment discontinuation as dictated by standard of care at each individual site).~Number of subjects reflects participants who started EBR/GZR or SOF/LDV- based treatment (with or without RBV) during Phase 1 and 2." (NCT02786537)
Timeframe: 12-24 weeks post HCV treatment

InterventionParticipants (Count of Participants)
EBR/GZR With RBV40
EBR/GZR516
SOF/LDV With RBV14
SOF/LDV335

[back to top]

Post-treatment Progression/Regression of Liver Disease-Fib-4

Mean change (delta) in FIB-4, an indirect non-invasive measure of liver fibrosis, calculated as baseline median -long term follow up median, following SVR after any of the study treatment regimens. Change in FIB-4 where negative values indicate improvement in liver fibrosis score and positive values indicate worsening of fibrosis score. There is no upper or lower limit for change. FIB-4 = age (years) * AST(IU/L)/Platelets (10^3/L) * ALT^.5(IU/L). In general, Score of 0-1.29 is low risk for advanced fibrosis, 1.30-1.67: intermediate risk for advanced liver fibrosis, >2.67: high risk for advanced fibrosis. (NCT02786537)
Timeframe: Baseline to up to 3 years post treatment discontinuation

Interventionscore on a scale (Median)
EBR/GZR, SOF/LDV or PrOD Based HCV Treatment-1.36

[back to top]

Sustained Virologic Response (SVR12) mITT With Imputation-Phase 1 and 2 EBR/GZR, SOF/LDV

"SVR (Sustained Virologic Response) 12 will be defined as undetectable hepatitis C virus (HCV) RNA at 12 week follow-up visit (12 -24 weeks after HCV treatment discontinuation at discretion of provider).~mITT with imputation (missing=failure). Total number of subjects reflects participants from EBR/GZR with or without RBV and SOF/LDV with or without RBV randomized during Phase 1 and Phase 2." (NCT02786537)
Timeframe: 12 weeks post-treatment

InterventionParticipants (Count of Participants)
EBR/GZR With RBV40
EBR/GZR516
SOF/LDV With RBV14
SOF/LDV335

[back to top]

Treatment Non-Adherence Probability Estimates

"The Voils Medication Adherence Survey (VMAS) was used to evaluate medication adherence during HCV treatment. Participants responded to three questions about the extent of adherence during the past seven days of treatment (during early and late on-treatment occasions). Participants responded using a five-point ordinal scale of missed dosing from 1 (none of the time) to 5 (all of the time). On each occasion participants were coded as being Non-adherent if any response was > 1, otherwise they were coded as Adherent. Probability estimates of percentage of patients reporting non-adherence were calculated per HCV treatment (Direct Acting Antiviral-DAA) regimen: 1)EBR/GZV (elbasvir/grazoprevir, 2)SOF/LDV (sofosbuvir/ledipasvir), 3)PrOD" (NCT02786537)
Timeframe: 12-16 weeks of HCV treatment

Interventionpercentage of patients (Number)
EBR/GZR23
SOF/LDV19
PrOD26

[back to top]

Change in Functional Status (HCV-PRO) Within Treatment

"HCV-PRO score, a validated PROMIS survey used to evaluate overall functioning and well-being in HCV patients, was utilized to compare long-term 'within treatment' changes of functional well-being. In general, lower score is worst outcome and higher scores indicate greater well-being and functioning. However, for ease of interpretation, HCV-PRO scale has been transformed by using '100 - HCV-PRO' ultimately revising the score to mean 0 (lowest score) is best to 100 (worst outcome). A positive estimate (Post treatment to baseline) suggests baseline functional well-being improvement.~Total score = (SUM-N)/(4*N)*100, where N is the number of questions answered." (NCT02786537)
Timeframe: Treatment start date up to 2 years post-treatment

,
Interventionscore on a scale (Mean)
9 months post treatment20 months post treatment
EBR/GZR Regimen8.029.87
SOF/LDV Regimen9.9011.54

[back to top]

Change in HCV-associated Symptoms (PROMIS Measures) After HCV Treatment Initiation

Change in HCV-associated symptoms was calculated as the mean differences of mean scores from multiple surveys from the NIH Patient-Reported Outcomes Measurement Information System (PROMIS)-- Fatigue, nausea, belly pain, sleep disturbance, and diarrhea) and functional status (well-being) when comparing baseline to early post-treatment and late post treatment surveys. Mean change scores were calculated by comparing baseline to early post-treatment (1 yr) and late post-treatment (approximately 3 years) surveys. T-scores for the PROMIS Nausea and Vomiting 4a scale range from 45.0 - 80.1. Higher scores indicate worse nausea. Negative values for mean change represent improvement.Negative numbers suggest better symptoms (improvement in HCV-associated symptoms). PROMIS Fatigue Score scale per question: 1=Never, 2=Rarely, 3=Sometimes, 4=Often, 5=Always with 7 questions for a total maximum score of 35.HCV-PRO positive estimates suggest baseline functional well-being improvement. (NCT02786537)
Timeframe: 1 year post treatment discontinuation (Early post-tx)

,
Interventionunits on a scale (Mean)
NauseaBelly PainDiarrheaFatigueSleep DisturbanceCognitive ImpairmentHCV-PRO
EBR/GZR Regimen0.00-0.82-1.12-2.080.65-0.548.02
SOF/LDV Regimen-4.99-6.47-5.77-7.59-1.72-4.489.90

[back to top]

Impact of Baseline NS5A RASs on Treatment Outcomes-Phase 2

Number of participants who achieved SVR (sustained virologic response), defined as undetectable HCV RNA 12 weeks post-treatment with RASs (Resistant Associated Substitutions) after treatment with EBR/GZR or SOF/LDV regimen (NCT02786537)
Timeframe: 12 weeks post HCV treatment

,
InterventionParticipants (Count of Participants)
With NS5a RASWithout NS5a RAS
EBR/GZR Regimen47485
SOF/LDV Regimen42286

[back to top]

16 Wk EBR/GZR With RBV Efficacy on Patients With HCV RASs

"Efficacy of Hepatitis C Virus (HCV) Treatment with Zepatier (Elbasvir/Grazobevir) with Ribavirin (RBV) for 16 weeks when used in Genotype 1a patients with Baseline RASs (NS5a Resistance Associated Substitutions or RAPs -Resistance Associated Polymorphisms).~Efficacy defined as HCV RNA undetectable 12 weeks post treatment. Table excludes Genotype 1b patients." (NCT02786537)
Timeframe: 12 weeks post treatment

InterventionParticipants (Count of Participants)
EBR/GZR With RBV-16 Weeks34

[back to top]

HCV SVR Durability -No Cirrhosis

Number/Percentage of patients with persistence of viral cure, SVR (SVR = Sustained Virologic Response)- defined as undetectable HCV RNA at least 24 weeks following HCV Treatment. (NCT02786537)
Timeframe: 24 weeks post-end of treatment up to 153 weeks

InterventionParticipants (Count of Participants)
EBR/GZR255
EBR/GZR With Ribavirin17
SOF/LDV146
SOF/LDV With RBV2
PrOD14
PrOD With RBV36

[back to top]

HCV SVR Durability-Patients With Cirrhosis

Percentage of Cirrhotic patients with persistence of viral cure, SVR, (SVR= Sustained Virologic Response) defined as undetectable HCV RNA at least 24 weeks following HCV Treatment. (NCT02786537)
Timeframe: Up to 132 weeks post HCV treatment

InterventionParticipants (Count of Participants)
EBR/GZR43
EBR/GZR With RBV7
SOF/LDV35
SOF/LDV With RBV7
PrOD6
PrOD With RBV7

[back to top]

Mean Change in Fatigue PRO -EBR/GZR vs SOF/LDV

Fatigue severity collected from validated, Patient Reported Outcomes survey (PROs), 'PROMIS Fatigue Short Form'. PROMIS® T-scores were computated to compare difference between baseline value of PROMIS score to the highest (worst) score during treatment. Results presented as computated t-score from baseline to average of on Treatment Scores. A positive (+) score suggests improvements in functional well-being. A negative (-) PRO change score is suggestive of symptom improvement or lack of drug side effect. PROMIS Fatigue Score scale per question: 1=Never, 2=Rarely, 3=Sometimes, 4=Often, 5=Always with 7 questions for a total maximum score of 35. (NCT02786537)
Timeframe: Baseline and Average On-Treatment Score

Interventionscore on a scale (Mean)
EBR/GZR With RBV-1.0
EBR/GZR-2.1
SOF/LDV With RBV-3.7
SOF/LDV-2.2

[back to top]

Percentage of Participants With Viral Breakthrough

Viral breakthrough was defined as at least 1 documented hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL followed by HCV RNA level greater than or equal to 50 IU/mL during treatment. (NCT02803138)
Timeframe: Up to 24 weeks

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 40.0

[back to top]

Percentage of Participants With Relapse

Relapse was defined as hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL at the end of treatment followed by HCV RNA level greater than or equal to 50 IU/mL. (NCT02803138)
Timeframe: Up to 48 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 40.0

[back to top]

Percentage of Participants With Sufficient Follow-up Who Achieved Sustained Virological Response 12 Weeks Posttreatment

"Sustained virologic response was defined as hepatitis C virus ribonucleic acid (HCV RNA) levels less than 50 IU/mL 12 weeks after the last dose of study drug.~The core population with sufficient follow-up data 12 weeks after the last actual dose of study drug (CPSFU12) was defined as all participants who fulfilled one of the following criteria:~evaluable HCV RNA data ≥70 days after the last actual dose of the ABBVIE REGIMEN~an HCV RNA value ≥50 IU/mL at the last measurement post-baseline~HCV RNA <50 IU/mL at the last measurement post-baseline, but no HCV RNA measurement ≥70 days after the last actual dose of the ABBVIE REGIMEN due to reasons related to safety (e.g. dropped out due to AE) or virologic failure" (NCT02803138)
Timeframe: 12 weeks (at least 70 days) after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 495.8

[back to top]

Percentage of Participants With Virologic Response at End of Treatment (EoT)

Virologic response was defined as hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL at the end of treatment. (NCT02803138)
Timeframe: Up to 24 weeks

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 482.0

[back to top]

Percentage of Participants Achieving Sustained Virologic Response 12 Weeks Post-treatment (SVR12)

SVR12 was defined as hepatitis C virus ribonucleic acid (HCV RNA) level less than 50 IU/mL 12 weeks after the last actual dose of study drug. (NCT02803138)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 470.6

[back to top]

Percentage of Participants Meeting Premature Study Drug Discontinuation Criteria

Premature study drug discontinuation was defined as participants who prematurely discontinued study drug with no on-treatment virologic failure. (NCT02803138)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 41.8

[back to top]

Percentage of Participants Meeting Relapse Criteria

Relapse was defined as hepatitis C virus ribonucleic acid (HCV RNA) less than 50 IU/mL at end of treatment or at the last on-treatment HCV RNA measurement followed by HCV RNA greater than or equal to 50 IU/mL post-treatment. (NCT02803138)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 40

[back to top]

Percentage of Participants With Missing Sustained Virologic Response 12 Weeks Post-Treatment (SVR12) Data and/or Nonresponders Who Did Not Meet Specific SVR12 Nonresponder Criteria

The number of participants with missing SVR12 data or SVR12 nonresponder participants who did not meet criteria for on-treatment virologic failure, relapse, premature treatment discontinuation, and who did not have an Insufficient virological response reported was documented. (NCT02803138)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 425.9

[back to top]

Percentage of Participants With On-treatment Virologic Failure

On-treatment virologic failure was defined as breakthrough (at least 1 documented hepatitis C virus ribonucleic acid (HCV RNA) less than 50 IU/mL followed by HCV RNA greater than or equal to 50 IU/mL during treatment) or failure to suppress (each measured on-treatment HCV RNA value greater than or equal to 50 IU/mL). (NCT02803138)
Timeframe: 12 weeks after the last actual dose of study drug

Interventionpercentage of participants (Number)
Participants With HCV Genotype 1 or 41.3

[back to top]

Percentage of Subjects With Sustained Virologic Response (SVR12) 12 Weeks Post-treatment

SVR12, defined as undetectable HCV RNA 12 weeks after the last day of study drug administration (NCT03020004)
Timeframe: 24 weeks

InterventionParticipants (Count of Participants)
Danoprevir,Ritonavir, Peg-IFN,RBV66

[back to top]

Proportion of Participants With Sustained Virologic Response 12 Weeks After Discontinuation of Therapy (SVR12)

(NCT03020082)
Timeframe: 12 weeks

InterventionParticipants (Count of Participants)
Danoprevir, Ritonavir, Peg-IFN,RBV136

[back to top]

Percentage of Subjects With Sustained Virologic Response (SVR12) 12 Weeks Post-treatment

SVR12, defined as undetectable HCV RNA 12 weeks after the last day of study drug administration. (NCT03020095)
Timeframe: 12 weeks

InterventionParticipants (Count of Participants)
Ravidasvir,Danoprevir/r,RBV38

[back to top]

"Change From Baseline J-Tpeakc With Balanced Ion Channel Drugs (Ranolazine, Verapamil, Lopinavir / Ritonavir)"

"The primary outcome measure for the balanced ion channel drugs (ranolazine, verapamil, lopinavir / ritonavir) is for the upper bound of the 2-sided 90% confidence interval (CI) to be <10 msec for the projected placebo-corrected change from baseline J-Tpeakc effect at the peak plasma level on Day 3 using a linear mixed-effects exposure response model. Placebo drug concentration was set to 0 (see SAP)." (NCT03070470)
Timeframe: 3 days

Interventionms (Median)
Ranolazine-8.3
Verapamil-7.8
Lopinavir / Ritonavir-11.5
Placebo-10.9

[back to top]

"Change From Baseline QTc With Predominant hERG Blocking Drug (Chloroquine)"

"The primary outcome measure for the predominant hERG drug (chloroquine) is for the upper bound of the 2-sided 90% CI to be ≥10 msec for the projected placebo-corrected change from baseline QTc effect at the peak plasma level on Day 1 using a linear mixed-effects exposure response model" (NCT03070470)
Timeframe: 3 days

Interventionms (Median)
Chloroquine17.7
Placebo-9.3

[back to top]

QTc Shortening From Calcium Block (Diltiazem) in the Presence of hERG Block (Dofetilide)

"It will be assessed whether the projected QTc effect of dofetilide alone is significantly greater (i.e., p<0.05) than the projected QTc effect of the combination of dofetilide + diltiazem. This will be assessed at the dofetilide peak plasma level on Day 3 (computed from the combination of dofetilide + diltiazem) on the pooled dofetilide alone, diltiazem alone, and dofetilide + diltiazem data using a linear mixed effects model.~Subsequently, and if the test is significant for QTc, the same test will be performed to assess calcium block (diltiazem) effects on J-Tpeakc." (NCT03070470)
Timeframe: 3 days

Interventionms (Median)
Dofetilide2.2
Diltiazem+Dofetilide-1.7

[back to top]

Part 1: AUC (0-infinity) Following Administration of GSK2838232 Tablet in Fasted and Fed State

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as tablet formulation in fed and fasted state. Pharmacokinetic parameters were determined using standard non-compartmental methods. (NCT03234036)
Timeframe: Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96, and 120 hours post-dose in treatment periods 1, 2 and 3 of Part 1

InterventionHours*nanogram per milliliter (Geometric Mean)
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed4437.98
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fasted1816.27

[back to top]

Part 1: Area Under the Concentration-time Curve From Time Zero (Pre-dose) Extrapolated to Infinite Time (AUC [0-infinity]) Following Administration of GSK2838232 Tablet and Capsule Formulation in Fed State

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as tablet and capsule formulation in fed state. Pharmacokinetic parameters were determined using standard non-compartmental methods. Pharmacokinetic Population comprised of all participants in the Safety Population who had at least 1 non-missing pharmacokinetic assessment (Non-quantifiable [NQ] values were considered as non-missing values). (NCT03234036)
Timeframe: Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96, and 120 hours post-dose in treatment periods 1 and 2 of Part 1

InterventionHours*nanogram per milliliter (Geometric Mean)
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed4672.28
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed4437.98

[back to top]

Part 1: Change From Baseline in Blood Pressure

SBP and DBP were measured in supine position after 10 minutes rest for the participant at indicated time points. Baseline was defined as the latest pre-dose assessment, including those from unscheduled visits. Change from Baseline was defined as any visit value minus Baseline value. (NCT03234036)
Timeframe: Baseline (Day -1) and 1, 2, 4, 6, 8, 12, 24, 48, 72 Hours on Day 1

,,
InterventionMillimeter of mercury (Mean)
DBP; DAY 1, 1 HourDBP; DAY 1, 2 HoursDBP; DAY 1, 4 HoursDBP; DAY 1, 6 HoursDBP; DAY 1, 8 HoursDBP; DAY 1, 12 HoursDBP; DAY 1, 24 HoursDBP; DAY 1, 48 HoursDBP; DAY 1, 72 HoursSBP; DAY 1, 1 HourSBP; DAY 1, 2 HoursSBP; DAY 1, 4 HoursSBP; DAY 1, 6 HoursSBP; DAY 1, 8 HoursSBP; DAY 1, 12 HoursSBP; DAY 1, 24 HoursSBP; DAY 1, 48 HoursSBP; DAY 1, 72 Hours
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed-2.756-3.823-2.223-1.5562.3772.9112.4440.6440.644-2.156-3.289-0.4230.1773.9111.977-1.956-1.289-4.089
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fasted0.429-0.8570.500-0.8571.071-0.7140.000-2.286-2.500-0.358-0.644-0.1441.356-0.8580.928-1.358-4.8580.571
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed-2.112-3.712-1.845-2.1790.6881.5550.2881.0880.8210.111-3.222-2.289-1.3552.3114.711-0.822-1.222-2.355

[back to top]

Part 1: Blood Pressure at Indicated Time Points

SBP and DBP of participants were measured in supine position after 10 minutes rest at indicated time points. (NCT03234036)
Timeframe: Day -2; Day -1; Pre-dose, 1, 2, 4, 6, 8, 12, 24, 48, 72 Hours on Day 1

,,
InterventionMillimeter of mercury (Mean)
DBP; DAY -2DBP; DAY -1DBP; DAY 1, PREDOSEDBP; DAY 1, 1 HourDBP; DAY 1, 2 HoursDBP; DAY 1, 4 HoursDBP; DAY 1, 6 HoursDBP; DAY 1, 8 HoursDBP; DAY 1, 12 HoursDBP; DAY 1, 24 HoursDBP; DAY 1, 48 HoursDBP; DAY 1, 72 HoursSBP; DAY -2SBP; DAY -1SBP; DAY 1, PREDOSESBP; DAY 1, 1 HourSBP; DAY 1, 2 HoursSBP; DAY 1, 4 HoursSBP; DAY 1, 6 HoursSBP; DAY 1, 8 HoursSBP; DAY 1, 12 HoursSBP; DAY 1, 24 HoursSBP; DAY 1, 48 HoursSBP; DAY 1, 72 Hours
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed68.66767.46765.95663.20062.13363.73364.40068.33368.86768.40066.60066.600108.667110.067108.156106.000104.867107.733108.333112.067110.133106.200106.867104.067
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fasted65.00065.64366.92967.35766.07167.42966.07168.00066.21466.92964.64364.429105.786108.429106.858106.500106.214106.714108.214106.000107.786105.500102.000107.429
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed69.53368.46766.24564.13362.53364.40064.06766.93367.80066.53367.33367.067109.733104.000108.289108.400105.067106.000106.933110.600113.000107.467107.067105.933

[back to top]

Part 2: Observed Concentration at the End of the Dosing Interval (Ctau) Following Administration of Non-boosted Once-daily Doses of GSK2838232 Tablet in Fed State

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as a non-boosted once-daily dosing tablet in fed state. Pharmacokinetic parameters were determined using standard non-compartmental methods. (NCT03234036)
Timeframe: Pre-dose, 0.5,1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12 and 24 hours post-dose on Day 1; Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96 and 120 hours post-dose on Day 11 in Part 2

InterventionNanograms per milliliter (Geometric Mean)
Day 1Day 11
Part 2: GSK2838232 500 mg Tablet Fed21.62027.668

[back to top]

Part 2: Time of Last Quantifiable Concentration (Tlast) Following Administration of Non-boosted Once-daily Doses of GSK2838232 Tablet in Fed State on Day 11

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as a non-boosted once-daily dosing tablet in fed state. Pharmacokinetic parameters were determined using standard non-compartmental methods. (NCT03234036)
Timeframe: Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96 and 120 hours post-dose on Day 11 in Part 2

InterventionHours (Median)
Part 2: GSK2838232 500 mg Tablet Fed118.800

[back to top]

Part 2: Lag-time (Tlag) Following Administration of Non-boosted Once-daily Doses of GSK2838232 Tablet in Fed State on Day 1

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as a non-boosted once-daily dosing tablet in fed state. Tlag is a time delay between drug administration and first observed concentration. Pharmacokinetic parameters were determined using standard non-compartmental methods. (NCT03234036)
Timeframe: Pre-dose, 0.5,1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12 and 24 hours post-dose on Day 1 in Part 2

InterventionHours (Median)
Part 2: GSK2838232 500 mg Tablet Fed0.500

[back to top]

Part 2: Pulse Rate at Indicated Time Points

Pulse rate of participants was measured in supine position after 10 minutes rest at indicated time points. (NCT03234036)
Timeframe: Day -1; Pre-dose, 1, 4 hours on Day 1; 24 hours (Day 2); 48 hours (Day 3); 72 hours (Day 4), Days 5, 6, 7, 8, 9, 10; Pre-dose, 1, 4 , 24, 48, 72 hours on Day 11; Follow-up (Day 25)

,
InterventionBeats per minute (Mean)
DAY -1DAY 1, PREDOSEDAY 1, 1 HourDAY 1, 4 HoursDAY 2, 24 HoursDAY 3, 48 HoursDAY 4, 72 HoursDAY 5DAY 6DAY 7DAY 8DAY 9DAY 10DAY 11, PREDOSEDAY 11, 1 HourDAY 11, 4 HoursDAY 11, 24 HoursDAY 11, 48 HoursDAY 11, 72 HoursFollow-Up (DAY 25)
Part 2: GSK2838232 500 mg Tablet Fed64.14363.71472.28666.00058.28659.42958.28666.42959.14363.42962.00060.71460.14360.81070.14368.85764.57161.71460.85763.857
Part 2: Placebo63.66763.66368.00064.66757.66758.00058.66760.66762.33359.33364.33361.00063.66761.88772.66768.00067.66761.66763.00070.667

[back to top]

Part 2: AUC(0-infinity) Following Administration of Non-boosted Once-daily Doses of GSK2838232 Tablet in Fed State on Day 11

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as a non-boosted once-daily dosing tablet in fed state. Pharmacokinetic parameters were determined using standard non-compartmental methods. (NCT03234036)
Timeframe: Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96 and 120 hours post-dose on Day 11 in Part 2

InterventionHours*nanograms per milliliter (Geometric Mean)
Part 2: GSK2838232 500 mg Tablet Fed1816.34

[back to top]

Part 1: Plasma Drug Concentration at 24 Hours (C24) Following Administration of GSK2838232 Tablet and Capsule Formulation in Fed State and Tablet Formulation in Fasted State

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as capsule and tablet formulation in fed state as well as tablet formulation in fasted state. Pharmacokinetic parameters were determined using standard non-compartmental methods. (NCT03234036)
Timeframe: Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96, and 120 hours post-dose in treatment periods 1, 2 and 3 of Part 1

InterventionNanograms per milliliter (Geometric Mean)
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed84.536
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed75.643
Part 1: GSK 200 mg/Ritonavir Tablet Fasted26.852

[back to top]

Part 2: Accumulation Ratio Calculated From Ctau Following Administration of GSK2838232 as Non-boosted Once-daily Doses of a Tablet Formulation

Blood sample were collected at indicated time points to calculate accumulation ratio from Ctau following administration of GSK2838232 as non-boosted once-daily doses of a tablet formulation. Accumulation ratio of GSK2838232 was evaluated by Day 11, Ctau divided by Day 1, Ctau. (NCT03234036)
Timeframe: Pre-dose, 0.5,1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12 and 24 hours post-dose on Day 1; Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96 and 120 hours post-dose on Day 11 in Part 2

InterventionRatio of Ctau (Mean)
Part 2: GSK2838232 500 mg Tablet Fed1.3205

[back to top]

Part 2: Accumulation Ratio Calculated From Cmax Following Administration of GSK2838232 as Non-boosted Once-daily Doses of a Tablet Formulation

Blood sample were collected at indicated time points to calculate accumulation ratio from Cmax following administration of GSK2838232 as non-boosted once-daily doses of a tablet formulation. Accumulation ratio of GSK2838232 was evaluated by Day 11, Cmax divided by Day 1, Cmax. (NCT03234036)
Timeframe: Pre-dose, 0.5,1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12 and 24 hours post-dose on Day 1; Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96 and 120 hours post-dose on Day 11 in Part 2

InterventionRatio of Cmax (Mean)
Part 2: GSK2838232 500 mg Tablet Fed1.2118

[back to top]

Part 2: Accumulation Ratio Calculated From AUC(0-tau) Following Administration of GSK2838232 as Non-boosted Once-daily Doses of a Tablet Formulation

Blood sample were collected at indicated time points to calculate accumulation ratio from AUC(0-tau) following administration of GSK2838232 as non-boosted once-daily doses of a tablet formulation. Accumulation ratio of GSK2838232 was evaluated by Day 11, AUC (0-tau) divided by Day 1, AUC (0-tau). (NCT03234036)
Timeframe: Pre-dose, 0.5,1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12 and 24 hours post-dose on Day 1; Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96 and 120 hours post-dose on Day 11 in Part 2

InterventionRatio of AUC (Mean)
Part 2: GSK2838232 500 mg Tablet Fed1.3340

[back to top]

Part 2: Slope of Pre-dose Concentration of GSK2838232 Administered as Non-boosted Once-daily Doses of a Tablet Formulation to Assess Achievement of Steady State

Blood sample were collected at indicated time points to assess pre-dose concentration of GSK2838232 when administered as non-boosted once-daily doses of a tablet formulation for 11 days in Part 2. Slope and 90 percent confidence interval have been presented. (NCT03234036)
Timeframe: Pre-dose on Day 3, Day 4, Day 5, Day 6, Day 7, Day 8, Day 9, Day 10 and Day 11 in Part 2

InterventionNanograms per milliliter per study day (Number)
Pre-dose, Days 3-11Pre-dose, Days 4-11Pre-dose, Days 5-11Pre-dose, Days 6-11Pre-dose, Days 7-11Pre-dose, Days 8-11Pre-dose, Days 9-11Pre-dose, Days 10-11
Part 2: GSK2838232 500 mg Tablet Fed-0.00910-0.0150-0.0379-0.0490-0.0679-0.0672-0.107-0.263

[back to top]

Part 2: Tmax Following Administration of Non-boosted Once-daily Doses of GSK2838232 Tablet in Fed State

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as a non-boosted once-daily dosing tablet in fed state. Pharmacokinetic parameters were determined using standard non-compartmental methods. (NCT03234036)
Timeframe: Pre-dose, 0.5,1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12 and 24 hours post-dose on Day 1; Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96 and 120 hours post-dose on Day 11 in Part 2

InterventionHours (Median)
Day 1Day 11
Part 2: GSK2838232 500 mg Tablet Fed4.0003.500

[back to top]

Part 1: Maximum Observed Concentration (Cmax) Following Administration of GSK2838232 Tablet and Capsule Formulation in Fed State

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as tablet and capsule formulation in fed state. Pharmacokinetic parameters were determined using standard non-compartmental methods. (NCT03234036)
Timeframe: Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96, and 120 hours post-dose in treatment periods 1 and 2 of Part 1

InterventionNanograms per milliliter (Geometric Mean)
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed109.054
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed118.118

[back to top]

Part 2: Number of Participants With Worst Case Urinalysis Results Relative to Normal Range Criteria

"Urine samples were collected from participants for analysis of following urinalysis parameters; specific gravity, potential of hydrogen (pH), presence of glucose, protein, occult blood, ketones in urine analyzed by dipstick method. The dipstick test gives results in a semi-quantitative manner. Urine specific gravity is a measure of the concentration of solutes in the urine and provides information on the kidney's ability to concentrate urine. The reference range is 1.002-1.030. Urine pH is an acid-base measurement. Normal urine has a slightly acid pH (5.0 - 6.0). Participants were counted in the worst case category that their value changes to (low, normal or high), unless there is no change in their category. Participants whose laboratory value category was unchanged (e.g., High to High), or whose value became normal, were recorded in the To Normal or No Change category." (NCT03234036)
Timeframe: Up to 25 days

,
InterventionParticipants (Count of Participants)
Glucose; To LowGlucose; To Normal or No ChangeGlucose; To HighGlucose; To AbnormalKetones; To LowKetones; To Normal or No ChangeKetones; To HighKetones; To AbnormalOccult blood; To LowOccult blood; To Normal or No ChangeOccult blood; To HighOccult blood; To AbnormalProtein; To LowProtein; To Normal or No ChangeProtein; To HighProtein; To AbnormalSpecific gravity; To LowSpecific gravity; To Normal or No ChangeSpecific gravity; To HighSpecific gravity; To AbnormalUrine pH; To LowUrine pH; To Normal or No ChangeUrine pH; To HighUrine pH; To Abnormal
Part 2: GSK2838232 500 mg Tablet Fed070007000601070007000700
Part 2: Placebo030003000201030012000300

[back to top]

Part 2: Apparent Terminal Phase Half-life (T1/2) Following Administration of Non-boosted Once-daily Doses of GSK2838232 Tablet in Fed State on Day 11

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as a non-boosted once-daily dosing tablet in fed state. Pharmacokinetic parameters were determined using standard non-compartmental methods. (NCT03234036)
Timeframe: Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96 and 120 hours post-dose on Day 11 in Part 2

InterventionHours (Geometric Mean)
Part 2: GSK2838232 500 mg Tablet Fed23.217

[back to top]

Part 1: Cmax Following Administration of GSK2838232 Tablet in Fasted and Fed State

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as tablet formulation in fed and fasted state. Pharmacokinetic parameters were determined using standard non-compartmental methods. (NCT03234036)
Timeframe: Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96, and 120 hours post-dose in treatment periods 1, 2 and 3 of Part 1

InterventionNanograms per milliliter (Geometric Mean)
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed118.118
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fasted50.437

[back to top]

Part 1: AUC(0-t) Following Administration of GSK2838232 Tablet and Capsule Formulation in Fed State and Tablet Formulation in Fasted State

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as capsule and tablet formulation in fed state as well as tablet formulation in fasted state. Pharmacokinetic parameters were determined using standard non-compartmental methods. (NCT03234036)
Timeframe: Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96, and 120 hours post-dose in treatment periods 1, 2 and 3 of Part 1

InterventionHours* nanograms per milliliter (Geometric Mean)
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed4595.35
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed4371.47
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fasted1776.21

[back to top]

Part 2: Number of Participants With Abnormal Electrocardiogram (ECG) Findings

Single and triplicate 12-lead ECG's were obtained at least 5 minutes apart in the supine position after 10 minutes of rest at indicated time points using an ECG machine that automatically calculates the heart rate and measures PR, QRS, QT and QT duration corrected for heart rate by Fridericia's formula (QTcF) intervals. Clinically significant (CS) and not clinically significant (NCS) abnormal ECG findings have been presented. CS abnormal findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. (NCT03234036)
Timeframe: Day -1; 1, 4, 12 hours on Day 1; Days 2, 3, 5, 8; 1, 4, 12, 24 Hours on Day 11; Follow-up (Day 25)

,
InterventionParticipants (Count of Participants)
NCS: DAY -1CS: DAY -1NCS: DAY 1, 1 HourCS: DAY 1, 1 HourNCS: DAY 1, 4 HoursCS: DAY 1, 4 HoursNCS: DAY 1, 12 HoursCS: DAY 1, 12 HoursNCS: DAY 2CS: DAY 2NCS: DAY 3CS: DAY 3NCS: DAY 5CS: DAY 5NCS: DAY 8CS: DAY 8NCS: DAY 11, 1 HourCS: DAY 11, 1 HourNCS: DAY 11, 4 HoursCS: DAY 11, 4 HoursNCS: DAY 11, 12 HoursCS: DAY 11, 12 HoursNCS: DAY 11, 24 HoursCS: DAY 11, 24 HoursNCS: Follow-up (DAY 25)CS: Follow-up (DAY 25)
Part 2: GSK2838232 500 mg Tablet Fed60406060505060703050405060
Part 2: Placebo20202030203030202020202010

[back to top]

Part 1: T1/2 Following Administration of GSK2838232 Tablet and Capsule Formulation in Fed State and Tablet Formulation in Fasted State

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as capsule and tablet formulation in fed state as well as tablet formulation in fasted state. Pharmacokinetic parameters were determined using standard non-compartmental methods. (NCT03234036)
Timeframe: Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96, and 120 hours post-dose in treatment periods 1, 2 and 3 of Part 1

InterventionHours (Geometric Mean)
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed15.606
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed15.692
Part 1: GSK 200 mg/Ritonavir Tablet Fasted17.061

[back to top]

Part 1: Time of Occurrence of Cmax (Tmax) Following Administration of GSK2838232 Tablet in Fasted and Fed State

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as tablet formulation in fed and fasted state. Pharmacokinetic parameters were determined using standard non-compartmental methods. (NCT03234036)
Timeframe: Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96, and 120 hours post-dose in treatment periods 1, 2 and 3 of Part 1

InterventionHours (Median)
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed5.983
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fasted3.508

[back to top]

Part 1: Tlag Following Administration of GSK2838232 Tablet and Capsule Formulation in Fed State and Tablet Formulation in Fasted State

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as capsule and tablet formulation in fed state as well as tablet formulation in fasted state. Tlag is a time delay between drug administration and first observed concentration. Pharmacokinetic parameters were determined using standard non-compartmental methods. (NCT03234036)
Timeframe: Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96, and 120 hours post-dose in treatment periods 1, 2 and 3 of Part 1

InterventionHours (Median)
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed1.000
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed1.000
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fasted0.500

[back to top]

Part 1: Tlast Following Administration of GSK2838232 Tablet and Capsule Formulation in Fed State and Tablet Formulation in Fasted State

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as capsule and tablet formulation in fed state as well as tablet formulation in fasted state. Pharmacokinetic parameters were determined using standard non-compartmental methods. (NCT03234036)
Timeframe: Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96, and 120 hours post-dose in treatment periods 1, 2 and 3 of Part 1

InterventionHours (Median)
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed119.983
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed119.983
Part 1: GSK 200 mg/Ritonavir Tablet Fasted120.017

[back to top]

Part 1: Tmax Following Administration of GSK2838232 Tablet and Capsule Formulation in Fed State and Tablet Formulation in Fasted State

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as capsule and tablet formulation in fed state as well as tablet formulation in fasted state. Pharmacokinetic parameters were determined using standard non-compartmental methods. (NCT03234036)
Timeframe: Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96, and 120 hours post-dose in treatment periods 1, 2 and 3 of Part 1

InterventionHours (Median)
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed11.967
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed5.983
Part 1: GSK 200 mg/Ritonavir Tablet Fasted3.508

[back to top]

Part 1: Change From Baseline in Mean Heart Rate Values as ECG Parameter

Single and triplicate 12-lead ECG's were obtained at least 5 minutes apart in the supine position after 10 minutes of rest at indicated time points using an ECG machine that automatically calculates the heart rate. Baseline was defined as the latest pre-dose assessment, including those from unscheduled visits. Change from Baseline was defined as any visit value minus Baseline value. (NCT03234036)
Timeframe: Baseline (Day -1) and 1, 2, 4, 6, 8, 12, 24, 48, 72 hours on Day 1

,,
InterventionBeats per minute (Mean)
DAY 1, 1 Hour; n=15,15,14DAY 1, 2 Hours; n=15,15,13DAY 1, 4 Hours; n=15,14,14DAY 1, 6 Hours; n=14,14,14DAY 1, 8 Hours; n=15,15,14DAY 1, 12 Hours; n=15,15,14DAY 1, 24 Hours; n=15,15,14DAY 1, 48 Hours; n=15,15,14DAY 1, 72 Hours; n=15,15,14
Part 1: GSK 200 mg/Ritonavir Tablet Fasted-0.642-1.2302.1445.7862.9296.0722.7862.3582.286
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed11.62310.6234.1566.0722.2234.089-2.3771.023-0.911
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed7.9558.4892.5001.6431.489-0.178-4.711-2.378-3.045

[back to top]

Part 1: Change From Baseline in PR Interval, QRS, QT Interval, and QTcF Interval as ECG Parameters

Single and triplicate 12-lead ECG's were obtained at least 5 minutes apart in the supine position after 10 minutes of rest at indicated time points using an ECG machine that automatically calculates measures PR, QRS, QT and QTcF intervals. Baseline was defined as the latest pre-dose assessment, including those from unscheduled visits. Change from Baseline was defined as any visit value minus Baseline value. (NCT03234036)
Timeframe: Baseline (Day -1) and 1, 2, 4, 6, 8, 12, 24, 48, 72 hours on Day 1

,,
InterventionMilliseconds (Mean)
PR interval; DAY 1, 1 Hour; n= 15,15,14PR interval; DAY 1, 2 Hours; n=15,15,13PR interval; DAY 1, 4 Hours; n=15,14,14PR interval; DAY 1, 6 Hours; n=14,14,14PR interval; DAY 1, 8 Hours; n=15,15,14PR interval; DAY 1, 12 Hours; n=15,15,14PR interval; DAY 1, 24 Hours; n=15,15,14PR interval; DAY 1, 48 Hours; n=15,15,14PR interval; DAY 1, 72 Hours; n=15,15,14QRS interval; DAY 1, 1 Hour; n=15,15,14QRS interval; DAY 1, 2 Hours; n=15,15,13QRS interval; DAY 1, 4 Hours; n=15,14,14QRS interval; DAY 1, 6 Hours; n=14,14,14QRS interval; DAY 1, 8 Hours; n=15,15,14QRS interval; DAY 1, 12 Hours; n=15,15,14QRS interval; DAY 1, 24 Hours; n=15,15,14QRS interval; DAY 1, 48 Hours; n=15,15,14QRS interval; DAY 1, 72 Hours; n=15,15,14QT interval; DAY 1, 1 Hour; n=15,15,14QT interval; DAY 1, 2 Hours; n=15,15,13QT interval; DAY 1, 4 Hours; n=15,14,14QT interval; DAY 1, 6 Hours; n=14,14,14QT interval; DAY 1, 8 Hours; n=15,15,14QT interval; DAY 1, 12 Hours; n=15,15,14QT interval; DAY 1, 24 Hours; n=15,15,14QT interval; DAY 1, 48 Hours; n=15,15,14QT interval; DAY 1, 72 Hours; n=15,15,14QTcF interval; DAY 1, 1 Hour; n=15,15,14QTcF interval; DAY 1, 2 Hours; n=15,15,13QTcF interval; DAY 1, 4 Hours; n=15,14,14QTcF interval; DAY 1, 6 Hours; n=14,14,14QTcF interval; DAY 1, 8 Hours; n=15,15,14QTcF interval; DAY 1, 12 Hours; n=15,15,14QTcF interval; DAY 1, 24 Hours; n=15,15,14QTcF interval; DAY 1, 48 Hours; n=15,15,14QTcF interval; DAY 1, 72 Hours; n=15,15,14
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed-4.667-6.001-7.067-5.286-2.2673.5990.1330.5330.2660.177-1.289-0.2231.190-1.0230.577-0.489-0.4890.311-27.511-30.844-17.377-15.524-9.511-16.044-2.044-3.644-0.977-4.666-10.133-8.466-3.714-4.333-7.199-6.933-0.999-2.466
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fasted-3.000-1.846-2.571-7.286-7.571-5.143-4.000-2.8570.286-1.191-1.949-1.334-0.048-1.476-0.905-1.762-2.905-1.905-0.3341.435-2.191-23.334-12.762-24.334-11.476-7.476-12.762-2.285-2.0761.858-10.499-6.356-11.214-5.856-3.785-7.785
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed-3.067-8.000-5.905-4.0480.2673.6000.6670.267-0.800-1.067-3.601-3.144-1.096-1.2010.266-0.001-3.601-1.067-23.333-26.266-15.523-10.619-6.266-9.4660.801-0.6660.134-8.868-11.068-11.335-7.573-3.268-9.735-9.601-5.601-6.401

[back to top]

Part 1: Change From Baseline in Pulse Rate

Pulse rate was measured in supine position after 10 minutes rest for the participant at indicated time points. Baseline was defined as the latest pre-dose assessment, including those from unscheduled visits. Change from Baseline was defined as any visit value minus Baseline value. (NCT03234036)
Timeframe: Baseline (Day -1) and 1, 2, 4, 6, 8, 12, 24, 48, 72 Hours on Day 1

,,
InterventionBeats per minute (Mean)
DAY 1, 1 HourDAY 1, 2 HoursDAY 1, 4 HoursDAY 1, 6 HoursDAY 1, 8 HoursDAY 1, 12 HoursDAY 1, 24 HoursDAY 1, 48 HoursDAY 1, 72 Hours
Part 1: GSK 200 mg/Ritonavir Tablet Fasted-1.2860.3571.3576.6433.7147.2864.2143.4292.429
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed9.6447.6443.1113.8441.511-0.156-1.623-0.689-2.756
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed8.0238.6235.1563.8232.6231.556-3.044-0.777-1.844

[back to top]

Part 1: Number of Participants With Abnormal ECG Findings

Single and triplicate 12-lead ECG's were obtained at least 5 minutes apart in the supine position after 10 minutes of rest at indicated time points using an ECG machine that automatically calculates the heart rate and measures PR, QRS, QT and QTcF intervals. CS and NCS abnormal ECG findings have been presented. CS abnormal findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. (NCT03234036)
Timeframe: Day -2, Day -1; 1, 2, 4, 6, 8, , 12, 24, 48, 72 hours on Day 1

,,
InterventionParticipants (Count of Participants)
NCS: DAY -2; n=15, 15, 14CS: DAY -2; n=15, 15, 14NCS: DAY -1; n=15, 14, 14CS: DAY -1; n=15, 14, 14NCS: DAY 1, 1 Hour; n=15, 15, 14CS: DAY 1, 1 Hour; n=15, 15, 14NCS: DAY 1, 2 Hours; n=15, 15, 13CS: DAY 1, 2 Hours; n=15, 15, 13NCS: DAY 1, 4 Hours; n=15, 13, 14CS: DAY 1, 4 Hours; n=15, 13, 14NCS: DAY 1, 6 Hours; n=14, 14, 14CS: DAY 1, 6 Hours; n=14, 14, 14NCS: DAY 1, 8 Hours; n=15, 15, 14CS: DAY 1, 8 Hours; n=15, 15, 14NCS: DAY 1, 12 Hours; n=15, 14, 14CS: DAY 1, 12 Hours; n=15, 14, 14NCS: DAY 1, 24 Hours; n=14, 15, 14CS: DAY 1, 24 Hours; n=14, 15, 14NCS: DAY 1, 48 Hours; n=15, 15, 14CS: DAY 1, 48 Hours; n=15, 15, 14NCS: DAY 1, 72 Hours; n=15, 15, 14CS: DAY 1, 72 Hours; n=15, 15, 14
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed1007060505030707011080100
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fasted90120110100130508090100110110
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed90905050303050401009080

[back to top]

Part 1: Number of Participants With SAEs and Non-SAEs

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study treatment, whether or not considered related to the study treatment. SAE is defined as any untoward medical occurrence that, at any dose results in death, is life threatening, requires hospitalization or prolongation of existing hospitalization, results in persistent disability/incapacity, is a congenital anomaly/ birth defect, is associated with liver injury or impaired liver function or any other situations as per medical or scientific judgment. Number of participants with SAEs and common non-SAEs (>=5%) are presented. (NCT03234036)
Timeframe: Up to 60 days

,,
InterventionParticipants (Count of Participants)
Any SAEAny non-SAE
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed03
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fasted01
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed02

[back to top]

Part 1: Number of Participants With Worst Case Clinical Chemistry Results to PCI Criteria

"Blood samples were collected from participants for analysis of following clinical chemistry parameters; glucose, ALT, albumin, alkaline phosphatase, AST, bilirubin, calcium, potassium and sodium. PCI ranges were <30 g/L for albumin, <2 or 2.75 mmol/L for calcium, <3 or >9 mmol/L for glucose, >=2 times ULN U/L for ALT, >=2 times ULN U/L for alkaline phosphatase, >=2 times ULN U/L for AST, >=1.5 times ULN µmol/L for bilirubin, <3 or >5.5 mmol/L for potassium, and <130 or >150 mmol/L for sodium. Participants were counted in the worst case category that their value changes to (low, within range or no change,or high), unless there is no change in their category. Participants whose laboratory value category was unchanged (e.g., High to High), or whose value became within range, were recorded in the To within Range or No Change category." (NCT03234036)
Timeframe: Up to 60 days

,,
InterventionParticipants (Count of Participants)
Glucose; To LowGlucose; To within Range or No ChangeGlucose; To HighALT; To LowALT; To within Range or No ChangeALT; To HighAlbumin; To LowAlbumin; To within Range or No ChangeAlbumin; To HighAlkaline phosphatase; To LowAlkaline phosphatase; To within Range or No ChangeAlkaline phosphatase; To HighAST; To LowAST; To within Range or No ChangeAST; To HighBilirubin; To LowBilirubin; To within Range or No ChangeBilirubin; To HighCalcium; To LowCalcium; To within Range or No ChangeCalcium; To HighPotassium; To LowPotassium; To within Range or No ChangePotassium; To HighSodium; To LowSodium; To within Range or No ChangeSodium; To High
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed015001500150015001500150015001500150
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fasted014001400140014001400140014001400140
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed015001500150015001500150015001500150

[back to top]

Part 1: Number of Participants With Worst Case Hematology Results to PCI Criteria

"Blood samples were collected from participants for analysis of following hematology parameters; hematocrit, hemoglobin, leukocytes, lymphocytes, neutrophils and platelets. PCI ranges were >0.54 as proportion of red blood cells in blood for hematocrit, >180 g/L for hemoglobin, < 3 or >20 cells/L for leukocytes, 0.8 x10^9 cells/L for lymphocytes, 1.5 x10^9 cells/L for neutrophils, and <100 or >550 cells/L for platelets. Participants were counted in the worst case category that their value changes to (low, within range or no change, or high), unless there is no change in their category. Participants whose laboratory value category was unchanged (e.g., High to High), or whose value became within range, were recorded in the To within Range or No Change category." (NCT03234036)
Timeframe: Up to 60 days

,,
InterventionParticipants (Count of Participants)
Hematocrit; To LowHematocrit; To within Range or No ChangeHematocrit; To HighHemoglobin; To LowHemoglobin; To within Range or No ChangeHemoglobin; To HighLeukocytes; To LowLeukocytes; To within Range or No ChangeLeukocytes; To HighLymphocytes; To LowLymphocytes; To within Range or No ChangeLymphocytes; To HighNeutrophils; To LowNeutrophils; To within Range or No ChangeNeutrophils; To HighPlatelets; To LowPlatelets; To within Range or No ChangePlatelets; To High
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed015001500150015001500150
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fasted014001400140014001400140
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed015001500150015001500150

[back to top]

Part 1: Number of Participants With Worst Case Urinalysis Results Relative to Normal Range Criteria

"Urine samples were collected from participants for analysis of following urinalysis parameters; specific gravity, pH, presence of glucose, protein, occult blood, ketones in urine analyzed by dipstick method. The dipstick test gives results in a semi-quantitative manner. Urine specific gravity is a measure of the concentration of solutes in the urine and provides information on the kidney's ability to concentrate urine. The reference range is 1.002-1.030. Urine pH is an acid-base measurement. Normal urine has a slightly acid pH (5.0 - 6.0). Participants were counted in the worst case category that their value changes to (low, normal or high), unless there is no change in their category. Participants whose laboratory value category was unchanged (e.g., High to High), or whose value became normal, were recorded in the To Normal or No Change category." (NCT03234036)
Timeframe: Up to 60 days

,,
InterventionParticipants (Count of Participants)
Glucose; To LowGlucose; To Normal or No ChangeGlucose; To HighGlucose; To AbnormalKetones; To LowKetones; To Normal or No ChangeKetones; To HighKetones To AbnormalOccult blood; To LowOccult blood ; To Normal or No ChangeOccult blood; To HighOccult blood To AbnormalProtein; To LowProtein; To Normal or No ChangeProtein; To HighProtein To AbnormalSpecific gravity; To LowSpecific gravity; To Normal or No ChangeSpecific gravity; To HighSpecific gravity To AbnormalUrine pH; To LowUrine pH; To Normal or No ChangeUrine pH; To HighUrine pH; To Abnormal
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed015000140101401015001140001500
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fasted014000140001004014002120001400
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed015000150001302015000150001500

[back to top]

Part 1: Pulse Rate at Indicated Time Points

Pulse rate was measured in supine position after 10 minutes rest for the participant at indicated time points. (NCT03234036)
Timeframe: Day -2; Day -1; Pre-dose, 1, 2, 4, 6, 8, 12, 24, 48, 72 Hours on Day 1

,,
InterventionBeats per minute (Mean)
DAY -2DAY -1DAY 1, PREDOSEDAY 1, 1 HourDAY 1, 2 HoursDAY 1, 4 HoursDAY 1, 6 HoursDAY 1, 8 HoursDAY 1, 12 HoursDAY 1, 24 HoursDAY 1, 48 HoursDAY 1, 72 Hours
Part 1: GSK 200 mg/Ritonavir Tablet Fasted60.21459.07159.85758.57160.21461.21466.50063.57167.14364.07163.28662.286
Part 1: GSK2838232 200 mg/Ritonavir Capsule Fed64.06762.33365.02374.66772.66768.13368.86766.53364.86763.40064.33362.267
Part 1: GSK2838232 200 mg/Ritonavir Tablet Fed62.33363.06764.31172.33372.93369.46768.13366.93365.86761.26763.53362.467

[back to top]

Part 2: Area Under the Plasma Drug Concentration Time Curve From Pre-dose to the End of the Dosing Interval at Steady State (AUC[0-tau]) Following Administration of Non-boosted Once-daily Doses of GSK2838232 Tablet in Fed State

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as a non-boosted once-daily dosing tablet in fed state. Pharmacokinetic parameters were determined using standard non-compartmental methods. (NCT03234036)
Timeframe: Pre-dose, 0.5,1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12 and 24 hours post-dose on Day 1; Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96 and 120 hours post-dose on Day 11 in Part 2

InterventionHours*nanograms per milliliter (Geometric Mean)
Day 1Day 11
Part 2: GSK2838232 500 mg Tablet Fed895.421184.55

[back to top]

Part 2: Blood Pressure at Indicated Time Points

Systolic blood pressure (SBP) and diastolic blood pressure (DBP) were measured in supine position after 10 minutes rest for the participants at indicated time points. (NCT03234036)
Timeframe: Day -1; Pre-dose, 1, 4 hours on Day 1; 24 hours (Day 2); 48 hours (Day 3); 72 hours (Day 4), Days 5, 6, 7, 8, 9, 10; Pre-dose, 1, 4 , 24, 48, 72 hours on Day 11; Follow-up (Day 25)

,
InterventionMillimeter of mercury (Mean)
DBP; DAY -1DBP; DAY 1, PREDOSEDBP; DAY 1, 1 HourDBP; DAY 1, 4 HoursDBP; DAY 2, 24 HoursDBP; DAY 3, 48 HoursDBP; DAY 4, 72 HoursDBP; DAY 5DBP; DAY 6DBP; DAY 7DBP; DAY 8DBP; DAY 9DBP; DAY 10DBP; DAY 11, PREDOSEDBP; DAY 11, 1 HourDBP; DAY 11, 4 HoursDBP; DAY 11, 24 HoursDBP; DAY 11, 48 HoursDBP; DAY 11, 72 HoursDBP; Follow-Up (DAY 25)SBP; DAY -1SBP; DAY 1, PREDOSESBP; DAY 1, 1 HourSBP; DAY 1, 4 HoursSBP; DAY 2, 24 HoursSBP; DAY 3, 48 HoursSBP; DAY 4, 72 HoursSBP; DAY 5SBP; DAY 6SBP; DAY 7SBP; DAY 8SBP; DAY 9SBP; DAY 10SBP; DAY 11, PREDOSESBP; DAY 11, 1 HourSBP; DAY 11, 4 HoursSBP; DAY 11, 24 HoursSBP; DAY 11, 48 HoursSBP; DAY 11, 72 HoursSBP; Follow-Up (DAY 25)
Part 2: GSK2838232 500 mg Tablet Fed73.14372.33364.71463.71473.71468.00065.14365.00066.14367.28669.28666.71466.57167.00066.57165.57164.71468.00065.00076.143123.000121.190113.000106.286113.286111.714109.000109.000107.857112.143109.571110.143106.714110.333111.571109.857105.143109.286110.286123.714
Part 2: Placebo73.66765.44354.66762.00073.66766.00069.00062.33362.66764.33366.00069.00065.00065.33362.66763.33367.66766.00066.33371.000121.000107.55399.333105.000110.667111.333108.333105.667106.667103.333105.667107.333110.000107.113113.000108.667107.333109.667110.333115.667

[back to top]

Part 2: Change From Baseline in Blood Pressure

SBP and DBP were measured in supine position after 10 minutes rest for participants at indicated time points. Baseline was defined as the latest pre-dose assessment, including those from unscheduled visits. Change from Baseline was defined as any visit value minus Baseline value. (NCT03234036)
Timeframe: Baseline (Day -1) and 1, 4 hours on Day 1; 24 hours (Day 2); 48 hours (Day 3); 72 hours (Day 4), Days 5, 6, 7, 8, 9, 10; Pre-dose, 1, 4 , 24, 48, 72 hours on Day 11; Follow-up (Day 25)

,
InterventionMillimeter of mercury (Mean)
DBP; DAY 1, 1 HourDBP; DAY 1, 4 HoursDBP; DAY 2, 24 HoursDBP; DAY 3, 48 HoursDBP; DAY 4, 72 HoursDBP; DAY 5DBP; DAY 6DBP; DAY 7DBP; DAY 8DBP; DAY 9DBP; DAY 10DBP; DAY 11, PREDOSEDBP; DAY 11, 1 HourDBP; DAY 11, 4 HoursDBP; DAY 11, 24 HoursDBP; DAY 11, 48 HoursDBP; DAY 11, 72 HoursDBP; Follow-Up (DAY 25)SBP; DAY 1, 1 HourSBP; DAY 1, 4 HoursSBP; DAY 2, 24 HoursSBP; DAY 3, 48 HoursSBP; DAY 4, 72 HoursSBP; DAY 5SBP; DAY 6SBP; DAY 7SBP; DAY 8SBP; DAY 9SBP; DAY 10SBP; DAY 11, PREDOSESBP; DAY 11, 1 HourSBP; DAY 11, 4 HoursSBP; DAY 11, 24 HoursSBP; DAY 11, 48 HoursSBP; DAY 11, 72 HoursSBP; Follow-Up (DAY 25)
Part 2: GSK2838232 500 mg Tablet Fed-7.619-8.6191.381-4.333-7.190-7.333-6.190-5.047-3.047-5.619-5.761-5.333-5.761-6.761-7.619-4.333-7.3333.810-8.190-14.904-7.904-9.476-12.190-12.190-13.333-9.047-11.619-11.047-14.476-10.857-9.619-11.333-16.047-11.904-10.9042.524
Part 2: Placebo-10.777-3.4438.2230.5573.557-3.110-2.777-1.1100.5573.557-0.443-0.110-2.777-2.1102.2230.5570.8905.557-8.220-2.5533.1133.7800.780-1.887-0.887-4.220-1.887-0.2202.447-0.4405.4471.113-0.2202.1132.7808.113

[back to top]

Part 2: Change From Baseline in Mean Heart Rate Values as ECG Parameter

Single and triplicate 12-lead ECG's were obtained at least 5 minutes apart in the supine position after 10 minutes of rest at indicated time points using an ECG machine that automatically calculates the heart rate. Baseline was defined as the latest pre-dose assessment, including those from unscheduled visits. Change from Baseline was defined as any visit value minus Baseline value. (NCT03234036)
Timeframe: Baseline (Day -1) and 1, 4, 12 hours on Day 1; Days 2, 3 5, 8; Pre-dose, 1, 4, 12, 24 hours Day 11; Follow-up (Day 25)

,
InterventionBeats per minute (Mean)
DAY 1, 1 HourDAY 1, 4 HoursDAY 1, 12 HoursDAY 2DAY 3DAY 5DAY 8DAY 11, PREDOSEDAY 11, 1 HourDAY 11, 4 HoursDAY 11, 12 HoursDAY 11, 24 HoursFollow-up (DAY 25)
Part 2: GSK2838232 500 mg Tablet Fed10.2401.9540.526-5.331-1.0462.6690.954-0.9036.3834.811-0.1890.9540.669
Part 2: Placebo4.0002.000-6.000-8.000-5.333-3.667-2.000-4.33310.3331.000-0.6672.6672.333

[back to top]

Part 2: Change From Baseline in PR Interval, QRS, QT Interval, and QTcF Interval as ECG Parameters

Single and triplicate 12-lead ECG's were obtained at least 5 minutes apart in the supine position after 10 minutes of rest at indicated time points using an ECG machine that automatically calculates measures PR, QRS, QT and QTcF intervals. Baseline was defined as the latest pre-dose assessment, including those from unscheduled visits. Change from Baseline was defined as any visit value minus Baseline value. (NCT03234036)
Timeframe: Baseline (Day -1) and 1, 4, 12 hours on Day 1; Days 2, 3 5, 8; Pre-dose, 1, 4, 12, 24 hours Day 11; Follow-up (Day 25)

,
InterventionMilliseconds (Mean)
PR interval; DAY 1, 1 HourPR interval; DAY 1, 4 HoursPR interval; DAY 1, 12 HoursPR interval; DAY 2PR interval; DAY 3PR interval; DAY 5PR interval; DAY 8PR interval; DAY 11, PREDOSEPR interval; DAY 11, 1 HourPR interval; DAY 11, 4 HoursPR interval; DAY 11, 12 HoursPR interval; DAY 11, 24 HoursPR interval; Follow-up (DAY 25)QRS interval; DAY 1, 1 HourQRS interval; DAY 1, 4 HoursQRS interval; DAY 1, 12 HoursQRS interval; DAY 2QRS interval; DAY 3QRS interval; DAY 5QRS interval; DAY 8QRS interval; DAY 11, PREDOSEQRS interval; DAY 11, 1 HourQRS interval; DAY 11, 4 HoursQRS interval; DAY 11, 12 HoursQRS interval; DAY 11, 24 HoursQRS interval; Follow-up (DAY 25)QT interval; DAY 1, 1 HourQT interval; DAY 1, 4 HoursQT interval; DAY 1, 12 HoursQT interval; DAY 2QT interval; DAY 3QT interval; DAY 5QT interval; DAY 8QT interval; DAY 11, PREDOSEQT interval; DAY 11, 1 HourQT interval; DAY 11, 4 HoursQT interval; DAY 11, 12 HoursQT interval; DAY 11, 24 HoursQT interval; Follow-up (DAY 25)QTcF interval; DAY 1, 1 HourQTcF interval; DAY 1, 4 HoursQTcF interval; DAY 1, 12 HoursQTcF interval; DAY 2QTcF interval; DAY 3QTcF interval; DAY 5QTcF interval; DAY 8QTcF interval; DAY 11, PREDOSEQTcF interval; DAY 11, 1 HourQTcF interval; DAY 11, 4 HoursQTcF interval; DAY 11, 12 HoursQTcF interval; DAY 11, 24 HoursQTcF interval; Follow-up (DAY 25)
Part 2: GSK2838232 500 mg Tablet Fed-3.999-12.2843.4303.7164.5732.2877.4307.6202.287-0.5701.7163.1442.001-1.713-1.7131.7160.287-4.570-1.999-3.141-0.570-0.856-2.570-0.856-0.5701.716-26.094-11.523-7.2370.763-8.951-10.666-8.951-4.283-28.380-20.380-13.237-13.523-3.237-5.857-7.429-6.286-10.143-10.714-6.286-6.714-6.476-15.286-11.000-12.571-10.429-2.571
Part 2: Placebo-7.110-6.443-0.4434.8901.5576.2236.2235.113-5.777-3.1104.2232.890-10.4433.3301.9976.6631.3302.6633.997-0.0031.107-0.003-1.3373.330-1.3371.997-8.223-3.55718.4435.7775.777-0.8901.1105.553-18.223-8.2235.777-10.8900.443-0.6671.0007.000-10.667-5.000-7.667-2.000-2.8871.000-5.6675.333-5.6674.000

[back to top]

Part 2: Change From Baseline in Pulse Rate

Pulse rate was measured in supine position after 10 minutes rest at indicated time points. Baseline was defined as the latest pre-dose assessment, including those from unscheduled visits. Change from Baseline was defined as any visit value minus Baseline value. (NCT03234036)
Timeframe: Baseline (Day -1) and 1, 4 hours on Day 1; 24 hours (Day 2); 48 hours (Day 3); 72 hours (Day 4), Days 5, 6, 7, 8, 9, 10; Pre-dose, 1, 4 , 24, 48, 72 hours on Day 11; Follow-up (Day 25)

,
InterventionBeats per minute (Mean)
DAY 1, 1 HourDAY 1, 4 HoursDAY 2, 24 HoursDAY 3, 48 HoursDAY 4, 72 HoursDAY 5DAY 6DAY 7DAY 8DAY 9DAY 10DAY 11, PREDOSEDAY 11, 1 HourDAY 11, 4 HoursDAY 11, 24 HoursDAY 11, 48 HoursDAY 11, 72 HoursFollow-Up (DAY 25)
Part 2: GSK2838232 500 mg Tablet Fed8.5712.286-5.429-4.286-5.4292.714-4.571-0.286-1.714-3.000-3.571-2.9046.4295.1430.857-2.000-2.8570.143
Part 2: Placebo4.3371.003-5.997-5.663-4.997-2.997-1.330-4.3300.670-2.6630.003-1.7779.0034.3374.003-1.997-0.6637.003

[back to top]

Part 2: Cmax Following Administration of Non-boosted Once-daily Doses of GSK2838232 Tablet in Fed State

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232 given as a non-boosted once-daily dosing tablet in fed state. Pharmacokinetic parameters were determined using standard non-compartmental methods. (NCT03234036)
Timeframe: Pre-dose, 0.5,1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12 and 24 hours post-dose on Day 1; Pre-dose, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6, 8, 12, 24, 36, 48, 72, 96 and 120 hours post-dose on Day 11 in Part 2

InterventionNanograms per milliliter (Geometric Mean)
Day 1Day 11
Part 2: GSK2838232 500 mg Tablet Fed79.57594.239

[back to top]

Part 2: Number of Participants With Serious Adverse Events (SAEs) and Non-SAEs

An adverse event (AE) is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study treatment, whether or not considered related to the study treatment. SAE is defined as any untoward medical occurrence that, at any dose results in death, is life threatening, requires hospitalization or prolongation of existing hospitalization, results in persistent disability/incapacity, is a congenital anomaly/ birth defect, is associated with liver injury or impaired liver function or any other situations as per medical or scientific judgment. Safety Population comprised of all participants who received at least 1 dose of the study treatment (including placebo) with at least 1 post-Baseline safety assessment. Number of participants with SAEs and common non-SAEs (>=5%) are presented. (NCT03234036)
Timeframe: Up to 25 days

,
InterventionParticipants (Count of Participants)
Any SAEAny non-SAE
Part 2: GSK2838232 500 mg Tablet Fed00
Part 2: Placebo01

[back to top]

Part 2: Number of Participants With Worst Case Clinical Chemistry Results to PCI Criteria

"Blood samples were collected from participants for analysis of following clinical chemistry parameters; glucose, alanine aminotransferase (ALT), albumin, alkaline phosphatase, aspartate aminotransferase (AST), bilirubin, calcium, potassium and sodium. PCI ranges were <30 g/L for albumin, <2 or >2.75 millimoles per liter (mmol/L) for calcium, <3 or >9 mmol/L for glucose, >=2 times Upper limit of Normal (ULN) units per liter (U/L) for ALT, >=2 times ULN U/L for alkaline phosphatase, >=2 times ULN U/L for AST, >=1.5 times ULN micromoles per liter (µmol/L) for bilirubin, <3 or >5.5 mmol/L for potassium, and <130 or >150 mmol/L for sodium. Participants were counted in the worst case category that their value changes to (low, within range or no change,or high), unless there is no change in their category. Participants whose laboratory value category was unchanged (e.g., High to High), or whose value became within range, were recorded in the To within Range or No Change category." (NCT03234036)
Timeframe: Up to 25 days

,
InterventionParticipants (Count of Participants)
Glucose; To LowGlucose; To within Range or No ChangeGlucose; To HighALT; To LowALT; To within Range or No ChangeALT; To HighAlbumin; To LowAlbumin; To within Range or No ChangeAlbumin; To HighAlkaline phosphatase; To LowAlkaline phosphatase; To within Range or No ChangeAlkaline phosphatase; To HighAST; To LowAST; To within Range or No ChangeAST; To HighBilirubin; To LowBilirubin; To within Range or No ChangeBilirubin; To HighCalcium; To LowCalcium; To within Range or No ChangeCalcium; To HighPotassium; To LowPotassium; To within Range or No ChangePotassium; To HighSodium; To LowSodium; To within Range or No ChangeSodium; To High
Part 2: GSK2838232 500 mg Tablet Fed070070070070070070070070070
Part 2: Placebo030030030030030030030030030

[back to top]

Part 2: Number of Participants With Worst Case Hematology Results to Potential Clinical Importance (PCI) Criteria

"Blood samples were collected from participants for analysis of following hematology parameters; hematocrit, hemoglobin, leukocytes, lymphocytes, neutrophils and platelets. PCI ranges were < 0.075 or >0.54 proportion of red blood cells in blood for hematocrit, <25 or >180 grams per liter (g/L) for hemoglobin, < 3 or >20 cells per liter (cells/L) for leukocytes, 0.8 x10^9 cells/L for lymphocytes, 1.5 x10^9 cells/L for neutrophils, and <100 or >550 cells/L for platelets. Participants were counted in the worst case category that their value changes to (low, within range or no change, or high), unless there is no change in their category. Participants whose laboratory value category was unchanged (example given [e.g.], High to High), or whose value became within range, were recorded in the To within Range or No Change category." (NCT03234036)
Timeframe: Up to 25 days

,
InterventionParticipants (Count of Participants)
Hematocrit; To LowHematocrit; To within Range or No ChangeHematocrit; To HighHemoglobin; To LowHemoglobin; To within Range or No ChangeHemoglobin; To HighLeukocytes; To LowLeukocytes; To within Range or No ChangeLeukocytes; To HighLymphocytes; To LowLymphocytes; To within Range or No ChangeLymphocytes; To HighNeutrophils; To LowNeutrophils; To within Range or No ChangeNeutrophils; To HighPlatelets; To LowPlatelets; To within Range or No ChangePlatelets; To High
Part 2: GSK2838232 500 mg Tablet Fed070070070070160070
Part 2: Placebo030030030030120030

[back to top]

Best Objective Response as Confirmed By The Investigator's Assessment in Participants With HER2-Expressing Advanced Solid Malignant Tumors

Best objective response (BOR) was assessed by the investigator based on Response Evaluation Criteria In Solid Tumors (RECIST) version 1.1. Completed response (CR) was defined as a disappearance of all target lesions, PR was defined as at least a 30% decrease in the sum of diameters of target lesions, and stable disease (SD) was defined as neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for progressive disease (PD; at least a 20% increase in the sum of diameters of target lesions. (NCT03383692)
Timeframe: Baseline up to disease progression, death, lost to follow up, or study discontinuation (whichever comes first), up to approximately 9 months post-dose

,
InterventionParticipants (Count of Participants)
Complete response (CR)Partial response (PR)Stable disease (SD)Non-CR/Non-PRProgressive disease (PD)Non evaluable
Cohort 1: DS-8201a + Ritonavir098000
Cohort 2: DS-8201a + Itraconazole0108010

[back to top]

Pharmacokinetic Parameter of Area Under the Serum Concentration-time Curve Following Treatment With DS-8201a and Itraconazole - Cohort 2

Area under the serum concentration-time curve from time zero to 17 days (AUC17d) and during the dosing interval (AUCtau) were assessed for DS-8201a and total anti-HER2 antibody. (NCT03383692)
Timeframe: Cycle 1: Day 1, pre-dose and end of infusion (EOI); Cycles 2 and 3: Day 1, pre-dose, EOI, 2 hours, 4 hours, 7 hours; Day 2, 4, 8, 12, 17, and 22; Cycles 4, 6 and 8: Day 1, pre-dose and EOI (each cycle is 22 days)

Interventionug*d/mL (Mean)
DS-8201a, Cycle 2 (DS-8201a only): AUC17dDS-8201a, Cycle 2 (DS-8201a only): AUCtauDS-8201a, Cycle 3 (DS-8201a + ritonavir): AUC17dDS-8201a, Cycle 3 (DS-8201a + ritonavir): AUCtauTotal Anti-HER2 antibody, Cycle 2 (DS-8201a only): AUC17dTotal Anti-HER2 antibody, Cycle 2 (DS-8201a only): AUCtauTotal Anti-HER2 antibody, Cycle 3 (DS-8201a + ritonavir): AUC17dTotal Anti-HER2 antibody, Cycle 3 (DS-8201a + ritonavir): AUCtau
Cohort 2: DS-8201a + Itraconazole644706710789707790781883

[back to top]

Pharmacokinetic Parameter of Area Under the Serum Concentration-time Curve Following Treatment With DS-8201a and Ritonavir - Cohort 1

Area under the serum concentration-time curve from time zero to 17 days (AUC17d) and during the dosing interval (AUCtau) for DS-8201a and total anti-HER2 antibody were assessed. (NCT03383692)
Timeframe: Cycle 1: Day 1, pre-dose and end of infusion (EOI); Cycles 2 and 3: Day 1, pre-dose, EOI, 2 hours, 4 hours, 7 hours; Day 2, 4, 8, 12, 17, and 22; Cycles 4, 6 and 8: Day 1, pre-dose and EOI (each cycle is 22 days)

Interventionug*d/mL (Mean)
DS-8201a, Cycle 2 (DS-8201a only): AUC17dDS-8201a, Cycle 2 (DS-8201a only): AUCtauDS-8201a, Cycle 3 (DS-8201a + ritonavir): AUC17dDS-8201a, Cycle 3 (DS-8201a + ritonavir): AUCtauTotal Anti-HER2 antibody, Cycle 2 (DS-8201a only): AUC17dTotal Anti-HER2 antibody, Cycle 2 (DS-8201a only): AUCtauTotal Anti-HER2 antibody, Cycle 3 (DS-8201a + ritonavir): AUC17dTotal Anti-HER2 antibody, Cycle 3 (DS-8201a + ritonavir): AUCtau
Cohort 1: DS-8201a + Ritonavir650701754810723791796860

[back to top]

Pharmacokinetic Parameter of Area Under the Serum Concentration-time Curve of MAAA-1181a Following Treatment With DS-8201a and Itraconazole - Cohort 2

Area under the serum concentration-time curve from time zero to 17 days (AUC17d) and during the dosing interval (AUCtau) were assessed for MAAA-1181a. (NCT03383692)
Timeframe: Cycle 1: Day 1, pre-dose and end of infusion (EOI); Cycles 2 and 3: Day 1, pre-dose, EOI, 2 hours, 4 hours, 7 hours; Day 2, 4, 8, 12, 17, and 22; Cycles 4, 6 and 8: Day 1, pre-dose and EOI (each cycle is 22 days)

Interventionng*d/mL (Mean)
MAAA-1181a, Cycle 2 (DS-8201a only): AUC17dMAAA-1181a, Cycle 2 (DS-8201a only): AUCtauMAAA-1181a, Cycle 3 (DS-8201a + ritonavir): AUC17dMAAA-1181a, Cycle 3 (DS-8201a + ritonavir): AUCtau
Cohort 2: DS-8201a + Itraconazole29.932.434.837.7

[back to top]

Pharmacokinetic Parameter of Maximum (Peak) Observed Serum Concentration (Cmax) Following Treatment With DS-8201a and Itraconazole - Cohort 2

Maximum concentration (Cmax) was assessed for DS-8201a and total Anti-HER2 antibody. (NCT03383692)
Timeframe: Cycle 1: Day 1, pre-dose and end of infusion (EOI); Cycles 2 and 3: Day 1, pre-dose, EOI, 2 hours, 4 hours, 7 hours; Day 2, 4, 8, 12, 17, and 22; Cycles 4, 6 and 8: Day 1, pre-dose and EOI (each cycle is 22 days)

Interventionug/mL (Mean)
DS-8201a, Cycle 2: DS-8201a onlyDS-8201a, Cycle 3: DS-8201a + ritonavirTotal Anti-HER2 antibody, Cycle 2: DS-8201a onlyTotal Anti-HER2 antibody, Cycle 3: DS-8201a + ritonavir
Cohort 1: DS-8201a + Itraconazole139142119130

[back to top]

Pharmacokinetic Parameter of Maximum (Peak) Observed Serum Concentration (Cmax) Following Treatment With DS-8201a and Ritonavir - Cohort 1

Maximum concentration (Cmax) was assessed for DS-8201a and total Anti-HER2 antibody. (NCT03383692)
Timeframe: Cycle 1: Day 1, pre-dose and end of infusion (EOI); Cycles 2 and 3: Day 1, pre-dose, EOI, 2 hours, 4 hours, 7 hours; Day 2, 4, 8, 12, 17, and 22; Cycles 4, 6 and 8: Day 1, pre-dose and EOI (each cycle is 22 days)

Interventionug/mL (Mean)
DS-8201a, Cycle 2: DS-8201a onlyDS-8201a, Cycle 3: DS-8201a + ritonavirTotal Anti-HER2 antibody, Cycle 2: DS-8201a onlyTotal Anti-HER2 antibody, Cycle 3: DS-8201a + ritonavir
Cohort 1: DS-8201a + Ritonavir133140121126

[back to top]

Pharmacokinetic Parameter of Maximum (Peak) Observed Serum Concentration (Cmax) For MAAA-1181a Following Treatment With DS-8201a and Ritonavir - Cohort 1

Maximum concentration (Cmax) was assessed for MAAA-1181a. (NCT03383692)
Timeframe: Cycle 1: Day 1, pre-dose and end of infusion (EOI); Cycles 2 and 3: Day 1, pre-dose, EOI, 2 hours, 4 hours, 7 hours; Day 2, 4, 8, 12, 17, and 22; Cycles 4, 6 and 8: Day 1, pre-dose and EOI (each cycle is 22 days)

Interventionng/mL (Mean)
MAAA-1181a, Cycle 2: DS-8201a onlyMAAA-1181a, Cycle 3: DS-8201a + ritonavir
Cohort 1: DS-8201a + Ritonavir8.988.95

[back to top]

Pharmacokinetic Parameter of Maximum (Peak) Observed Serum Concentration (Cmax) of MAAA-1181a Following Treatment With DS-8201a and Itraconazole - Cohort 2

Maximum concentration (Cmax) was assessed for MAAA-1181a. (NCT03383692)
Timeframe: Cycle 1: Day 1, pre-dose and end of infusion (EOI); Cycles 2 and 3: Day 1, pre-dose, EOI, 2 hours, 4 hours, 7 hours; Day 2, 4, 8, 12, 17, and 22; Cycles 4, 6 and 8: Day 1, pre-dose and EOI (each cycle is 22 days)

Interventionng/mL (Mean)
MAAA-1181a, Cycle 2: DS-8201a onlyMAAA-1181a, Cycle 3: DS-8201a + ritonavir
Cohort 2: DS-8201a + Itraconazole8.658.93

[back to top]

Pharmacokinetic Parameter of Area Under the Serum Concentration-time Curve of MAAA-1181a Following Treatment With DS-8201a and Ritonavir - Cohort 1

Area under the serum concentration-time curve from time zero to 17 days (AUC17d) and during the dosing interval (AUCtau) were assessed for MAAA-1181a. (NCT03383692)
Timeframe: Cycle 1: Day 1, pre-dose and end of infusion (EOI); Cycles 2 and 3: Day 1, pre-dose, EOI, 2 hours, 4 hours, 7 hours; Day 2, 4, 8, 12, 17, and 22; Cycles 4, 6 and 8: Day 1, pre-dose and EOI (each cycle is 22 days)

Interventionng*d/mL (Mean)
MAAA-1181a, Cycle 2 (DS-8201a only): AUC17dMAAA-1181a, Cycle 2 (DS-8201a only): AUCtauMAAA-1181a, Cycle 3 (DS-8201a + ritonavir): AUC17dMAAA-1181a, Cycle 3 (DS-8201a + ritonavir): AUCtau
Cohort 1: DS-8201a + Ritonavir32.735.037.239.2

[back to top]

Objective Response Rate as Confirmed By The Investigator's Assessment in Participants With HER2-Expressing Advanced Solid Malignant Tumors

Objective response rate (defined as participants who achieved CR and PR) was assessed by the investigator based on Response Evaluation Criteria In Solid Tumors (RECIST) version 1.1. CR was defined as a disappearance of all target lesions and PR was defined as at least a 30% decrease in the sum of diameters of target lesions. ORR with confirmation is Objective Response Rate applying a confirmed response of CR/PR in RECIST version 1.1. (NCT03383692)
Timeframe: Baseline up to disease progression, death, lost to follow up, or study discontinuation (whichever comes first), up to approximately 9 months post-dose

InterventionParticipants (Count of Participants)
Cohort 1: DS-8201a + Ritonavir8
Cohort 2: DS-8201a + Itraconazole7

[back to top]

Objective Response Ratio (ORR) as Confirmed By The Investigator's Assessment in Participants With HER2-Expressing Advanced Solid Malignant Tumors

Objective response ratio (ORR) was defined as the proportion of participants who achieved CR and PR as assessed by the investigator based on Response Evaluation Criteria In Solid Tumors (RECIST) version 1.1. CR was defined as a disappearance of all target lesions and PR was defined as at least a 30% decrease in the sum of diameters of target lesions. (NCT03383692)
Timeframe: Baseline up to disease progression, death, lost to follow up, or study discontinuation (whichever comes first), up to approximately 9 months post-dose

InterventionParticipants (Count of Participants)
Cohort 1: DS-8201a + Ritonavir9
Cohort 2: DS-8201a + Itraconazole10

[back to top]

AUCtau of Raltegravir

Area Under the Concentration-time curve during a dosing interval τ at steady state of Tenofovir at Day 5 of treatment B and C of Part 2 of the study (NCT03537404)
Timeframe: Day 5 Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12 hrs post-dose of Day 5 of treatment B and C (Part 2)

Interventionng*h/ml (Geometric Mean)
Treatment B (Part 2)2912.45
Treatment C (Part 2)2653.65

[back to top]

Number of Patients With Abnormal ECG Changes

There were no subjects with abnormal ECG changes during the study (NCT03537404)
Timeframe: Up to 14 Days after the last dose of treatment in period 3 of each Part of the Study

,,
InterventionParticipants (Count of Participants)
Part 1Part 2
Treatment A (Part 1/Part 2)00
Treatment B (Part 1/Part 2)00
Treatment C (Part 1/Part 2)00

[back to top]

Number of Patients With Abnormal Laboratory Values

(NCT03537404)
Timeframe: Up to 14 Days after the last dose of treatment in period 3 of each Part of the Study

,,
InterventionParticipants (Count of Participants)
Part 1Part 2
Treatment A (Part 1)00
Treatment B (Part 1)00
Treatment C (Part 1)21

[back to top]

AUCtau of Tenofovir

Area Under the Concentration-time curve during a dosing interval τ at steady state of Tenofovir at Day 5 of treatment B and C of Part 1 of the study (NCT03537404)
Timeframe: Day 5 Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16, 18 and 24 hrs post-dose on Day 5 of treatment B and C (Part 1)

Interventionng*h/ml (Geometric Mean)
Treatment B (Part 1)2599.95
Treatment C (Part 1)2799.72

[back to top]

Cmax of Narlaprevir

Maximum observed Concentration of Narlaprevir at Day 5 of treatment A and C of Part 1 or 2 of the study (NCT03537404)
Timeframe: Day 5 Pre-dose and 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 10, 12, 16 and 24 hours post-dose on Day 5 of treatment A and C (Part 1/ Part 2)

Interventionng/ml (Geometric Mean)
Treatment A (Part 1)2130.2742
Treatment C (Part 1)2172.233
Treatment A (Part 2)2946.131
Treatment C (Part 2)2880.612

[back to top]

Cmax of Tenofovir

Maximum observed Concentration of Tenofovir at Day 5 of treatment B and C of Part 1 of the study (NCT03537404)
Timeframe: Day 5 Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16, 18 and 24 hrs post-dose on Day 5 of treatment B and C (Part 1)

Interventionng/ml (Geometric Mean)
Treatment B (Part 1)263.037
Treatment C (Part 1)344.796

[back to top]

Number of Patients With Adverse Events

(NCT03537404)
Timeframe: Up to 14 Days after the last dose of treatment in period 3 of each Part of the Study

InterventionParticipants (Count of Participants)
Treatment A (Part 1)1
Treatment B (Part 1)0
Treatment C (Part 1)4
Treatment A (Part 2)0
Treatment B (Part 2)0
Treatment C (Part 2)1

[back to top]

Cmax of Raltegravir

Maximum observed Concentration of Raltegravir at Day 5 of treatment B and C of Part 2 of the study (NCT03537404)
Timeframe: Day 5 Pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12 hrs post-dose of Day 5 of treatment B and C (Part 2)

Interventionng/ml (Geometric Mean)
Treatment B (Part 2)830.204
Treatment C (Part 2)715.726

[back to top]

AUCtau of Narlaprevir

Area Under the Concentration-time curve during a dosing interval τ at steady state of Narlaprevir at Day 5 of treatment A and C of Part 1/ Part 2 of the study (NCT03537404)
Timeframe: Day 5 Pre-dose and 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 10, 12, 16 and 24 hours post-dose on Day 5 of treatment A and C (Part 1/ Part 2)

Interventionng*h/ml (Geometric Mean)
Treatment A (Part 1)20504.31
Treatment C (Part 1)21366.2
Treatment A (Part 2)26199.19
Treatment C (Part 2)24458.48

[back to top]

Number of Patients With Changes in Vital Signs

There were no subjects with abnormal changes in vital signs (NCT03537404)
Timeframe: Up to 14 Days after the last dose of treatment in period 3 of each Part of the Study

InterventionParticipants (Count of Participants)
Treatment A (Part 1)0
Treatment B (Part 1)0
Treatment C (Part 1)0
Treatment A (Part 2)0
Treatment B (Part 2)0
Treatment C (Part 2)0

[back to top]

Number of Participants With Reduction of Hepatic Venous Pressure Gradient (HVPG)

Reduction in hepatic venous pressure gradient (HVPG) measurements by >25% of baseline OR normalization of HVPG (<5 mm Hg) at 24 weeks after completing therapy (NCT03600714)
Timeframe: Baseline and 24 weeks after completing therapy

InterventionParticipants (Count of Participants)
Treatment10

[back to top]

Number of Participants With Sustained Virologic Response

Undetectable HDV RNA at both 12 and 24 weeks post treatment follow-up visits (NCT03600714)
Timeframe: 12 and 24 weeks after completing therapy

InterventionParticipants (Count of Participants)
Treatment3

[back to top]

Number of Participants With Loss of HBsAg at Week 24

Loss of HBsAg from the serum at week 24 (NCT03600714)
Timeframe: Week 24

InterventionParticipants (Count of Participants)
Treatment0

[back to top]

Number of Participants With Loss of HBsAg at Week 12 Weeks After Completing Therapy

Loss of HBsAg from the serum at 12 weeks after completing therapy (NCT03600714)
Timeframe: 12 weeks after completing therapy

InterventionParticipants (Count of Participants)
Treatment0

[back to top]

Number of Participants With Loss of HBsAg at 24 Weeks After Completing Therapy

Loss of HBsAg from the serum at 24 weeks after completing therapy (NCT03600714)
Timeframe: 24 weeks after completing therapy

InterventionParticipants (Count of Participants)
Treatment0

[back to top]

Number of Participants With Decline of Hepatitis D Virus (HDV) RNA Viral Titer of >2 Logs

Decline of HDV RNA viral titer of >2 logs from baseline at 24 weeks of therapy (NCT03600714)
Timeframe: Baseline and 24 weeks

InterventionParticipants (Count of Participants)
Treatment18

[back to top]

Number of Participants Who Discontinue Medication

Discontinuation of the medication before 24 weeks by the clinical team or patient will be considered a failure to tolerate the medicine. (NCT03600714)
Timeframe: 24 weeks

InterventionParticipants (Count of Participants)
Treatment3

[back to top]

Change in Quantitative Log HBsAg Levels From Baseline to Week 24

Change in quantitative log HBsAg levels at from baseline to week 24 (NCT03600714)
Timeframe: Baseline and week 24

Interventionlog IU/mL (Mean)
Treatment0.15

[back to top]

Change in Quantitative Log HBsAg Levels From Baseline 24 Weeks After Completing Therapy

Change in quantitative log HBsAg levels at from baseline to 24 weeks after completing therapy (NCT03600714)
Timeframe: Baseline and 24 weeks after completing therapy

Interventionlog IU/mL (Mean)
Treatment0.16

[back to top]

Number of Participants With Normalization of Serum ALT

Normalization of serum ALT (ALT <20 in females and ALT <31 in males) at the end of therapy, at week 12 of posttherapy follow-up and at week 24 of post-therapy follow-up, OR reduction in serum ALT by >50% of baseline at week 12 of post therapy follow up and week 24 of post therapy follow up. (NCT03600714)
Timeframe: End of therapy, and 12 and 24 weeks after completing therapy

InterventionParticipants (Count of Participants)
Treatment2

[back to top]

Number of Participants With Reduction in Fibroscan Transient Elastography Values

Reduction in Fibroscan transient elastography values by >25% of baseline at end of therapy. (NCT03600714)
Timeframe: Baseline and 24 weeks

InterventionParticipants (Count of Participants)
Treatment4

[back to top]

Number of Participants With Reduction in Histologic Inflammatory Scores (Modified HAI)

Reduction in histologic inflammatory scores (modified HAI) by at least two points with no progression in histologic fibrosis (Ishak) at week 24 post-treatment follow-up. (NCT03600714)
Timeframe: End of treatment and 24 weeks after completing therapy.

InterventionParticipants (Count of Participants)
Treatment6

[back to top]

Number of Participants With Hospitalization or Mortality

Number of participants with hospitalization or mortality (NCT04354428)
Timeframe: Day 28 after enrolment

InterventionParticipants (Count of Participants)
Ascorbic Acid and Folic Acid4
Hydroxychloroquine and Folic Acid2
Hydroxychloroquine and Azithromycin3
Lopinavir-ritonavir0
Ascorbic Acid1

[back to top]

Number of Participants With Serious Adverse Events and Adverse Events Resulting in Treatment Discontinuation

Serious adverse events (including death and hospitalization) and adverse events resulting in treatment discontinuation (NCT04354428)
Timeframe: 28 days from enrolment

InterventionParticipants (Count of Participants)
Ascorbic Acid and Folic Acid6
Hydroxychloroquine and Folic Acid6
Hydroxychloroquine and Azithromycin8
Lopinavir-ritonavir9
Ascorbic Acid1

[back to top]

Number of Persons With Lower Respiratory Tract Infection (LRTI), Defined as Resting Blood Oxygen Saturation (SpO2<93%) Level Sustained for 2 Readings 2 Hours Apart and Presence of Subjective Dyspnea or Cough

Resting blood oxygen saturation (SpO2<93%) level sustained for 2 readings 2 hours apart and presence of subjective dyspnea or cough (NCT04354428)
Timeframe: 28 days from enrolment

InterventionParticipants (Count of Participants)
Ascorbic Acid and Folic Acid2
Hydroxychloroquine and Folic Acid0
Hydroxychloroquine and Azithromycin4
Lopinavir-ritonavir2
Ascorbic Acid1

[back to top]

Time to Clearance of Nasal SARS-CoV-2

Time to clearance of nasal SARS-CoV-2, defined as 2 consecutive negative swabs (NCT04354428)
Timeframe: Day 1 through Day 14 after enrolment

InterventionDays (Median)
Ascorbic Acid and Folic Acid7
Hydroxychloroquine and Folic Acid5
Hydroxychloroquine and Azithromycin6
Lopinavir-ritonavir4
Ascorbic Acid5

[back to top]

Time to Resolution of COVID-19 Symptom Resolution in Days

"COVID-19 symptoms are based on the following criteria:~At least TWO of the following symptoms: Fever (≥ 38ºC), chills, rigors, myalgia, headache, sore throat, new olfactory and taste disorder(s), OR~At least ONE of the following symptoms: cough, shortness of breath or difficulty breathing, OR~Severe respiratory illness with at least 1 of the following:~Clinical or radiological evidence of pneumonia, OR~Acute respiratory distress syndrome (ARDS), OR~LRTI, defined by resting SpO2<93% sustained for 2 readings 2 hours apart AND presence of subjective dyspnea or cough Death or COVID-19-related hospitalizations will count as a failure to resolve symptoms." (NCT04354428)
Timeframe: Day 1 through Day 14 after enrolment

InterventionDays (Median)
Ascorbic Acid and Folic Acid11.5
Hydroxychloroquine and Folic Acid10.5
Hydroxychloroquine and AzithromycinNA
Lopinavir-ritonavirNA
Ascorbic AcidNA

[back to top] [back to top]

Hospital-free Days: Day 1 to Day 29 (Group 2 and Placebo Control Group)

Number of days outside the hospital from Day 1 to Day 29 (Group 2 and Placebo Control Group) (NCT04372628)
Timeframe: Day 1 to Day 29

Interventiondays (Median)
Group 2 - Lopinavir/Ritonavir17
Placebo Control Group17

[back to top]

ICU-free Days: Day 1 to Day 29 (Group 2 and Placebo Control Group)

Number of days outside the ICU from Day 1 to Day 29 (Group 2 and Placebo Control Group) (NCT04372628)
Timeframe: Day 1 to Day 29

Interventiondays (Median)
Group 2 - Lopinavir/Ritonavir17
Placebo Control Group17

[back to top]

Oxygen-free Days: Day 1 to Day 29 (Group 2 and Placebo Control Group)

Number of Days without oxygen Day 1 to Day 29 (Group 2 and Placebo Control Group) (NCT04372628)
Timeframe: Day 1 to Day 29

Interventiondays (Median)
Group 2 - Lopinavir/Ritonavir17
Placebo Control Group17

[back to top]

Time to Hospitalization Day 1 to Day 29 (Group 2 and Placebo Control Group)

Number of days from enrollment to hospitalization (Group 2 and Placebo Control Group) (NCT04372628)
Timeframe: Day 1 to Day 29

Interventiondays (Median)
Group 2 - Lopinavir/Ritonavir6
Placebo Control Group7

[back to top]

Time to Symptom Resolution: Day 1 to Day 29 (Group 2 and Placebo Control Group)

Number of days from enrollment to resolution of COVID-19 symptoms (Group 2 and Placebo Control Group) (NCT04372628)
Timeframe: Day 1 to Day 29

Interventiondays (Median)
Group 2 - Lopinavir/Ritonavir11
Placebo Control Group11

[back to top]

Vasopressor-free Days Through Study Day 29 (Group 2 and Placebo Control Group)

Number of vasopressor-free days through Study Day 29 (Group 2 and Placebo Control Group) (NCT04372628)
Timeframe: Day 1 to Day 29

Interventiondays (Median)
Group 2 - Lopinavir/Ritonavir17
Placebo Control Group17

[back to top]

Fever-free Days: Day 1 to Day 29 (Group 2 and Placebo Control Group)

Number of days without fever from Day 1 to Day 29 (Group 2 and Placebo Control Group) (NCT04372628)
Timeframe: Day 1 to Day 29

Interventiondays (Median)
Group 2 - Lopinavir/Ritonavir16
Placebo Control Group17

[back to top]

Ventilator-free Days: Day 1 to Day 29 (Group 2 and Placebo Control Group)

Number of days without ventilator use from Day 1 to Day 29 (Group 2 and Placebo Control Group) (NCT04372628)
Timeframe: Day 1 to Day 29

Interventiondays (Median)
Group 2 - Lopinavir/Ritonavir17
Placebo Control Group17

[back to top]

Cohort 1: AUC(0-tau) of DRV

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionHours*micrograms per milliliter (Geometric Mean)
Cohort 1: DRV/RTV 600/100 mg57.47
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg53.37

[back to top]

Cohort 1: AUC(0-tau) of RTV

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionHours* micrograms per milliliter (Geometric Mean)
Cohort 1: DRV/RTV 600/100 mg7.303
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg7.790

[back to top]

Cohort 1: Cmax of DRV

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 1: DRV/RTV 600/100 mg7.037
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg7.268

[back to top]

Cohort 1: Cmax of RTV

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 1: DRV/RTV 600/100 mg1.178
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg1.306

[back to top]

Cohort 1: Ctau of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 1: GSK3640254 200 mg0.8152
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg0.9530

[back to top]

Cohort 1: Ctau of RTV

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 1: DRV/RTV 600/100 mg0.3194
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg0.3314

[back to top]

Cohort 1: Maximum Observed Concentration (Cmax) of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 1: GSK3640254 200 mg1.752
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg1.863

[back to top]

Cohort 1: Plasma Concentration at the End of the Dosing Interval (Ctau) of DRV

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 1: DRV/RTV 600/100 mg2.957
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg2.637

[back to top]

Cohort 1: Time of Maximum Observed Concentration (Tmax) of DRV

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionHours (Median)
Cohort 1: DRV/RTV 600/100 mg3.000
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg3.000

[back to top]

Cohort 1: Tmax of RTV

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionHours (Median)
Cohort 1: DRV/RTV 600/100 mg4.000
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg4.000

[back to top]

Cohort 2: AUC(0-tau) of ETR

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionHours*micrograms per milliliter (Geometric Mean)
Cohort 2: ETR 200 mg8.340
Cohort 2: GSK3640254 200 mg + ETR 200 mg9.791

[back to top]

Cohort 2: AUC(0-tau) of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 3

InterventionHours*micrograms per milliliter (Geometric Mean)
Cohort 2: GSK3640254 200 mg27.86
Cohort 2: GSK3640254 200 mg + ETR 200 mg14.73

[back to top]

Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Urinalysis Parameters

Urine samples were collected for urinalysis parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Erythrocytes High, Grade 3: Gross, with or without clots OR with RBC casts OR intervention indicated, Grade 4: Life-threatening consequences; Glucose High, Grade 3: >2+ (proportionate concentration by dipstick test) or >500 mg, Grade 4: >500 mg; Protein High, Grade 3: 3+ (proportionate concentration by dipstick test) or higher, Grade 4: Not Applicable. Baseline was defined as the latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 26

,
InterventionParticipants (Count of Participants)
Erythrocytes, High, Increase to Grade 3Erythrocytes, High, Increase to Grade 4Glucose, High, Increase to Grade 3Glucose, High, Increase to Grade 4Protein, High, Increase to Grade 3Protein, High, Increase to Grade 4
Cohort 3: GSK3640254 200 mg000000
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg000000

[back to top]

Cohort 1: Area Under the Plasma Concentration-time Curve From Time Zero to the End of the Dosing Interval at Steady State (AUC[0-tau]) of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 3

InterventionHours*micrograms per milliliter (Geometric Mean)
Cohort 1: GSK3640254 200 mg27.03
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg30.70

[back to top]

Cohort 3: AUC(0-tau) of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 2

InterventionHours*micrograms per milliliter (Geometric Mean)
Cohort 3: GSK3640254 200 mg24.99
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg22.78

[back to top]

Cohort 2: Tmax of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 3

InterventionHours (Median)
Cohort 2: GSK3640254 200 mg4.000
Cohort 2: GSK3640254 200 mg + ETR 200 mg3.000

[back to top]

Cohort 2: Tmax of ETR

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionHours (Median)
Cohort 2: ETR 200 mg3.500
Cohort 2: GSK3640254 200 mg + ETR 200 mg4.000

[back to top]

Cohort 2: Number of Participants With Clinically Significant Abnormal ECG Findings

A 12-lead ECG was recorded with the participant in a supine position using an automated ECG machine. Clinically significant abnormal findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. Data for number of participants with clinically significant abnormal ECG findings were reported. Data has been presented for the participants with respect to the actual treatment received in respective treatment periods. (NCT04630002)
Timeframe: Day 1 (2,4,6 Hours), Day 7 and Day 11 in Treatment Period 1; Day 12 (2,4,6 Hours), Day 21 in Treatment Period 2; Day 22 (2,4,6 Hours), Day 26 and Day 36 in Treatment Period 3

InterventionParticipants (Count of Participants)
2 Hours, Day 14 Hours, Day 16 Hours; Day 1Day 7Day 11
Cohort 2: GSK3640254 200 mg00000

[back to top]

Cohort 2: Ctau of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 2: GSK3640254 200 mg0.7706
Cohort 2: GSK3640254 200 mg + ETR 200 mg0.3901

[back to top]

Cohort 2: Ctau of ETR

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 2: ETR 200 mg0.4705
Cohort 2: GSK3640254 200 mg + ETR 200 mg0.5837

[back to top]

Cohort 2: Cmax of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 2: GSK3640254 200 mg1.889
Cohort 2: GSK3640254 200 mg + ETR 200 mg1.136

[back to top]

Cohort 2: Cmax of ETR

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 2: ETR 200 mg0.9749
Cohort 2: GSK3640254 200 mg + ETR 200 mg1.102

[back to top]

Cohort 1: Number of Participants With Vital Sign Values of Potential Clinical Importance (PCI) Criteria

Vital signs including systolic blood pressure (SBP), diastolic blood pressure (DBP) and pulse rate were measured in a supine position after atleast 5 minutes of rest. The PCI ranges for vitals were as follows; for SBP <85 or >140 millimeters of mercury (mmHg), for DBP <45 or >90 mmHg, for pulse rate <40 or >100 beats per minute. The number of participants with vital signs of PCI were presented. (NCT04630002)
Timeframe: Up to Day 35

,,
InterventionParticipants (Count of Participants)
SBPDBPPulse rate
Cohort 1: DRV/RTV 600/100 mg200
Cohort 1: GSK3640254 200 mg000
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg000

[back to top]

Cohort 2: Number of Participants With AEs Leading to Discontinuations and Deaths

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study intervention, whether or not considered related to the study intervention. Number of participants with AEs leading to discontinuations and deaths were reported. (NCT04630002)
Timeframe: Up to Day 36

,,
InterventionParticipants (Count of Participants)
AEs leading to discontinuationsAES leading to deaths
Cohort 2: ETR 200 mg00
Cohort 2: GSK3640254 200 mg00
Cohort 2: GSK3640254 200 mg + ETR 200 mg10

[back to top]

Cohort 1: Number of Participants With Clinically Significant Abnormal Electrocardiogram (ECG) Findings

A 12-lead ECG was recorded with the participant in a supine position using an automated ECG machine. Clinically significant abnormal findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. Data for number of participants with clinically significant abnormal ECG findings were reported. Data has been presented for the participants with respect to the actual treatment received in respective treatment periods. (NCT04630002)
Timeframe: Day 1 (2,4,6 Hours), Day 7 and Day 11 in Treatment Period 1; Day 12 (2,4,6 Hours), Day 21 in Treatment Period 2; Day 22 (2,4,6 Hours), Day 26 and Day 35 in Treatment Period 3

InterventionParticipants (Count of Participants)
2 Hours, Day 14 Hours, Day 16 Hours; Day 1Day 7Day 11
Cohort 1: GSK3640254 200 mg00000

[back to top]

Cohort 2: Number of Participants With Clinically Significant Abnormal ECG Findings

A 12-lead ECG was recorded with the participant in a supine position using an automated ECG machine. Clinically significant abnormal findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. Data for number of participants with clinically significant abnormal ECG findings were reported. Data has been presented for the participants with respect to the actual treatment received in respective treatment periods. (NCT04630002)
Timeframe: Day 1 (2,4,6 Hours), Day 7 and Day 11 in Treatment Period 1; Day 12 (2,4,6 Hours), Day 21 in Treatment Period 2; Day 22 (2,4,6 Hours), Day 26 and Day 36 in Treatment Period 3

InterventionParticipants (Count of Participants)
2 Hours, Day 124 Hours, Day 126 Hours; Day 12Day 21
Cohort 2: ETR 200 mg0000

[back to top]

Cohort 1: Number of Participants With Clinically Significant Abnormal Electrocardiogram (ECG) Findings

A 12-lead ECG was recorded with the participant in a supine position using an automated ECG machine. Clinically significant abnormal findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. Data for number of participants with clinically significant abnormal ECG findings were reported. Data has been presented for the participants with respect to the actual treatment received in respective treatment periods. (NCT04630002)
Timeframe: Day 1 (2,4,6 Hours), Day 7 and Day 11 in Treatment Period 1; Day 12 (2,4,6 Hours), Day 21 in Treatment Period 2; Day 22 (2,4,6 Hours), Day 26 and Day 35 in Treatment Period 3

InterventionParticipants (Count of Participants)
2 Hours, Day 124 Hours, Day 126 Hours; Day 12Day 21
Cohort 1: DRV/RTV 600/100 mg0000

[back to top]

Cohort 1: Number of Participants With AEs Leading to Discontinuations and Deaths

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study intervention, whether or not considered related to the study intervention. Number of participants with AEs leading to discontinuations and deaths were reported. (NCT04630002)
Timeframe: Up to Day 35

,,
InterventionParticipants (Count of Participants)
AEs leading to discontinuationsAES leading to deaths
Cohort 1: DRV/RTV 600/100 mg10
Cohort 1: GSK3640254 200 mg00
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg00

[back to top]

Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Calcium, Creatine Kinase, Creatinine, Phosphate, Potassium and Sodium

Blood samples were collected for analysis of clinical chemistry parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Calcium High, Grade 3: 12.5 to <13.5 mg/dL, Grade 4: >=13.5 mg/dL; Calcium Low, Grade 3: 6.1 to <7.0 mg/dL, Grade 4: <6.1 mg/dL; Creatine Kinase High, Grade 3: 10 to <20 × ULN, Grade 4: >=20 × ULN; Creatinine High, Grade 3: >1.8 to <3.5 ULN, Grade 4: >=3.5 × ULN; Phosphate Low, Grade 3: 1.0 to <1.4 mg/dL, Grade 4: <1.0 mg/dL; Potassium High, Grade 3: 6.5 to <7.0 mEq/L,Grade 4: >=7.0 mEq/L; Potassium Low, Grade 3: 2.0 to <2.5 mEq/L, Grade 4: <2.00 mEq/L; Sodium High, Grade 3: 154 to <160 mEq/L, Grade 4:>=160 mEq/L; Sodium Low, Grade 3: 121 to <125 mEq/L, Grade 4:<=120 mEq/L. Baseline was defined as the latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 36

,,
InterventionParticipants (Count of Participants)
Calcium, Low, Increase to Grade 3Calcium, Low, Increase to Grade 4Calcium, High, Increase to Grade 3Calcium, High, Increase to Grade 4Creatine Kinase, High, Increase to Grade 3Creatine Kinase, High, Increase to Grade 4Creatinine, High, Increase to Grade 3Creatinine, High, Increase to Grade 4Phosphate, Low, Increase to Grade 3Phosphate, Low, Increase to Grade 4Potassium, Low, Increase to Grade 3Potassium, Low, Increase to Grade 4Potassium, High, Increase to Grade 3Potassium, High, Increase to Grade 4Sodium, Low, Increase to Grade 3Sodium, Low, Increase to Grade 4Sodium, High, Increase to Grade 3Sodium, High, Increase to Grade 4
Cohort 2: ETR 200 mg000000000000000000
Cohort 2: GSK3640254 200 mg000000000000000000
Cohort 2: GSK3640254 200 mg + ETR 200 mg000000000000000000

[back to top]

Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Alanine Aminotransferase, Albumin, Alkaline Phosphatase, Amylase, Aspartate Aminotransferase, Bilirubin and Direct Bilirubin

Blood samples were collected for analysis of clinical chemistry parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Alanine Aminotransferase High; Grade 3: 5.0 to <10.0 × ULN, Grade 4: >=10.0 × ULN; Albumin Low, Grade 3: <2.0 g/dL, Grade 4: Not Applicable; Alkaline Phosphatase High, Grade 3: 5.0 to <10.0 × ULN, Grade 4: >=10.0 × ULN; Amylase High, Grade 3: 3.0 to <5.0 × ULN, Grade 4: >=5.0 × ULN; Aspartate Aminotransferase High, Grade 3: 5.0 to <10.0 × ULN, Grade 4: >=10.0 × ULN; Bilirubin High, Grade 3: 2.6 to<5.0 × ULN, Grade 4: >=5.0 × ULN and Direct Bilirubin High, Grade 3: >ULN with other signs and symptoms of hepatotoxicity, Grade 4: >ULN with life-threatening consequences. Baseline was defined as latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 36

,,
InterventionParticipants (Count of Participants)
Alanine Aminotransferase, High, Increase to Grade 3Alanine Aminotransferase, High, Increase to Grade 4Albumin, Low, Increase to Grade 3Albumin, Low, Increase to Grade 4Alkaline Phosphatase, High, Increase to Grade 3Alkaline Phosphatase, High, Increase to Grade 4Amylase, High, Increase to Grade 3Amylase, High, Increase to Grade 4Aspartate Aminotransferase, High, Increase to Grade 3Aspartate Aminotransferase, High, Increase to Grade 4Bilirubin, High, Increase to Grade 3Bilirubin, High, Increase to Grade 4Direct Bilirubin, High, Increase to Grade 3Direct Bilirubin, High, Increase to Grade 4
Cohort 2: ETR 200 mg00000000000000
Cohort 2: GSK3640254 200 mg00000000000000
Cohort 2: GSK3640254 200 mg + ETR 200 mg00000000000000

[back to top]

Cohort 2: Number of Participants With Clinically Significant Abnormal ECG Findings

A 12-lead ECG was recorded with the participant in a supine position using an automated ECG machine. Clinically significant abnormal findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. Data for number of participants with clinically significant abnormal ECG findings were reported. Data has been presented for the participants with respect to the actual treatment received in respective treatment periods. (NCT04630002)
Timeframe: Day 1 (2,4,6 Hours), Day 7 and Day 11 in Treatment Period 1; Day 12 (2,4,6 Hours), Day 21 in Treatment Period 2; Day 22 (2,4,6 Hours), Day 26 and Day 36 in Treatment Period 3

InterventionParticipants (Count of Participants)
2 Hours; Day 224 Hours; Day 226 Hours; Day 22Day 26Day 36
Cohort 2: GSK3640254 200 mg + ETR 200 mg00000

[back to top]

Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Glucose, Triglycerides, Lipase, Urate and Cholesterol

Blood samples were collected for analysis of clinical chemistry parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Glucose High, Grade 3: >250 to 500 mg/dL, Grade 4: >=500 mg/dL, Glucose Low, Grade 3: 30 to<40 mg/dL, Grade 4:<30 mg/dL; Triglycerides High, Grade 3: >500 to <1.000 mg/dL, Grade 4:>1000 mg/dL; Lipase High, Grade 3: 3.0 to <5.0×ULN, Grade 4:>=5.0×ULN; Urate High, Grade 3: 12.0 to <15.0 mEq/L, Grade 4:>=15.0 mEq/L; Cholesterol High, Grade 3: >=300 mg/dL, Grade 4: Not Applicable. Baseline was defined as the latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 36

,,
InterventionParticipants (Count of Participants)
Glucose, Low, Increase to Grade 3Glucose, Low, Increase to Grade 4Glucose, High, Increase to Grade 3Glucose, High, Increase to Grade 4Triglycerides, High, Increase to Grade 3Triglycerides, High, Increase to Grade 4Lipase, High, Increase to Grade 3Lipase, High, Increase to Grade 4Urate, High, Increase to Grade 3Urate, High, Increase to Grade 4Cholesterol, High, Increase to Grade 3Cholesterol, High, Increase to Grade 4
Cohort 2: ETR 200 mg000000000000
Cohort 2: GSK3640254 200 mg000000000000
Cohort 2: GSK3640254 200 mg + ETR 200 mg000000000000

[back to top]

Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Hematology Parameters

Blood samples were collected for analysis of hematology parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Hemoglobin Low, Grade 3: 7.0 to <9.0 g/dL (males) and 6.5 to <8.5 g/dL (females),Grade 4: <7.0 g/dL (males) and <6.5 g/dL (females); Leukocytes Low, Grade 3: 1000 to 1499 cells/mm^3,Grade 4: <1000 cells/mm^3; Lymphocytes Low, Grade 3: 350 to <500 cells/L,Grade 4: <350 cells/L; Neutrophils Low, Grade 3: 400 to 599 cells/mm^3, Grade 4: <400 cells/mm^3; Platelets Low, Grade 3: 25,000 to <50,000 cells/mm^3, Grade 4: <25,000 cells/mm^3. Baseline was defined as latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 36

,,
InterventionParticipants (Count of Participants)
Hemoglobin, Low, Increase to Grade 3Hemoglobin, Low, Increase to Grade 4Leukocytes, Low, Increase to Grade 3Leukocytes, Low, Increase to Grade 4Lymphocytes, Low, Increase to Grade 3Lymphocytes, Low, Increase to Grade 4Neutrophils, Low, Increase to Grade 3Neutrophils, Low, Increase to Grade 4Platelets, Low, Increase to Grade 3Platelets, Low, Increase to Grade 4
Cohort 2: ETR 200 mg0000000000
Cohort 2: GSK3640254 200 mg0000000000
Cohort 2: GSK3640254 200 mg + ETR 200 mg0000000000

[back to top]

Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Urinalysis Parameters

Urine samples were collected for urinalysis parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Erythrocytes High, Grade 3: Gross, with or without clots OR with RBC casts OR intervention indicated, Grade 4: Life-threatening consequences; Glucose High, Grade 3: >2+ (proportionate concentration by dipstick test) or >500 mg, Grade 4: >500 mg; Protein High, Grade 3: 3+ (proportionate concentration by dipstick test) or higher, Grade 4: Not Applicable. Baseline was defined as the latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 36

,,
InterventionParticipants (Count of Participants)
Erythrocytes, High, Increase to Grade 3Erythrocytes, High, Increase to Grade 4Glucose, High, Increase to Grade 3Glucose, High, Increase to Grade 4Protein, High, Increase to Grade 3Protein, High, Increase to Grade 4
Cohort 2: ETR 200 mg000000
Cohort 2: GSK3640254 200 mg000000
Cohort 2: GSK3640254 200 mg + ETR 200 mg000000

[back to top]

Cohort 2: Number of Participants With SAEs and Non-SAEs

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study intervention, whether or not considered related to the study intervention. An SAE is defined as any untoward medical occurrence that, at any dose that results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect or other situations as per medical or scientific judgment. Adverse events which were not Serious were considered as Non-Serious adverse events. (NCT04630002)
Timeframe: Up to Day 36

,,
InterventionParticipants (Count of Participants)
SAEnon-SAE
Cohort 2: ETR 200 mg04
Cohort 2: GSK3640254 200 mg03
Cohort 2: GSK3640254 200 mg + ETR 200 mg07

[back to top]

Cohort 2: Number of Participants With Vital Sign Values of PCI Criteria

Vital signs including SBP, DBP and pulse rate were measured in a supine position after atleast 5 minutes of rest. The PCI ranges for vitals were as follows; for SBP <85 or >140 mmHg, for DBP <45 or >90 mmHg, for pulse rate <40 or >100 beats per minute. The number of participants with vital signs of PCI were presented. (NCT04630002)
Timeframe: Up to Day 36

,,
InterventionParticipants (Count of Participants)
SBPDBPPulse rate
Cohort 2: ETR 200 mg000
Cohort 2: GSK3640254 200 mg000
Cohort 2: GSK3640254 200 mg + ETR 200 mg010

[back to top]

Cohort 3: Number of Participants With AEs Leading to Discontinuations and Deaths

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study intervention, whether or not considered related to the study intervention. Number of participants with AEs leading to discontinuations and deaths were reported. (NCT04630002)
Timeframe: Up to Day 26

,
InterventionParticipants (Count of Participants)
AEs leading to discontinuationsAES leading to deaths
Cohort 3: GSK3640254 200 mg00
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg10

[back to top]

Cohort 3: Number of Participants With Clinically Significant Abnormal ECG Findings

A 12-lead ECG was recorded with the participant in a supine position using an automated ECG machine. Clinically significant abnormal findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. Data for number of participants with clinically significant abnormal ECG findings were reported. Data has been presented for the participants with respect to the actual treatment received in respective treatment periods. (NCT04630002)
Timeframe: Day 1 (2,4,6 Hours) in Treatment Period 1; Day 8 (2,4,6 Hours), Day 9 (2,4,6 Hours), Day 26 in Treatment Period 2

InterventionParticipants (Count of Participants)
2 Hours, Day 14 Hours, Day 16 Hours, Day 1
Cohort 3: GSK3640254 200 mg000

[back to top]

Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Alanine Aminotransferase, Albumin, Alkaline Phosphatase, Amylase, Aspartate Aminotransferase, Bilirubin and Direct Bilirubin

Blood samples were collected for analysis of clinical chemistry parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Alanine Aminotransferase High; Grade 3: 5.0 to <10.0 × ULN, Grade 4: >=10.0 × ULN; Albumin Low, Grade 3: <2.0 g/dL, Grade 4: Not Applicable; Alkaline Phosphatase High, Grade 3: 5.0 to <10.0 × ULN, Grade 4: >=10.0 × ULN; Amylase High, Grade 3: 3.0 to <5.0 × ULN, Grade 4: >=5.0 × ULN; Aspartate Aminotransferase High, Grade 3: 5.0 to <10.0 × ULN, Grade 4: >=10.0 × ULN; Bilirubin High, Grade 3: 2.6 to<5.0 × ULN, Grade 4: >=5.0 × ULN and Direct Bilirubin High, Grade 3: >ULN with other signs and symptoms of hepatotoxicity, Grade 4: >ULN with life-threatening consequences. Baseline was defined as latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 26

,
InterventionParticipants (Count of Participants)
Alanine Aminotransferase, High, Increase to Grade 3Alanine Aminotransferase, High, Increase to Grade 4Albumin, Low, Increase to Grade 3Albumin, Low, Increase to Grade 4Alkaline Phosphatase, High, Increase to Grade 3Alkaline Phosphatase, High, Increase to Grade 4Amylase, High, Increase to Grade 3Amylase, High, Increase to Grade 4Aspartate Aminotransferase, High, Increase to Grade 3Aspartate Aminotransferase, High, Increase to Grade 4Bilirubin, High, Increase to Grade 3Bilirubin, High, Increase to Grade 4Direct Bilirubin, High, Increase to Grade 3Direct Bilirubin, High, Increase to Grade 4
Cohort 3: GSK3640254 200 mg00000000000000
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg00000000000000

[back to top]

Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Calcium, Creatine Kinase, Creatinine, Phosphate, Potassium and Sodium

Blood samples were collected for analysis of clinical chemistry parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Calcium High, Grade 3: 12.5 to <13.5 mg/dL, Grade 4: >=13.5 mg/dL; Calcium Low, Grade 3: 6.1 to <7.0 mg/dL, Grade 4: <6.1 mg/dL; Creatine Kinase High, Grade 3: 10 to <20 × ULN, Grade 4: >=20 × ULN; Creatinine High, Grade 3: >1.8 to <3.5 ULN, Grade 4: >=3.5 × ULN; Phosphate Low, Grade 3: 1.0 to <1.4 mg/dL, Grade 4: <1.0 mg/dL; Potassium High, Grade 3: 6.5 to <7.0 mEq/L,Grade 4: >=7.0 mEq/L; Potassium Low, Grade 3: 2.0 to <2.5 mEq/L, Grade 4: <2.00 mEq/L; Sodium High, Grade 3: 154 to <160 mEq/L, Grade 4:>=160 mEq/L; Sodium Low, Grade 3: 121 to <125 mEq/L, Grade 4:<=120 mEq/L. Baseline was defined as the latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 26

,
InterventionParticipants (Count of Participants)
Calcium, Low, Increase to Grade 3Calcium, Low, Increase to Grade 4Calcium, High, Increase to Grade 3Calcium, High, Increase to Grade 4Creatine Kinase, High, Increase to Grade 3Creatine Kinase, High, Increase to Grade 4Creatinine, High, Increase to Grade 3Creatinine, High, Increase to Grade 4Phosphate, Low, Increase to Grade 3Phosphate, Low, Increase to Grade 4Potassium, Low, Increase to Grade 3Potassium, Low, Increase to Grade 4Potassium, High, Increase to Grade 3Potassium, High, Increase to Grade 4Sodium, Low, Increase to Grade 3Sodium, Low, Increase to Grade 4Sodium, High, Increase to Grade 3Sodium, High, Increase to Grade 4
Cohort 3: GSK3640254 200 mg000000000000000000
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg000000000000000000

[back to top]

Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Glucose, Triglycerides, Lipase, Urate and Cholesterol

Blood samples were collected for analysis of clinical chemistry parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Glucose High, Grade 3: >250 to 500 mg/dL, Grade 4: >=500 mg/dL, Glucose Low, Grade 3: 30 to<40 mg/dL, Grade 4:<30 mg/dL; Triglycerides High, Grade 3: >500 to <1.000 mg/dL, Grade 4:>1000 mg/dL; Lipase High, Grade 3: 3.0 to <5.0×ULN, Grade 4:>=5.0×ULN; Urate High, Grade 3: 12.0 to <15.0 mEq/L, Grade 4:>=15.0 mEq/L; Cholesterol High, Grade 3: >=300 mg/dL, Grade 4: Not Applicable. Baseline was defined as the latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 26

,
InterventionParticipants (Count of Participants)
Glucose, Low, Increase to Grade 3Glucose, Low, Increase to Grade 4Glucose, High, Increase to Grade 3Glucose, High, Increase to Grade 4Triglycerides, High, Increase to Grade 3Triglycerides, High, Increase to Grade 4Lipase, High, Increase to Grade 3Lipase, High, Increase to Grade 4Urate, High, Increase to Grade 3Urate, High, Increase to Grade 4Cholesterol, High, Increase to Grade 3Cholesterol, High, Increase to Grade 4
Cohort 3: GSK3640254 200 mg000000000000
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg000000000000

[back to top]

Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Hematology Parameters

Blood samples were collected for analysis of hematology parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Hemoglobin Low, Grade 3: 7.0 to <9.0 g/dL (males) and 6.5 to <8.5 g/dL (females),Grade 4: <7.0 g/dL (males) and <6.5 g/dL (females); Leukocytes Low, Grade 3: 1000 to 1499 cells/mm^3,Grade 4: <1000 cells/mm^3; Lymphocytes Low, Grade 3: 350 to <500 cells/L,Grade 4: <350 cells/L; Neutrophils Low, Grade 3: 400 to 599 cells/mm^3, Grade 4: <400 cells/mm^3; Platelets Low, Grade 3: 25,000 to <50,000 cells/mm^3, Grade 4: <25,000 cells/mm^3. Baseline was defined as latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 26

,
InterventionParticipants (Count of Participants)
Hemoglobin, Low, Increase to Grade 3Hemoglobin, Low, Increase to Grade 4Leukocytes, Low, Increase to Grade 3Leukocytes, Low, Increase to Grade 4Lymphocytes, Low, Increase to Grade 3Lymphocytes, Low, Increase to Grade 4Neutrophils, Low, Increase to Grade 3Neutrophils, Low, Increase to Grade 4Platelets, Low, Increase to Grade 3Platelets, Low, Increase to Grade 4
Cohort 3: GSK3640254 200 mg0000000000
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg0000000000

[back to top]

Cohort 3: Number of Participants With Clinically Significant Abnormal ECG Findings

A 12-lead ECG was recorded with the participant in a supine position using an automated ECG machine. Clinically significant abnormal findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. Data for number of participants with clinically significant abnormal ECG findings were reported. Data has been presented for the participants with respect to the actual treatment received in respective treatment periods. (NCT04630002)
Timeframe: Day 1 (2,4,6 Hours) in Treatment Period 1; Day 8 (2,4,6 Hours), Day 9 (2,4,6 Hours), Day 26 in Treatment Period 2

InterventionParticipants (Count of Participants)
2 Hours, Day 84 Hours, Day 86 Hours, Day 82 Hours; Day 94 Hours; Day 96 Hours; Day 9Day 26
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg0000000

[back to top]

Cohort 3: Number of Participants With SAEs and Non-SAEs

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study intervention, whether or not considered related to the study intervention. An SAE is defined as any untoward medical occurrence that, at any dose that results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect or other situations as per medical or scientific judgment. Adverse events which were not Serious were considered as Non-Serious adverse events. (NCT04630002)
Timeframe: Up to Day 26

,
InterventionParticipants (Count of Participants)
SAEnon-SAE
Cohort 3: GSK3640254 200 mg01
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg08

[back to top]

Cohort 3: Number of Participants With Vital Sign Values of PCI Criteria

Vital signs including SBP, DBP and pulse rate were measured in a supine position after atleast 5 minutes of rest. The PCI ranges for vitals were as follows; for SBP <85 or >140 mmHg, for DBP <45 or >90 mmHg, for pulse rate <40 or >100 beats per minute. The number of participants with vital signs of PCI were presented. (NCT04630002)
Timeframe: Up to Day 26

,
InterventionParticipants (Count of Participants)
SBPDBPPulse rate
Cohort 3: GSK3640254 200 mg100
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg000

[back to top]

Cohort 3: Cmax of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 2

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 3: GSK3640254 200 mg1.578
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg1.383

[back to top]

Cohort 1: Number of Participants With Serious Adverse Events (SAEs) and Non-serious Adverse Events (Non-SAEs)

An adverse event (AE) is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study intervention, whether or not considered related to the study intervention. An SAE is defined as any untoward medical occurrence that, at any dose that results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect or other situations as per medical or scientific judgment. Adverse events which were not Serious were considered as Non-Serious adverse events. (NCT04630002)
Timeframe: Up to Day 35

,,
InterventionParticipants (Count of Participants)
SAEnon-SAE
Cohort 1: DRV/RTV 600/100 mg04
Cohort 1: GSK3640254 200 mg06
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg05

[back to top]

Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Urinalysis Parameters

Urine samples were collected for urinalysis parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Erythrocytes High, Grade 3: Gross, with or without clots OR with Red Blood Cells (RBC) casts OR intervention indicated, Grade 4: Life-threatening consequences; Glucose High, Grade 3: >2+ (proportionate concentration by dipstick test) or >500 mg, Grade 4: >500 mg; Protein High, Grade 3: 3+ (proportionate concentration by dipstick test) or higher, Grade 4: Not Applicable. Baseline was defined as the latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 35

,,
InterventionParticipants (Count of Participants)
Erythrocytes, High, Increase to Grade 3Erythrocytes, High, Increase to Grade 4Glucose, High, Increase to Grade 3Glucose, High, Increase to Grade 4Protein, High, Increase to Grade 3Protein, High, Increase to Grade 4
Cohort 1: DRV/RTV 600/100 mg000000
Cohort 1: GSK3640254 200 mg000000
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg000000

[back to top]

Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Hematology Parameters

Blood samples were collected for analysis of hematology parameters. Laboratory abnormalities were graded according to Division of Acquired Immunodeficiency Syndrome (DAIDS) grading table Version 2.1. For Hemoglobin Low, Grade 3: 7.0 to <9.0 Grams per deciliter (g/dL) (males) and 6.5 to <8.5 g/dL (females),Grade 4: <7.0 g/dL (males) and <6.5 g/dL (females); Leukocytes Low, Grade 3: 1000 to 1499 cells per cubic millimeter (cells/mm^3),Grade 4: <1000 cells/mm^3; Lymphocytes Low, Grade 3: 350 to <500 cells per liter (cells/L),Grade 4: <350 cells/L; Neutrophils Low, Grade 3: 400 to 599 cells/mm^3, Grade 4: <400 cells/mm^3; Platelets Low, Grade 3: 25,000 to <50,000 cells/mm^3, Grade 4: <25,000 cells/mm^3. Baseline was defined as latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 35

,,
InterventionParticipants (Count of Participants)
Hemoglobin, Low, Increase to Grade 3Hemoglobin, Low, Increase to Grade 4Leukocytes, Low, Increase to Grade 3Leukocytes, Low, Increase to Grade 4Lymphocytes, Low, Increase to Grade 3Lymphocytes, Low, Increase to Grade 4Neutrophils, Low, Increase to Grade 3Neutrophils, Low, Increase to Grade 4Platelets, Low, Increase to Grade 3Platelets, Low, Increase to Grade 4
Cohort 1: DRV/RTV 600/100 mg0000000000
Cohort 1: GSK3640254 200 mg0000000000
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg0000000000

[back to top]

Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Glucose, Triglycerides, Lipase, Urate and Cholesterol

Blood samples were collected for analysis of clinical chemistry parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Glucose High, Grade 3: >250 to 500 mg/dL, Grade 4: >=500 mg/dL, Glucose Low, Grade 3: 30 to<40 mg/dL, Grade 4:<30 mg/dL; Triglycerides High, Grade 3: >500 to <1.000 mg/dL, Grade 4:>1000 mg/dL; Lipase High, Grade 3: 3.0 to <5.0×ULN, Grade 4:>=5.0×ULN; Urate High, Grade 3: 12.0 to <15.0 mEq/L, Grade 4:>=15.0 mEq/L; Cholesterol High, Grade 3: >=300 mg/dL, Grade 4: Not Applicable. Baseline was defined as the latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 35

,,
InterventionParticipants (Count of Participants)
Glucose, Low, Increase to Grade 3Glucose, Low, Increase to Grade 4Glucose, High, Increase to Grade 3Glucose, High, Increase to Grade 4Triglycerides, High, Increase to Grade 3Triglycerides, High, Increase to Grade 4Lipase, High, Increase to Grade 3Lipase, High, Increase to Grade 4Urate, High, Increase to Grade 3Urate, High, Increase to Grade 4Cholesterol, High, Increase to Grade 3Cholesterol, High, Increase to Grade 4
Cohort 1: DRV/RTV 600/100 mg000000000000
Cohort 1: GSK3640254 200 mg000000000000
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg000000000000

[back to top]

Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Calcium, Creatine Kinase, Creatinine, Phosphate, Potassium and Sodium

Blood samples were collected for analysis of clinical chemistry parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Calcium High, Grade 3: 12.5 to <13.5 milligrams/deciliter (mg/dL), Grade 4: >=13.5 mg/dL; Calcium Low, Grade 3: 6.1 to <7.0 mg/dL, Grade 4: <6.1 mg/dL; Creatine Kinase High, Grade 3: 10 to <20 × ULN, Grade 4: >=20 × ULN; Creatinine High, Grade 3: >1.8 to <3.5 ULN, Grade 4: >=3.5 × ULN; Phosphate Low, Grade 3: 1.0 to <1.4 mg/dL, Grade 4: <1.0 mg/dL; Potassium High, Grade 3: 6.5 to <7.0 Milliequivalents per liter (mEq/L),Grade 4: >=7.0 mEq/L; Potassium Low, Grade 3: 2.0 to <2.5 mEq/L, Grade 4: <2.00 mEq/L; Sodium High, Grade 3: 154 to <160 mEq/L, Grade 4:>=160 mEq/L; Sodium Low, Grade 3: 121 to <125 mEq/L, Grade 4:<=120 mEq/L. Baseline was defined as the latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 35

,,
InterventionParticipants (Count of Participants)
Calcium, Low, Increase to Grade 3Calcium, Low, Increase to Grade 4Calcium, High, Increase to Grade 3Calcium, High, Increase to Grade 4Creatine Kinase, High, Increase to Grade 3Creatine Kinase, High, Increase to Grade 4Creatinine, High, Increase to Grade 3Creatinine, High, Increase to Grade 4Phosphate, Low, Increase to Grade 3Phosphate, Low, Increase to Grade 4Potassium, Low, Increase to Grade 3Potassium, Low, Increase to Grade 4Potassium, High, Increase to Grade 3Potassium, High, Increase to Grade 4Sodium, Low, Increase to Grade 3Sodium, Low, Increase to Grade 4Sodium, High, Increase to Grade 3Sodium, High, Increase to Grade 4
Cohort 1: DRV/RTV 600/100 mg000000000000000000
Cohort 1: GSK3640254 200 mg000000000000000000
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg000000000000000000

[back to top]

Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Alanine Aminotransferase, Albumin, Alkaline Phosphatase, Amylase, Aspartate Aminotransferase, Bilirubin and Direct Bilirubin

Blood samples were collected for analysis of clinical chemistry parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Alanine Aminotransferase High; Grade 3: 5.0 to <10.0 times (×) Upper Limit Normal (ULN), Grade 4: >=10.0 × ULN; Albumin Low, Grade 3: <2.0 grams per deciliter (g/dL), Grade 4: Not Applicable; Alkaline Phosphatase High, Grade 3: 5.0 to <10.0 × ULN, Grade 4: >=10.0 × ULN; Amylase High, Grade 3: 3.0 to <5.0 × ULN, Grade 4: >=5.0 × ULN; Aspartate Aminotransferase High, Grade 3: 5.0 to <10.0 × ULN, Grade 4: >=10.0 × ULN; Bilirubin High, Grade 3: 2.6 to<5.0 × ULN, Grade 4: >=5.0 × ULN and Direct Bilirubin High, Grade 3: >ULN with other signs and symptoms of hepatotoxicity, Grade 4: >ULN with life-threatening consequences. Baseline was defined as latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 35

,,
InterventionParticipants (Count of Participants)
Alanine Aminotransferase, High, Increase to Grade 3Alanine Aminotransferase, High, Increase to Grade 4Albumin, Low, Increase to Grade 3Albumin, Low, Increase to Grade 4Alkaline Phosphatase, High, Increase to Grade 3Alkaline Phosphatase, High, Increase to Grade 4Amylase, High, Increase to Grade 3Amylase, High, Increase to Grade 4Aspartate Aminotransferase, High, Increase to Grade 3Aspartate Aminotransferase, High, Increase to Grade 4Bilirubin, High, Increase to Grade 3Bilirubin, High, Increase to Grade 4Direct Bilirubin, High, Increase to Grade 3Direct Bilirubin, High, Increase to Grade 4
Cohort 1: DRV/RTV 600/100 mg00000000000000
Cohort 1: GSK3640254 200 mg00000000000000
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg00000000000000

[back to top]

Cohort 1: Number of Participants With Clinically Significant Abnormal Electrocardiogram (ECG) Findings

A 12-lead ECG was recorded with the participant in a supine position using an automated ECG machine. Clinically significant abnormal findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. Data for number of participants with clinically significant abnormal ECG findings were reported. Data has been presented for the participants with respect to the actual treatment received in respective treatment periods. (NCT04630002)
Timeframe: Day 1 (2,4,6 Hours), Day 7 and Day 11 in Treatment Period 1; Day 12 (2,4,6 Hours), Day 21 in Treatment Period 2; Day 22 (2,4,6 Hours), Day 26 and Day 35 in Treatment Period 3

InterventionParticipants (Count of Participants)
2 Hours; Day 224 Hours; Day 226 Hours; Day 22Day 26Day 35
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg00000

[back to top]

Cohort 1: Tmax of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 3

InterventionHours (Median)
Cohort 1: GSK3640254 200 mg4.000
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg4.000

[back to top]

Number of Participants With Clinical Laboratory Abnormalities

The hematology, clinical chemistry and urinalysis tests were included in the laboratory examination. The criteria for hematology evaluation included hemoglobin less than (<) 0.8*lower limit of normal (LLN), hematocrit <0.8*LLN, erythrocytes <0.8*LLN, erythrocyte mean corpuscular hemoglobin <0.9*LLN and lymphocytes <0.8*LLN. The criteria for clinical chemistry evaluation included neutrophils <0.8*LLN, eosinophils greater than (>) 1.2* upper limit of normal (ULN), monocytes >1.2*ULN, urea nitrogen >1.3*ULN, creatinine >1.3*ULN, urate >1.2*ULN, potassium >1.1*ULN and bicarbonate <0.9*LLN. The criteria for urinalysis evaluation included thyrotropin >1.2*ULN, glucose >1.5*ULN, fibrinogen >1.25*baseline, ketones greater than or equal to (>=) 1, urine protein >=1, nitrite >=1 and leukocyte esterase >=1. (NCT04909853)
Timeframe: Part 1 and Part 2: Day -1 up to maximum of 35 days after last dose (maximum of 38 days)

InterventionParticipants (Count of Participants)
Part 1: Normal Renal Function5
Part 1: Mild Renal Impairment4
Part 1: Moderate Renal Impairment8
Part 2: Severe Renal Impairment8

[back to top]

Number of Participants With Clinically Significant Findings in Physical Examination

A complete physical examination included, at a minimum, head, ears, eyes, nose, mouth, skin, heart and lung examinations, lymph nodes, and gastrointestinal, musculoskeletal, and neurological systems. Clinical significance was judged by investigator. (NCT04909853)
Timeframe: Part 1 and Part 2: Day -1 up to maximum of 35 days after last dose (maximum of 38 days)

InterventionParticipants (Count of Participants)
Part 1: Normal Renal Function0
Part 1: Mild Renal Impairment1
Part 1: Moderate Renal Impairment0
Part 2: Severe Renal Impairment0

[back to top]

Number of Participants With Clinically Significant Vital Signs Abnormalities

Supine blood pressure, pulse rate, respiratory rate and oral temperature were evaluated in vital signs examination. Clinical significance was judged by investigator. (NCT04909853)
Timeframe: Part 1 and Part 2: Day -1 up to maximum of 35 days after last dose (maximum of 38 days)

InterventionParticipants (Count of Participants)
Part 1: Normal Renal Function0
Part 1: Mild Renal Impairment0
Part 1: Moderate Renal Impairment0
Part 2: Severe Renal Impairment1

[back to top]

Plasma Concentration of PF-07321332 at 12 Hours Post Dose (C12)

(NCT04909853)
Timeframe: Part 1 and Part 2: 0 (pre-dose), 0.5, 1, 2, 3, 4, 6, 8, 10 and 12 hours post-dose on Day 1

InterventionNanogram per milliliter (Geometric Mean)
Part 1: Normal Renal Function341.9
Part 1: Mild Renal Impairment438.0
Part 1: Moderate Renal Impairment785.6
Part 2: Severe Renal Impairment1213

[back to top]

Plasma Concentration of PF-07321332 at 24 Hours Post Dose (C24)

(NCT04909853)
Timeframe: Part 1 and Part 2: 0 (pre-dose), 0.5, 1, 2, 3, 4, 6, 8, 10, 12 and 24 hours post-dose on Day 1

InterventionNanogram per milliliter (Geometric Mean)
Part 1: Normal Renal Function99.10
Part 1: Mild Renal Impairment112.8
Part 1: Moderate Renal Impairment179.1
Part 2: Severe Renal Impairment694.2

[back to top]

Renal Clearance (CLr) of PF-07321332

Renal clearance was calculated as total amount of unchanged drug excreted in the urine over 48 hours (Ae48) divided by area under the plasma concentration-time profile from time 0 to 48 hours post dose. (NCT04909853)
Timeframe: Part 1 and Part 2: 0 to 48 hours post dose on Day 1

InterventionLiters per hour (Geometric Mean)
Part 1: Normal Renal Function2.180
Part 1: Mild Renal Impairment2.395
Part 1: Moderate Renal Impairment1.154
Part 2: Severe Renal Impairment0.4398

[back to top]

Terminal Elimination Plasma Half-life (t1/2) of PF-07321332

t1/2 was calculated by Loge(2)/kel. kel was the terminal phase rate constant calculated by a linear regression of the log-linear concentration-time curve. (NCT04909853)
Timeframe: Part 1 and Part 2: 0 (pre-dose), 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36 and 48 hours post-dose on Day 1

InterventionHours (Mean)
Part 1: Normal Renal Function7.725
Part 1: Mild Renal Impairment6.606
Part 1: Moderate Renal Impairment9.948
Part 2: Severe Renal Impairment13.37

[back to top]

Time to Reach Maximum Observed Plasma Concentration (Tmax) of PF-07321332

Tmax was observed directly from data as time of first occurrence. (NCT04909853)
Timeframe: Part 1 and Part 2: 0 (pre-dose), 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36 and 48 hours post-dose on Day 1

InterventionHours (Median)
Part 1: Normal Renal Function2.000
Part 1: Mild Renal Impairment2.000
Part 1: Moderate Renal Impairment2.500
Part 2: Severe Renal Impairment3.000

[back to top] [back to top]

Number of Participants With Clinically Significant 12-Lead Electrocardiogram (ECG) Abnormalities

A standard 12-lead ECGs utilizing limb leads (with a 10 second rhythm strip) were collected using an ECG machine that automatically calculates the heart rate and measures PR, QT, and QTc intervals and QRS complex. All scheduled ECGs were performed after the participant had rested quietly for at least 5 minutes in a supine position. Clinical significance was judged by investigator. (NCT04909853)
Timeframe: Part 1 and Part 2: Day -1 up to maximum of 35 days after last dose (maximum of 38 days)

InterventionParticipants (Count of Participants)
Part 1: Normal Renal Function0
Part 1: Mild Renal Impairment0
Part 1: Moderate Renal Impairment0
Part 2: Severe Renal Impairment1

[back to top]

Maximum Observed Plasma Concentration (Cmax) of PF-07321332

(NCT04909853)
Timeframe: Part 1 and Part 2: 0 (pre-dose), 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36 and 48 hours post-dose on Day 1

InterventionNanogram per milliliter (Geometric Mean)
Part 1: Normal Renal Function1600
Part 1: Mild Renal Impairment2077
Part 1: Moderate Renal Impairment2210
Part 2: Severe Renal Impairment2369

[back to top]

Area Under the Plasma Concentration-time Profile From Time Zero (0) to Extrapolated Infinite Time (AUCinf) of PF-07321332

AUCinf was calculated by AUClast + (Clast/kel). AUClast was the area under the plasma concentration-time profile from time 0 to the time of Clast. Clast was the predicted plasma concentration at the last quantifiable time point estimated from the log-linear regression analysis and kel was the terminal phase rate constant calculated by a linear regression of the log-linear concentration-time curve. (NCT04909853)
Timeframe: Part 1 and Part 2: 0 (pre-dose), 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36 and 48 hours post-dose on Day 1

InterventionNanogram*hour per milliliter (Geometric Mean)
Part 1: Normal Renal Function14460
Part 1: Mild Renal Impairment17910
Part 1: Moderate Renal Impairment27110
Part 2: Severe Renal Impairment44040

[back to top]

Area Under the Plasma Concentration-time Profile From Time Zero (0) to the Time of the Last Quantifiable Concentration (AUClast) of PF-07321332

Area under the plasma concentration-time profile from time 0 to the time of the last quantifiable concentration was determined by linear/log trapezoidal method. (NCT04909853)
Timeframe: Part 1 and Part 2: 0 (pre-dose), 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36 and 48 hours post-dose on Day 1

InterventionNanogram*hour per milliliter (Geometric Mean)
Part 1: Normal Renal Function14270
Part 1: Mild Renal Impairment17770
Part 1: Moderate Renal Impairment26660
Part 2: Severe Renal Impairment39420

[back to top]

Apparent Volume of Distribution (Vz/F) of PF-07321332

Volume of distribution is defined as the theoretical volume in which the total amount of drug would need to be uniformly distributed to produce the desired plasma concentration of a drug. Apparent volume of distribution after oral dose (Vz/F) is influenced by the fraction absorbed. Vz/F was calculated by dose/(AUCinf*kel). kel was the terminal phase rate constant calculated by a linear regression of the log-linear concentration-time curve. (NCT04909853)
Timeframe: Part 1 and Part 2: 0 (pre-dose), 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36 and 48 hours post-dose on Day 1

InterventionLiters (Geometric Mean)
Part 1: Normal Renal Function74.95
Part 1: Mild Renal Impairment51.95
Part 1: Moderate Renal Impairment50.34
Part 2: Severe Renal Impairment42.73

[back to top]

Apparent Clearance (CL/F) of PF-07321332 From Plasma

Clearance of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. Clearance obtained after oral dose (apparent clearance) is influenced by the fraction of the dose absorbed. Drug clearance is a quantitative measure of the rate at which a drug substance is removed from the plasma. CL/F was calculated by Dose/AUCinf. (NCT04909853)
Timeframe: Part 1 and Part 2: 0 (pre-dose), 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 24, 36 and 48 hours post-dose on Day 1

InterventionLiters per hour (Geometric Mean)
Part 1: Normal Renal Function6.913
Part 1: Mild Renal Impairment5.581
Part 1: Moderate Renal Impairment3.689
Part 2: Severe Renal Impairment2.270

[back to top]

Amount of PF-07321332 Excreted Unchanged in Urine Over 48 Hours (Ae48)

Total amount of unchanged drug excreted in the urine over 48 hours. (NCT04909853)
Timeframe: Part 1 and Part 2: 0 to 48 hours post dose on Day 1

InterventionMilligram (Geometric Mean)
Part 1: Normal Renal Function31.20
Part 1: Mild Renal Impairment42.65
Part 1: Moderate Renal Impairment30.83
Part 2: Severe Renal Impairment18.46

[back to top]

Percentage of Participants With Severe Covid-19 Signs and Symptoms Through Day 28- mITT1 Population

Participants were required to record the severity of their Covid-19 symptoms over the past 24 hours daily on a 4-point scale ranging from 0 to 3, higher scores indicated more severity. The scale was reported as 0= no symptoms, 1=mild, 2=moderate and 3=severe. A participant with severe score for any targeted symptoms post-baseline was counted as severe. Percentage of participants with severe Covid-19 signs and symptoms were reported. (NCT04960202)
Timeframe: From Day 1 to Day 28

InterventionPercentage of participants (Number)
PF-07321332 300 mg + Ritonavir 100 mg19.006
Placebo20.829

[back to top]

Percentage of Participants With Severe Covid-19 Signs and Symptoms Through Day 28- mITT Population

Participants were required to record the severity of their Covid-19 symptoms over the past 24 hours daily on a 4-point scale ranging from 0 to 3, higher scores indicated more severity. The scale was reported as 0= no symptoms, 1=mild, 2=moderate and 3=severe. A participant with severe score for any targeted symptoms post-baseline was counted as severe. Percentage of participants with severe Covid-19 signs and symptoms were reported. (NCT04960202)
Timeframe: From Day 1 to Day 28

InterventionPercentage of participants (Number)
PF-07321332 300 mg + Ritonavir 100 mg17.930
Placebo20.326

[back to top] [back to top] [back to top]

Percentage of Participants Who Died Through Week 24- mITT2 Population

In this outcome measure, percentage of participants with death due to any cause was presented. (NCT04960202)
Timeframe: From Day 1 up to Week 24

InterventionPercentage of participants (Number)
PF-07321332 300 mg + Ritonavir 100 mg0
Placebo1.3

[back to top]

Percentage of Participants Who Died Through Week 24- mITT1 Population

In this outcome measure, percentage of participants with death due to any cause was presented. (NCT04960202)
Timeframe: From Day 1 up to Week 24

InterventionPercentage of participants (Number)
PF-07321332 300 mg + Ritonavir 100 mg0
Placebo1.4

[back to top]

Time to Sustained Alleviation of All Targeted COVID-19 Signs and Symptoms Through Day 28- Modified Intent-to-Treat 2 (mITT2) Population

Sustained alleviation of all targeted COVID-19 signs/symptoms was defined as the event occurring on the first 4 consecutive days when all symptoms scored as moderate or severe at the time of enrollment were scored as mild or absent and those scored mild or absent at the time of enrollment were scored as absent. The first day of the 4 consecutive-day period was considered date of first event. Time to sustained alleviation (event) was calculated as first event date minus first dose date plus 1, for participants with event. For participants who completed Day 28 or discontinued the study before Day 28 without sustained alleviation (censored), time was calculated as censoring date (last date on which symptom alleviation was assessed) minus first dose date plus 1 or Day 25 whichever occurred first. (NCT04960202)
Timeframe: From Day 1 (baseline) to Day 28

InterventionDays (Median)
PF-07321332 300 mg + Ritonavir 100 mg13.00
Placebo15.00

[back to top]

Percentage of Participants Who Died Through Week 24- mITT Population

In this outcome measure, percentage of participants with death due to any cause was presented. (NCT04960202)
Timeframe: From Day 1 up to Week 24

InterventionPercentage of participants (Number)
PF-07321332 300 mg + Ritonavir 100 mg0
Placebo1.6

[back to top]

Number of Participants With Treatment Emergent Adverse Events (TEAEs)

An adverse event (AE) was any untoward medical occurrence in a participant, temporarily associated with the use of study intervention, whether or not considered related to the study intervention. Serious adverse event (SAE) was any untoward medical occurrence that, at any dose: resulted in death; required inpatient hospitalization or prolongation of existing hospitalization; was life-threatening; resulted in persistent or significant disability/ incapacity; congenital anomaly/birth defect; a suspected transmission via a Pfizer product of an infectious agent, pathogenic or non-pathogenic and other important medical events. AEs included both SAEs and all non-SAEs. An AE was considered as TEAE if the event started on or after start date of study intervention. (NCT04960202)
Timeframe: From start of study intervention (Day 1) up to end of safety follow-up (Day 34)

InterventionParticipants (Count of Participants)
PF-07321332 300 mg + Ritonavir 100 mg256
Placebo270

[back to top]

Number of Participants With Progression to a Worsening Status in 1 or More Self-reported COVID-19 Associated Symptoms Through Day 28-mITT2 Population

Participants were required to record the severity of their Covid-19 symptoms over the past 24 hours daily on a 4-point scale where 0 = no symptoms; 1= mild; 2= moderate; and 3= severe. Vomiting and diarrhea were each rated on a 4-point frequency scale where 0= no occurrence, 1= mild for 1 to 2 times, 2= moderate for 3 to 4 times, and 3= severe for 5 or greater. Progression to a worsening status for any targeted symptom was based up on increasing severity (i.e. the first time any targeted symptoms worsened after treatment relative to baseline). (NCT04960202)
Timeframe: From Day 1 (baseline) to Day 28

InterventionParticipants (Count of Participants)
PF-07321332 300 mg + Ritonavir 100 mg819
Placebo822

[back to top]

Number of Participants With Progression to a Worsening Status in 1 or More Self-reported COVID-19 Associated Symptoms Through Day 28-mITT1 Population

Participants were required to record the severity of their Covid-19 symptoms over the past 24 hours daily on a 4-point scale where 0 = no symptoms; 1= mild; 2= moderate; and 3= severe. Vomiting and diarrhea were each rated on a 4-point frequency scale where 0= no occurrence, 1= mild for 1 to 2 times, 2= moderate for 3 to 4 times, and 3= severe for 5 or greater. Progression to a worsening status for any targeted symptom was based up on increasing severity (i.e. the first time any targeted symptoms worsened after treatment relative to baseline). (NCT04960202)
Timeframe: From Day 1 (baseline) to Day 28

InterventionParticipants (Count of Participants)
PF-07321332 300 mg + Ritonavir 100 mg763
Placebo768

[back to top]

Number of Participants With Progression to a Worsening Status in 1 or More Self-reported COVID-19 Associated Symptoms Through Day 28-mITT Population

Participants were required to record the severity of their Covid-19 symptoms over the past 24 hours daily on a 4-point scale where 0 = no symptoms; 1= mild; 2= moderate; and 3= severe. Vomiting and diarrhea were each rated on a 4-point frequency scale where 0= no occurrence, 1= mild for 1 to 2 times, 2= moderate for 3 to 4 times, and 3= severe for 5 or greater. Progression to a worsening status for any targeted symptom was based up on increasing severity (i.e. the first time any targeted symptoms worsened after treatment relative to baseline). (NCT04960202)
Timeframe: From Day 1 (baseline) to Day 28

InterventionParticipants (Count of Participants)
PF-07321332 300 mg + Ritonavir 100 mg523
Placebo504

[back to top]

Time to Sustained Resolution of Each Targeted COVID-19 Signs and Symptoms- mITT1 Population

Sustained resolution was defined as when each targeted symptom was scored as absent for 4 consecutive days. Time to sustained resolution (event) was calculated as first event date minus first dose date plus 1, for participants with event. For participants who completed Day 28 or discontinued the study before Day 28 without sustained resolution (censored), time was calculated as censoring date (last date on which symptom resolution was assessed) minus first dose date plus 1 or Day 25 whichever occurred first. (NCT04960202)
Timeframe: From Day 1 (baseline) to Day 28

,
InterventionDays (Median)
Muscle or body achesShortness of breath or difficulty breathingChills or shiveringCoughDiarrheaFeeling hot or feverishHeadacheNauseaStuffy or runny noseSore throatVomit
PF-07321332 300 mg + Ritonavir 100 mg9.0008.0005.00013.0006.0005.0009.0007.0009.0007.0003.000
Placebo12.00012.0007.00015.0006.0007.00011.0007.00010.0009.0003.000

[back to top]

Time to Sustained Resolution of Each Targeted COVID-19 Signs and Symptoms- mITT Population

Sustained resolution was defined as when each targeted symptom was scored as absent for 4 consecutive days. Time to sustained resolution (event) was calculated as first event date minus first dose date plus 1, for participants with event. For participants who completed Day 28 or discontinued the study before Day 28 without sustained resolution (censored), time was calculated as censoring date (last date on which symptom resolution was assessed) minus first dose date plus 1 or Day 25 whichever occurred first. (NCT04960202)
Timeframe: From Day 1 (baseline) to Day 28

,
InterventionDays (Median)
Muscle or body achesShortness of breath or difficulty breathingChills or shiveringCoughDiarrheaFeeling hot or feverishHeadacheNauseaStuffy or runny noseSore throatVomit
PF-07321332 300 mg + Ritonavir 100 mg9.0008.0005.00013.0006.0005.0008.0005.0009.0007.0003.000
Placebo12.00011.0006.00014.0006.0007.00011.0007.00010.0009.0003.000

[back to top]

Time to Sustained Alleviation of Each Targeted COVID-19 Signs and Symptoms- mITT2 Population

Sustained alleviation of each targeted COVID-19 signs/symptoms was defined as the event occurring on the first 4 consecutive days when each symptom scored as moderate or severe at the time of enrollment were scored as mild or absent and those scored mild or absent at the time of enrollment were scored as absent. The first day of the 4 consecutive-day period was considered date of first event. Time to sustained alleviation (event) was calculated as first event date minus first dose date plus 1, for participants with event. For participants who completed Day 28 or discontinued the study before Day 28 without sustained alleviation (censored), time was calculated as censoring date (last date on which symptom alleviation was assessed) minus first dose date plus 1 or Day 25 whichever occurred first. (NCT04960202)
Timeframe: From Day 1 (baseline) to Day 28

,
InterventionDays (Median)
Muscle or body achesShortness of breath or difficulty breathingChills or shiveringCoughDiarrheaFeeling hot or feverishHeadacheNauseaStuffy or runny noseSore throatVomit
PF-07321332 300 mg + Ritonavir 100 mg6.0006.0003.0008.0005.0003.0005.0005.0006.0005.0003.000
Placebo8.0008.0004.00010.0004.0004.0007.0006.0007.0006.0003.000

[back to top]

Time to Sustained Alleviation of Each Targeted COVID-19 Signs and Symptoms- mITT1 Population

Sustained alleviation of each targeted COVID-19 signs/symptoms was defined as the event occurring on the first 4 consecutive days when each symptom scored as moderate or severe at the time of enrollment were scored as mild or absent and those scored mild or absent at the time of enrollment were scored as absent. The first day of the 4 consecutive-day period was considered date of first event. Time to sustained alleviation (event) was calculated as first event date minus first dose date plus 1, for participants with event. For participants who completed Day 28 or discontinued the study before Day 28 without sustained alleviation (censored), time was calculated as censoring date (last date on which symptom alleviation was assessed) minus first dose date plus 1 or Day 25 whichever occurred first. (NCT04960202)
Timeframe: From Day 1 (baseline) to Day 28

,
InterventionDays (Median)
Muscle or body achesShortness of breath or difficulty breathingChills or shiveringCoughDiarrheaFeeling hot or feverishHeadacheNauseaStuffy or runny noseSore throatVomit
PF-07321332 300 mg + Ritonavir 100 mg6.006.003.008.005.003.005.005.006.005.003.00
Placebo7.008.004.0010.004.005.007.006.007.006.003.00

[back to top]

Time to Sustained Alleviation of Each Targeted COVID-19 Signs and Symptoms- mITT Population

Sustained alleviation of each targeted COVID-19 signs/symptoms was defined as the event occurring on the first 4 consecutive days when each symptom scored as moderate or severe at the time of enrollment were scored as mild or absent and those scored mild or absent at the time of enrollment were scored as absent. The first day of the 4 consecutive-day period was considered date of first event. Time to sustained alleviation (event) was calculated as first event date minus first dose date plus 1, for participants with event. For participants who completed Day 28 or discontinued the study before Day 28 without sustained alleviation (censored), time was calculated as censoring date (last date on which symptom alleviation was assessed) minus first dose date plus 1 or Day 25 whichever occurred first. (NCT04960202)
Timeframe: From Day 1 (baseline) to Day 28

,
InterventionDays (Median)
Muscle or body achesShortness of breath or difficulty breathingChills or shiveringCoughDiarrheaFeeling hot or feverishHeadacheNauseaStuffy or runny noseSore throatVomit
PF-07321332 300 mg + Ritonavir 100 mg6.006.003.008.004.003.005.004.006.005.003.00
Placebo7.007.004.0010.004.004.007.005.007.005.003.00

[back to top]

Plasma Concentration Versus Time Summary of PF-07321332

(NCT04960202)
Timeframe: 1 Hour post-dose on Day 1 and pre-dose on Day 5

InterventionNanograms per milliliter (Mean)
Day 1 (1 Hour post dose)Day 5 (Pre-dose)
PF-07321332 300 mg + Ritonavir 100 mg21862964

[back to top]

Percentage of Participants With a Resting Peripheral Oxygen Saturation >=95% at Days 1 and 5- mITT2 Population

In this outcome measure, the percentage of participants with a resting peripheral oxygen saturation >=95% were reported. (NCT04960202)
Timeframe: Day 1, 5

,
InterventionPercentage of participants (Number)
Day 1Day 5
PF-07321332 300 mg + Ritonavir 100 mg93.5991.81
Placebo92.1988.13

[back to top]

Percentage of Participants With a Resting Peripheral Oxygen Saturation >=95% at Days 1 and 5- mITT1 Population

In this outcome measure, the percentage of participants with a resting peripheral oxygen saturation >=95% were reported. (NCT04960202)
Timeframe: Day 1, 5

,
InterventionPercentage of participants (Number)
Day 1Day 5
PF-07321332 300 mg + Ritonavir 100 mg93.6491.77
Placebo92.6388.13

[back to top]

Percentage of Participants With a Resting Peripheral Oxygen Saturation >=95% at Days 1 and 5- mITT Population

In this outcome measure, the percentage of participants with a resting peripheral oxygen saturation >=95% were reported. (NCT04960202)
Timeframe: Day 1, 5

,
InterventionPercentage of participants (Number)
Day 1Day 5
PF-07321332 300 mg + Ritonavir 100 mg93.5493.09
Placebo92.3789.68

[back to top]

Number of Participants With AEs Leading to Discontinuation and Serious Adverse Events (SAEs)

An AE was any untoward medical occurrence in a participant, temporarily associated with the use of study treatment, whether or not considered related to the study treatment. An SAE was any untoward medical occurrence that, at any dose: resulted in death; required inpatient hospitalization or prolongation of existing hospitalization; was life-threatening; resulted in persistent or significant disability/ incapacity; congenital anomaly/birth defect; a suspected transmission via a Pfizer product of an infectious agent, pathogenic or non-pathogenic and other important medical events. (NCT04960202)
Timeframe: From start of study intervention (Day 1) up to end of safety follow-up (Day 34)

,
InterventionParticipants (Count of Participants)
AEs leading to study discontinuationSAEs
PF-07321332 300 mg + Ritonavir 100 mg019
Placebo1374

[back to top]

Number of Days in Hospital and ICU for the Treatment of COVID-19- mITT2 Population

(NCT04960202)
Timeframe: From Day 1 up to Day 34

,
InterventionDays (Mean)
Duration of hospitalization visitsDuration of ICU visitsDuration of non-ICU visits
PF-07321332 300 mg + Ritonavir 100 mg0.0860.0000.086
Placebo0.6970.1140.584

[back to top]

Number of Days in Hospital and ICU for the Treatment of COVID-19- mITT1 Population

(NCT04960202)
Timeframe: From Day 1 up to Day 34

,
InterventionDays (Mean)
Duration of hospitalization visitsDuration of ICU visitsDuration of non-ICU visits
PF-07321332 300 mg + Ritonavir 100 mg0.0820.0000.082
Placebo0.7330.1210.613

[back to top]

Number of Days in Hospital and ICU for the Treatment of COVID-19- mITT Population

(NCT04960202)
Timeframe: From Day 1 up to Day 34

,
InterventionDays (Mean)
Duration of hospitalization visitsDuration of ICU visitsDuration of non-ICU visits
PF-07321332 300 mg + Ritonavir 100 mg0.0850.0000.085
Placebo0.8010.1700.632

[back to top]

Change From Baseline in Logarithm to Base10 (Log10) Transformed Viral Load at Day 3, 5, 10 and 14- mITT Population

The viral load was measured in nasal or nasopharyngeal samples using reverse transcription polymerase chain reaction (RT-PCR). (NCT04960202)
Timeframe: Baseline, Day 3, 5, 10 and 14

,
InterventionLog10 copies per milliliter (Mean)
Day 3Day 5Day 10Day 14
PF-07321332 300 mg + Ritonavir 100 mg-1.821-3.202-4.532-5.098
Placebo-1.199-2.252-3.984-4.840

[back to top]

Change From Baseline in Log10 Transformed Viral Load at Day 3, 5, 10 and 14- mITT2 Population

The viral load was measured in nasal or nasopharyngeal samples using RT-PCR. (NCT04960202)
Timeframe: Baseline, Day 3, 5, 10 and 14

,
InterventionLog10 copies per milliliter (Mean)
Day 3Day 5Day 10Day 14
PF-07321332 300 mg + Ritonavir 100 mg-1.788-2.994-4.288-4.801
Placebo-1.187-2.184-3.778-4.506

[back to top]

Change From Baseline in Log10 Transformed Viral Load at Day 3, 5, 10 and 14- mITT1 Population

The viral load was measured in nasal or nasopharyngeal samples using RT-PCR. (NCT04960202)
Timeframe: Baseline, Day 3, 5, 10 and 14

,
InterventionLog10 copies per milliliter (Mean)
Day 3Day 5Day 10Day 14
PF-07321332 300 mg + Ritonavir 100 mg-1.756-2.977-4.274-4.813
Placebo-1.191-2.166-3.773-4.521

[back to top]

Time to Sustained Resolution of All Targeted COVID-19 Signs and Symptoms Through Day 28- mITT2 Population

Sustained resolution was defined as when all targeted symptoms were scored as absent for 4 consecutive days. Time to sustained resolution (event) was calculated as first event date minus first dose date plus 1, for participants with event. For participants who completed Day 28 or discontinued the study before Day 28 without sustained resolution (censored), time was calculated as censoring date (last date on which symptom resolution was assessed) minus first dose date plus 1 or Day 25 whichever occurred first. (NCT04960202)
Timeframe: From Day 1 (baseline) to Day 28

InterventionDays (Median)
PF-07321332 300 mg + Ritonavir 100 mg16.00
Placebo19.00

[back to top] [back to top]

Percentage of Participants With Severe Covid-19 Signs and Symptoms Through Day 28- mITT2 Population

Participants were required to record the severity of their Covid-19 symptoms over the past 24 hours daily on a 4-point scale ranging from 0 to 3, higher scores indicated more severity. The scale was reported as 0= no symptoms, 1=mild, 2=moderate and 3=severe. A participant with severe score for any targeted symptoms post-baseline was counted as severe. Percentage of participants with severe Covid-19 signs and symptoms were reported. (NCT04960202)
Timeframe: From Day 1 to Day 28

InterventionPercentage of participants (Number)
PF-07321332 300 mg + Ritonavir 100 mg19.854
Placebo21.267

[back to top]

Time to Sustained Resolution of All Targeted COVID-19 Signs and Symptoms Through Day 28- mITT1 Population

Sustained resolution was defined as when all targeted symptoms were scored as absent for 4 consecutive days. Time to sustained resolution (event) was calculated as first event date minus first dose date plus 1, for participants with event. For participants who completed Day 28 or discontinued the study before Day 28 without sustained resolution (censored), time was calculated as censoring date (last date on which symptom resolution was assessed) minus first dose date plus 1 or Day 25 whichever occurred first. (NCT04960202)
Timeframe: From Day 1 (baseline) to Day 28

InterventionDays (Median)
PF-07321332 300 mg + Ritonavir 100 mg16.00
Placebo19.00

[back to top]

Time to Sustained Resolution of All Targeted COVID-19 Signs and Symptoms Through Day 28- mITT Population

Sustained resolution was defined as when all targeted symptoms were scored as absent for 4 consecutive days. Time to sustained resolution (event) was calculated as first event date minus first dose date plus 1, for participants with event. For participants who completed Day 28 or discontinued the study before Day 28 without sustained resolution (censored), time was calculated as censoring date (last date on which symptom resolution was assessed) minus first dose date plus 1 or Day 25 whichever occurred first. (NCT04960202)
Timeframe: From Day 1 (baseline) to Day 28

InterventionDays (Median)
PF-07321332 300 mg + Ritonavir 100 mg16.00
Placebo18.00

[back to top] [back to top]

Time to Sustained Alleviation of All Targeted COVID-19 Signs and Symptoms Through Day 28- mITT1 Population

Sustained alleviation of all targeted COVID-19 signs/symptoms was defined as the event occurring on the first 4 consecutive days when all symptoms scored as moderate or severe at the time of enrollment were scored as mild or absent and those scored mild or absent at the time of enrollment were scored as absent. The first day of the 4 consecutive-day period was considered date of first event. Time to sustained alleviation (event) was calculated as first event date minus first dose date plus 1, for participants with event. For participants who completed Day 28 or discontinued the study before Day 28 without sustained alleviation (censored), time was calculated as censoring date (last date on which symptom alleviation was assessed) minus first dose date plus 1 or Day 25 whichever occurred first. (NCT04960202)
Timeframe: From Day 1 (baseline) to Day 28

InterventionDays (Median)
PF-07321332 300 mg + Ritonavir 100 mg13.00
Placebo15.00

[back to top]

Time to Sustained Alleviation of All Targeted COVID-19 Signs and Symptoms Through Day 28- mITT Population

Sustained alleviation of all targeted COVID-19 signs/symptoms was defined as the event occurring on the first 4 consecutive days when all symptoms scored as moderate or severe at the time of enrollment were scored as mild or absent and those scored mild or absent at the time of enrollment were scored as absent. The first day of the 4 consecutive-day period was considered date of first event. Time to sustained alleviation (event) was calculated as first event date minus first dose date plus 1, for participants with event. For participants who completed Day 28 or discontinued the study before Day 28 without sustained alleviation (censored), time was calculated as censoring date (last date on which symptom alleviation was assessed) minus first dose date plus 1 or Day 25 whichever occurred first. (NCT04960202)
Timeframe: From Day 1 (baseline) to Day 28

InterventionDays (Median)
PF-07321332 300 mg + Ritonavir 100 mg12.00
Placebo15.00

[back to top] [back to top]

Time to Sustained Resolution of Each Targeted COVID-19 Signs and Symptoms- mITT2 Population

Sustained resolution was defined as when each targeted symptom was scored as absent for 4 consecutive days. Time to sustained resolution (event) was calculated as first event date minus first dose date plus 1, for participants with event. For participants who completed Day 28 or discontinued the study before Day 28 without sustained resolution (censored), time was calculated as censoring date (last date on which symptom resolution was assessed) minus first dose date plus 1 or Day 25 whichever occurred first. (NCT04960202)
Timeframe: From Day 1 (baseline) to Day 28

,
InterventionDays (Median)
Muscle or body achesShortness of breath or difficulty breathingChills or shiveringCoughDiarrheaFeeling hot or feverishHeadacheNauseaStuffy or runny noseSore throatVomit
PF-07321332 300 mg + Ritonavir 100 mg9.0009.0005.00013.0006.0005.0009.0007.0009.0007.0003.000
Placebo12.00013.0006.00015.0006.0007.00011.0007.00011.0009.0004.000

[back to top]

Change From Baseline in Vital Signs Data - Supine Systolic Blood Pressure

Vital signs(systolic and diastolic blood pressure, and pulse rate) were measured with participants after having a rest for at least 5 minutes in a supine position. Vital signs assessment were performed after collection of ECGs and prior to collection of blood draws if scheduled at the same time. (NCT04962022)
Timeframe: Screening up to Day 9 of Period 2 or Early termination/discontinuation.

InterventionmmHg (Mean)
Period 1/Day 3 0 hour (H)Period 1/Day 3 1H 30min
PF-07321332 (Suspension)/Ritonavir 300/100 mg BID, Fasted1.41.5

[back to top]

Number of Participants With Clinically Significant 12-lead Electrocardiogram (ECG) Findings

Triplicate 12-lead ECG readings approximately 2 minutes apart were taken at each test. All ECG assessments were made after at least a 5-minute rest in a supine position and prior to any blood draws or vital sign measurements. (NCT04962022)
Timeframe: Screening up to Day 9 of Period 2 or Early termination/discontinuation.

,
InterventionParticipants (Count of Participants)
PR interval, Aggregate (msec) Value >= 300PR interval, Aggregate (msec) %Change >= 25/50%QRS Duration, Aggregate (msec) Value >= 140QRS Duration, Aggregate (msec) %Change >= 50%QTcF interval, Aggregate (msec) 450 < Value < = 480QTcF interval, Aggregate (msec) 480 < Value < = 500QTcF interval, Aggregate (msec) Value > 500QTcF interval, Aggregate (msec) 30 < Change < = 60QTcF interval, Aggregate (msec) Change > 60
Itraconazole 200 mg QD + PF-07321332(Suspension)/Ritonavir 300/100 mg BID, Fasted000000000
PF-07321332 (Suspension)/Ritonavir 300/100 mg BID, Fasted000000000

[back to top]

Number of Participants With Laboratory Abnormalities (Without Regard to Baseline Abnormality)

Safety laboratory assessments included urinalysis, hematology, chemistry and other. All the safety laboratory samples were collected following at least a 4-hour fast. (NCT04962022)
Timeframe: Screening up to Day 9 of Period 2 or Early termination/discontinuation.

,
InterventionParticipants (Count of Participants)
Monocytes/Leukocytes(%)> 1.2 ✖ ULNBicarbonate(mEq/L) > 1.1 ✖ ULNFibrinogen(mg/dl) > 1.25 ✖ BaselineLeukocyte Esterase >= 1
Itraconazole 200 mg QD + PF-07321332(Suspension)/Ritonavir 300/100 mg BID, Fasted2011
PF-07321332 (Suspension)/Ritonavir 300/100 mg BID, Fasted1600

[back to top]

Number of Participants With Treatment Emergent Adverse Events (TEAEs)

An Adverse event (AE) was any untoward medical occurrence in a participant. A serious AE was any untoward medical occurrence at any dose that resulted in death; was life-threatening; required hospitalization or prolongation of existing hospitalization; resulted in persistent or significant disability/incapacity; resulted in congenital anomaly/birth, was a suspected transmission via a Pfizer product of an infectious agent,pathogenic or non-pathogenic, was considered serious. The focus of AE summaries was on treatment-emergent AE (TEAE). An AE was considered TEAE if the event occurred during the on-treatment period. (NCT04962022)
Timeframe: Screening up to Day 35

,
InterventionParticipants (Count of Participants)
Participants with TEAEsParticipants with serious TEAEs
Itraconazole 200 mg QD + PF-07321332(Suspension)/Ritonavir 300/100 mg BID, Fasted100
PF-07321332 (Suspension)/Ritonavir 300/100 mg BID, Fasted70

[back to top]

Change From Baseline in Vital Signs Data - Supine Systolic Blood Pressure

Vital signs(systolic and diastolic blood pressure, and pulse rate) were measured with participants after having a rest for at least 5 minutes in a supine position. Vital signs assessment were performed after collection of ECGs and prior to collection of blood draws if scheduled at the same time. (NCT04962022)
Timeframe: Screening up to Day 9 of Period 2 or Early termination/discontinuation.

InterventionmmHg (Mean)
Period 2/Day 4 0HPeriod 2/Day6 0HPeriod 2/Day6 1HPeriod 2/Day6 1H 30minPeriod 2/Day6 72 H
Itraconazole 200 mg QD + PF-07321332(Suspension)/Ritonavir 300/100 mg BID, Fasted2.63.65.55.36.4

[back to top]

Apparent Clearance(CL/F) of PF-07321332

CL/F was apparent clearance. (NCT04962022)
Timeframe: Days 1, 2, 3 (pre-dose, 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours postdose on Day 3) in Period 1; Days 1, 4, 5, 6 (pre-dose, 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, 48, and 72 hours postdose on Day 6) of Period 2.

InterventionL/hr (Geometric Mean)
PF-07321332 (Suspension)/Ritonavir 300/100 mg BID, Fasted8.990
Itraconazole 200 mg QD + PF-07321332(Suspension)/Ritonavir 300/100 mg BID, Fasted6.478

[back to top]

Apparent Volume of Distribution (Vz/F) of PF-07321332

Vz/F was apparent volume of distribution. (NCT04962022)
Timeframe: Days 1, 2, 3 (pre-dose, 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours postdose on Day 3) in Period 1; Days 1, 4, 5, 6 (pre-dose, 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, 48, and 72 hours postdose on Day 6) of Period 2.

InterventionL (Geometric Mean)
PF-07321332 (Suspension)/Ritonavir 300/100 mg BID, Fasted104.7
Itraconazole 200 mg QD + PF-07321332(Suspension)/Ritonavir 300/100 mg BID, Fasted72.07

[back to top]

Area Under the Plasma Concentration-time Profile From Time Zero to the Time of the Last Quantifiable Concentration(AUClast) of PF-07321332

AUClast of PF-07321332 was determined by Linear/Log trapezoidal method. (NCT04962022)
Timeframe: Days 1, 2, 3 (pre-dose, 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours postdose on Day 3) in Period 1; Days 1, 4, 5, 6 (pre-dose, 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, 48, and 72 hours postdose on Day 6) of Period 2.

Interventionng*hr/mL (Geometric Mean)
PF-07321332 (Suspension)/Ritonavir 300/100 mg BID, Fasted41840
Itraconazole 200 mg QD + PF-07321332(Suspension)/Ritonavir 300/100 mg BID, Fasted74430

[back to top]

Area Under the Plasma Concentration-time Profile From Time Zero to Time Tau (τ), Where Tau=12-hour Dosing Interval(AUCtau) for PF-07321332

The AUCtau of PF-07321332 was determined by Linear/Log trapezoidal method. (NCT04962022)
Timeframe: Days 1, 2, 3 (pre-dose, 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours postdose on Day 3) in Period 1; Days 1, 4, 5, 6 (pre-dose, 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, 48, and 72 hours postdose on Day 6) of Period 2.

Interventionng*hr/mL (Geometric Mean)
PF-07321332 (Suspension)/Ritonavir 300/100 mg BID, Fasted33350
Itraconazole 200 mg QD + PF-07321332(Suspension)/Ritonavir 300/100 mg BID, Fasted46290

[back to top]

Maximum Observed Concentration (Cmax) of PF-07321332

The Cmax of PF-07321332 in the study was observed directly from data. (NCT04962022)
Timeframe: Days 1, 2, 3 (pre-dose, 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours postdose on Day 3) in Period 1; Days 1, 4, 5, 6 (pre-dose, 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, 48, and 72 hours postdose on Day 6) of Period 2.

Interventionng/mL (Geometric Mean)
PF-07321332 (Suspension)/Ritonavir 300/100 mg BID, Fasted4678
Itraconazole 200 mg QD + PF-07321332(Suspension)/Ritonavir 300/100 mg BID, Fasted5546

[back to top]

Terminal Half-life(t1/2) of PF-07321332

Terminal half-life was defined as the time measured for the plasma concentration of drug to decrease by one half. (NCT04962022)
Timeframe: Days 1, 2, 3 (pre-dose, 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours postdose on Day 3) in Period 1; Days 1, 4, 5, 6 (pre-dose, 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, 48, and 72 hours postdose on Day 6) of Period 2

Interventionhours(hr) (Mean)
PF-07321332 (Suspension)/Ritonavir 300/100 mg BID, Fasted8.255
Itraconazole 200 mg QD + PF-07321332(Suspension)/Ritonavir 300/100 mg BID, Fasted7.793

[back to top]

Time for Cmax (Tmax) for PF-07321332

PF-07321332 Tmax was observed directed from data (NCT04962022)
Timeframe: Days 1, 2, 3 (pre-dose, 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours postdose on Day 3) in Period 1; Days 1, 4, 5, 6 (pre-dose, 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, 48, and 72 hours postdose on Day 6) of Period 2

Interventionhours(hr) (Median)
PF-07321332 (Suspension)/Ritonavir 300/100 mg BID, Fasted1.020
Itraconazole 200 mg QD + PF-07321332(Suspension)/Ritonavir 300/100 mg BID, Fasted1.700

[back to top]

Change From Baseline in Vital Signs Data - Supine Diastolic Blood Pressure

Vital signs(systolic and diastolic blood pressure, and pulse rate) were measured with participants after having a rest for at least 5 minutes in a supine position. Vital signs assessment were performed after collection of ECGs and prior to collection of blood draws if scheduled at the same time. (NCT04962022)
Timeframe: Screening up to Day 9 of Period 2 or Early termination/discontinuation.

InterventionmmHg (Mean)
Period 1/Day 3 0HPeriod 1/Day 3 1H 30min
PF-07321332 (Suspension)/Ritonavir 300/100 mg BID, Fasted-0.90.5

[back to top]

Change From Baseline in Vital Signs Data - Supine Diastolic Blood Pressure

Vital signs(systolic and diastolic blood pressure, and pulse rate) were measured with participants after having a rest for at least 5 minutes in a supine position. Vital signs assessment were performed after collection of ECGs and prior to collection of blood draws if scheduled at the same time. (NCT04962022)
Timeframe: Screening up to Day 9 of Period 2 or Early termination/discontinuation.

InterventionmmHg (Mean)
Period 2/Day 4 0HPeriod 2/Day 6 0HPeriod 2/Day 6 1HPeriod 2/Day 6 1H 30minPeriod 2/Day 6 72H
Itraconazole 200 mg QD + PF-07321332(Suspension)/Ritonavir 300/100 mg BID, Fasted4.13.54.94.22.9

[back to top]

Change From Baseline in Vital Signs Data - Supine Pulse Rate

Vital signs(systolic and diastolic blood pressure, and pulse rate) were measured with participants after having a rest for at least 5 minutes in a supine position. Vital signs assessment were performed after collection of ECGs and prior to collection of blood draws if scheduled at the same time. (NCT04962022)
Timeframe: Screening up to Day 9 of Period 2 or Early termination/discontinuation.

Interventionbeats per minute(bpm) (Mean)
Period 1/Day 3 0 HPeriod 1/Day 3 1 H 30 min
PF-07321332 (Suspension)/Ritonavir 300/100 mg BID, Fasted 0/1-0.5-3.1

[back to top]

Change From Baseline in Vital Signs Data - Supine Pulse Rate

Vital signs(systolic and diastolic blood pressure, and pulse rate) were measured with participants after having a rest for at least 5 minutes in a supine position. Vital signs assessment were performed after collection of ECGs and prior to collection of blood draws if scheduled at the same time. (NCT04962022)
Timeframe: Screening up to Day 9 of Period 2 or Early termination/discontinuation.

Interventionbeats per minute(bpm) (Mean)
Period 2/Day 4 0 HPeriod 2/Day 6 0 HPeriod 2/Day 6 1 HPeriod 2/Day 6 1 H 30 minPeriod 2/Day 6 72 H
Itraconazole 200 mg QD + PF-07321332(Suspension)/Ritonavir 300/100 mg BID, Fasted-2.4-2.5-0.8-2.10.3

[back to top]

Vz/F of Ritonavir

Vz/F was defined as apparent volume of distribution. Vz/F is calculated as Dose/(AUCinf * kel), where kel is the terminal phase rate constant calculated by a linear regression of the log-linear concentration-time curve. (NCT04962230)
Timeframe: Day 1 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post dose in Period 1; Day 14 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post-dose or early termination/discontinuation in Period 2

InterventionL (Geometric Mean)
Period1: PF-07321332 300 mg/Ritonavir 100 mg234.0
Period2: Carbamazepine + PF-07321332 300 mg/Ritonavir 100 mg697.5

[back to top]

Number of Participants With Clinically Significant Change From Baseline in Vital Signs

Vital signs including single supine blood pressure and pulse rate were performed following at least a 5 minute rest in a supine position. Blood pressure (BP) and pulse rate (PR) assessments were performed after collection of electrocardiograms (ECGs) and prior to collection of blood draws if planned together. Respiratory rate (RR) was also evaluated. (NCT04962230)
Timeframe: Screening and Day 1 pre-dose, and at 1.5, 8, and 24 hours post dose in Period 1; Days 2, 3, 4, 6, 8, 10, 12, Day 14 pre-dose, and at 1.5, 8, 24, and 48 hours post-dose or early termination/discontinuation in Period 2

,
InterventionParticipants (Count of Participants)
Supine diastolic blood pressure value <50 mmHgSupine diastolic blood pressure change ≥20mmHg increaseSupine diastolic blood pressure change ≥20mmHg decreaseSupine pulse rate value <40 beats per minute(bpm)Supine pulse rate value >120 bpmSupine systolic blood pressure value <90mmHgSupine systolic blood pressure change ≥30mmHg decreaseSupine systolic blood pressure change ≥30mmHg increase
Period1: PF-07321332 300 mg/Ritonavir 100 mg00000100
Period2: Carbamazepine + PF-07321332 300 mg/Ritonavir 100 mg01000000

[back to top]

Tmax of Ritonavir

Tmax was defined as time to reach maximum observed plasma concentration and can be observed directly from data as time of first occurrence. (NCT04962230)
Timeframe: Day 1 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post dose in Period 1; Day 14 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post-dose or early termination/discontinuation in Period 2

Interventionhour (hr) (Median)
Period1: PF-07321332 300 mg/Ritonavir 100 mg3.98
Period2: Carbamazepine + PF-07321332 300 mg/Ritonavir 100 mg1.98

[back to top]

Terminal Half-Life ( t1/2) of PF-07321332

t1/2 was defined as the time measured for the plasma concentration of drug to decrease by one half. t1/2 is calculated as Loge(2)/kel, where kel is the terminal phase rate constant calculated by a linear regression of the log-linear concentration-time curve. Only those data points judged to describe the terminal log-linear decline was used in the regression. (NCT04962230)
Timeframe: Day 1 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post dose in Period 1; Day 14 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post-dose or early termination/discontinuation in Period 2

Interventionhours (hr) (Mean)
Period1: PF-07321332 300 mg/Ritonavir 100 mg6.053
Period2: Carbamazepine + PF-07321332 300 mg/Ritonavir 100 mg3.845

[back to top]

Number of Participants With Treatment-Emergent Adverse Events (TEAEs)

An Adverse event (AE) was any untoward medical occurrence in a participant. A serious AE was any untoward medical occurrence at any dose that resulted in death; was life-threatening; required hospitalization or prolongation of existing hospitalization; resulted in persistent or significant disability/incapacity, congenital anomaly/birth. An AE was considered treatment-emergent AE (TEAE) if the event occurred during the on-treatment period. A treatment-related AE was any untoward medical occurrence attributed to the study drug in a participant who received study drug. A treatment-related SAE was a treatment-related AE and was defined as any untoward medical occurrence at any dose that: was fatal or life-threatening; resulted in persistent or significant disability/incapacity; constituted a congenital anomaly/birth defect; was medically significant; required inpatient hospitalization or prolongation of existing hospitalization. Relatedness to study drug was assessed by the investigator. (NCT04962230)
Timeframe: Screening up to Day 3 in Period 1; Day 1 up to 35 days from administration of the final dose of study intervention or early termination/discontinuation in Period 2 (maximum of approximately 12 weeks)

,
InterventionParticipants (Count of Participants)
Participants with all-causality adverse eventsParticipants with all-causality serious adverse eventsParticipants with treatment related adverse eventsParticipants with treatment related serious adverse events
Period1: PF-07321332 300 mg/Ritonavir 100 mg4010
Period2: Carbamazepine + PF-07321332 300 mg/Ritonavir 100 mg9060

[back to top]

Number of Participants With Laboratory Abnormalities

Safety laboratory assessments included hematology, urinalysis, and clinical chemistry. All the safety laboratory samples were collected following at least a 4 hour fast. (NCT04962230)
Timeframe: Screening, Day -1 prior to dosing, and Day 2 in Period 1; Days 3, 6, 9, 12, 16 or early termination/discontinuation in Period 2

,
InterventionParticipants (Count of Participants)
Hemoglobin (g/dL) <0.8x lower limit of normal (LLN)Hematocrit (%) <0.8x LLNErythrocytes (10^6/mm^3) <0.8x LLNLymphocytes (10^3/mm^3) <0.8x LLNNeutrophils (10^3/mm^3) <0.8x LLNEosinophils/Leukocytes (%) >1.2x upper limit of normal (ULN)Monocytes/Leukocytes (%) >1.2x ULNAlanine Aminotransferase (U/L) >3.0x ULNSodium (milliequivalent per liter (mEq/L)) <0.95x LLNUrobilinogen (EU/dL) ≥1Fibrinogen (mg/dL) >1.25x BaselineURINE Hemoglobin ≥1
Period1: PF-07321332 300 mg/Ritonavir 100 mg000001100000
Period2: Carbamazepine + PF-07321332 300 mg/Ritonavir 100 mg111214541113

[back to top]

Apparent Oral Clearance (CL/F) of PF-07321332

CL/F was defined as apparent clearance. Clearance of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. Clearance obtained after oral dose (apparent oral clearance) is a quantitative measure of the rate at which a drug substance is removed from the blood. CL/F is calculated as dose/AUCinf. AUCinf = area under the plasma concentration versus time curve from time zero extrapolated to infinite time. (NCT04962230)
Timeframe: Day 1 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post dose in Period 1; Day 14 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post-dose or early termination/discontinuation in Period 2

InterventionLiter per hour (L/hr) (Geometric Mean)
Period1: PF-07321332 300 mg/Ritonavir 100 mg13.06
Period2: Carbamazepine + PF-07321332 300 mg/Ritonavir 100 mg29.17

[back to top]

Number of Participants With Clinically Significant Change in Electrocardiogram (ECG) Findings

All ECG assessments (triplicate) were made after at least a 10 minute rest in a supine position and prior to any blood draws or vital sign measurements. (NCT04962230)
Timeframe: Screening and Day 1 pre-dose, and at 1.5, 8, and 24 hours post dose in Period 1; Days 2, 3, 4, Day 14 pre-dose, and at 1.5, 8, 24, and 48 hours post-dose or early termination/discontinuation in Period 2

,
InterventionParticipants (Count of Participants)
PR interval, aggregate value ≥300 milliseconds (msec)PR interval, aggregate percent change ≥25/50%QRS duration, aggregate value ≥140 msecQRS duration, aggregate percent change ≥50%QTCF interval, aggregate value >450 msec and ≤480 msecQTCF interval, aggregate value >480 msec and ≤500 msecQTCF interval, aggregate value >500 msecQTCF interval, aggregate change ≥30 msec and ≤60 msecQTCF interval, aggregate change >60 msec
Period1: PF-07321332 300 mg/Ritonavir 100 mg000000000
Period2: Carbamazepine + PF-07321332 300 mg/Ritonavir 100 mg000000000

[back to top]

Apparent Volume of Distribution (Vz/F) of PF-07321332

Vz/F was defined as apparent volume of distribution. Vz/F is calculated as Dose/(AUCinf * kel), where kel is the terminal phase rate constant calculated by a linear regression of the log-linear concentration-time curve. (NCT04962230)
Timeframe: Day 1 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post dose in Period 1; Day 14 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post-dose or early termination/discontinuation in Period 2

InterventionLiter (L) (Geometric Mean)
Period1: PF-07321332 300 mg/Ritonavir 100 mg109.4
Period2: Carbamazepine + PF-07321332 300 mg/Ritonavir 100 mg157.2

[back to top]

Time to Reach Maximum Observed Plasma Concentration (Tmax) of PF-07321332

Tmax was defined as time to reach maximum observed plasma concentration and can be observed directly from data as time of first occurrence. (NCT04962230)
Timeframe: Day 1 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post dose in Period 1; Day 14 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post-dose or early termination/discontinuation in Period 2

Interventionhour (hr) (Median)
Period1: PF-07321332 300 mg/Ritonavir 100 mg3.00
Period2: Carbamazepine + PF-07321332 300 mg/Ritonavir 100 mg1.50

[back to top]

Area Under the Curve From Time Zero to Extrapolated Infinite Time (AUCinf) of PF-07321332

AUCinf was defined as area under the concentration-time curve from time 0 to infinity and was calculated as AUClast + (Clast*/kel), where Clast* is the predicted plasma concentration at the last quantifiable time point estimated from the log-linear regression analysis. (NCT04962230)
Timeframe: Day 1 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post dose in Period 1; Day 14 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post-dose or early termination/discontinuation in Period 2

Interventionnanogram*hour per milliliter (ng*hr/mL) (Geometric Mean)
Period1: PF-07321332 300 mg/Ritonavir 100 mg23010
Period2: Carbamazepine + PF-07321332 300 mg/Ritonavir 100 mg10280

[back to top]

Area Under the Curve From Time Zero to Last Quantifiable Concentration (AUClast) of PF-07321332

AUClast was defined as area under the plasma concentration-time profile from time 0 to the time of last observed quantifiable plasma concentration (Clast) and was determined by Linear/Log trapezoidal method. (NCT04962230)
Timeframe: Day 1 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post dose in Period 1; Day 14 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post-dose or early termination/discontinuation in Period 2

Interventionng*hr/mL (Geometric Mean)
Period1: PF-07321332 300 mg/Ritonavir 100 mg22450
Period2: Carbamazepine + PF-07321332 300 mg/Ritonavir 100 mg10050

[back to top]

AUCinf of Ritonavir

AUCinf was defined as area under the concentration-time curve from time 0 to infinity and was calculated as AUClast + (Clast*/kel), where Clast* is the predicted plasma concentration at the last quantifiable time point estimated from the log-linear regression analysis. (NCT04962230)
Timeframe: Day 1 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post dose in Period 1; Day 14 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post-dose or early termination/discontinuation in Period 2

Interventionng*hr/mL (Geometric Mean)
Period1: PF-07321332 300 mg/Ritonavir 100 mg3599
Period2: Carbamazepine + PF-07321332 300 mg/Ritonavir 100 mg677.6

[back to top]

AUClast of Ritonavir

AUClast was defined as area under the plasma concentration-time profile from time 0 to the time of Clast and was determined by Linear/Log trapezoidal method. (NCT04962230)
Timeframe: Day 1 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post dose in Period 1; Day 14 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post-dose or early termination/discontinuation in Period 2

Interventionng*hr/mL (Geometric Mean)
Period1: PF-07321332 300 mg/Ritonavir 100 mg3414
Period2: Carbamazepine + PF-07321332 300 mg/Ritonavir 100 mg466.2

[back to top]

CL/F of Ritonavir

CL/F was defined as apparent clearance. Clearance of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. Clearance obtained after oral dose (apparent oral clearance) is a quantitative measure of the rate at which a drug substance is removed from the blood. CL/F is calculated as dose/AUCinf. AUCinf = area under the plasma concentration versus time curve from time zero extrapolated to infinite time. (NCT04962230)
Timeframe: Day 1 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post dose in Period 1; Day 14 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post-dose or early termination/discontinuation in Period 2

InterventionL/hr (Geometric Mean)
Period1: PF-07321332 300 mg/Ritonavir 100 mg27.78
Period2: Carbamazepine + PF-07321332 300 mg/Ritonavir 100 mg147.6

[back to top]

Cmax of Ritonavir

Cmax was defined as maximum observed plasma concentration and can be observed directly from data. (NCT04962230)
Timeframe: Day 1 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post dose in Period 1; Day 14 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post-dose or early termination/discontinuation in Period 2

Interventionng/mL (Geometric Mean)
Period1: PF-07321332 300 mg/Ritonavir 100 mg359.3
Period2: Carbamazepine + PF-07321332 300 mg/Ritonavir 100 mg96.07

[back to top]

Maximum Observed Plasma Concentration (Cmax) of PF-07321332

Cmax was defined as maximum observed plasma concentration and can be observed directly from data. (NCT04962230)
Timeframe: Day 1 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post dose in Period 1; Day 14 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post-dose or early termination/discontinuation in Period 2

Interventionnanogram per milliliter (ng/mL) (Geometric Mean)
Period1: PF-07321332 300 mg/Ritonavir 100 mg2210
Period2: Carbamazepine + PF-07321332 300 mg/Ritonavir 100 mg1300

[back to top]

t1/2 of Ritonavir

t1/2 was defined as the time measured for the plasma concentration of drug to decrease by one half. t1/2 is calculated as Loge(2)/kel, where kel is the terminal phase rate constant calculated by a linear regression of the log-linear concentration-time curve. Only those data points judged to describe the terminal log-linear decline was used in the regression. (NCT04962230)
Timeframe: Day 1 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post dose in Period 1; Day 14 pre-dose, and at 0.5, 1, 1.5, 2, 4, 6, 8, 12, 16, 24, and 48 hours post-dose or early termination/discontinuation in Period 2

Interventionhr (Mean)
Period1: PF-07321332 300 mg/Ritonavir 100 mg6.149
Period2: Carbamazepine + PF-07321332 300 mg/Ritonavir 100 mg3.345

[back to top]

Number of Participants With Abnormal Laboratory Assessments (Without Regard to Baseline Abnormality)

Following laboratory parameters were assessed against pre-defined abnormality criteria: hematology; clinical chemistry; urinalysis. (NCT05005312)
Timeframe: Up to 2 months

,
InterventionParticipants (Count of Participants)
HEMATOLOGY: Platelets (10^3/mm^3) <0.5× lower limit of normal (LLN)HEMATOLOGY: Eosinophils (10^3/mm^3) >1.2× upper limit of normal (ULN)HEMATOLOGY: Monocytes (10^3/mm^3) >1.2× ULNCLINICAL CHEMISTRY: Direct Bilirubin (mg/dL) >1.5× ULNCLINICAL CHEMISTRY: Aspartate Aminotransferase (U/L) >3.0× ULNCLINICAL CHEMISTRY: Calcium (mg/dL) >1.1× ULNCLINICAL CHEMISTRY: Glucose (mg/dL) >1.5× ULNURINALYSIS: URINE Hemoglobin ≥1URINALYSIS: Urobilinogen ≥1URINALYSIS: Leukocyte Esterase ≥1URINALYSIS: Urine Microscopic Exam ≥1
Moderate Hepatic Impairment12111010110
Normal Hepatic Function01000101001

[back to top]

Area Under the Plasma Concentration-Time Profile From Time Zero Extrapolated to Infinite Time (AUCinf) of Plasma PF-07321332

AUCinf was defined as area under the plasma concentration-time curve from time zero to infinity. The geometric coefficient of variation was expressed in percentage. (NCT05005312)
Timeframe: Day 1 at 0 (pre-dose for PF-07321332), 0.5, 1, 2, 3, 4, 6, 8, 10, and 12 hours, Day 2 at 24 and 36 hours, and Day 3 at 48 hours

Interventionug*hr/mL (Geometric Mean)
Normal Hepatic Function15.24
Moderate Hepatic Impairment15.06

[back to top]

Maximum Observed Plasma Concentration (Cmax) of Plasma PF-07321332

Cmax was the maximum observed plasma concentration and was directly observed from data. Concentration values below the lower limit of quantification (LLQ) were set to zero. Geometric mean analysis was on the log scale. Zero values were not included in geometric mean and geometric coefficient of variation calculation. The geometric coefficient of variation was expressed in percentage. (NCT05005312)
Timeframe: Day 1 at 0 (pre-dose for PF-07321332), 0.5, 1, 2, 3, 4, 6, 8, 10, and 12 hours, Day 2 at 24 and 36 hours, and Day 3 at 48 hours

Interventionmicrograms per milliliter (ug/mL) (Geometric Mean)
Normal Hepatic Function1.886
Moderate Hepatic Impairment1.923

[back to top]

Number of Participants With Abnormal Electrocardiograms (ECGs)

"ECG categorical summarization criteria: 1. PR interval (the interval between the start of the P wave and the start of the QRS complex, corresponding to the time between the onset of the atrial depolarization and onset of ventricular depolarization): a) greater than or equal to (>=) 300 millisecond (msec), b) >=25% increase when baseline is > 200 msec or >=50% increase when baseline is less than or equal to (<=) 200 msec.~2. QRS duration (time from ECG Q wave to the end of the S wave corresponding to ventricle depolarization): a) >=140 msec, b) >=50% increase from baseline.~3. QTcF interval (QT corrected using the Fridericia formula): a) >450 msec and <=480 msec, b) >480 msec and <=500 msec, c) >500 msec, d) >30 msec and <=60 msec increase from baseline, e) >60 msec increase from baseline." (NCT05005312)
Timeframe: Up to 2 months

,
InterventionParticipants (Count of Participants)
PR INTERVAL NOT OTHERWISE SPECIFIED (MSEC): Value ≥ 300PR INTERVAL NOT OTHERWISE SPECIFIED (MSEC): %Change ≥ 25/50%QRS INTERVAL NOT OTHERWISE SPECIFIED (MSEC): Value ≥ 140QRS INTERVAL NOT OTHERWISE SPECIFIED (MSEC): %Change ≥ 50%QT INTERVAL NOT OTHERWISE SPECIFIED (MSEC): Value > 500QTC INTERVAL NOT OTHERWISE SPECIFIED (MSEC): 450 < Value ≤ 480QTC INTERVAL NOT OTHERWISE SPECIFIED (MSEC): 480 < Value ≤ 500QTC INTERVAL NOT OTHERWISE SPECIFIED (MSEC): Value > 500QTC INTERVAL NOT OTHERWISE SPECIFIED (MSEC): 30 < Change ≤ 60QTC INTERVAL NOT OTHERWISE SPECIFIED (MSEC): Change > 60QTCF - FRIDERICIA'S CORRECTION FORMULA NOT OTHERWISE SPECIFIED (MSEC): 450 < Value ≤ 480QTCF - FRIDERICIA'S CORRECTION FORMULA NOT OTHERWISE SPECIFIED (MSEC): 480 < Value ≤ 500QTCF - FRIDERICIA'S CORRECTION FORMULA NOT OTHERWISE SPECIFIED (MSEC): Value > 500QTCF - FRIDERICIA'S CORRECTION FORMULA NOT OTHERWISE SPECIFIED (MSEC): 30 < Change ≤ 60QTCF - FRIDERICIA'S CORRECTION FORMULA NOT OTHERWISE SPECIFIED (MSEC): Change > 60
Moderate Hepatic Impairment000000000000000
Normal Hepatic Function000000000000000

[back to top]

Area Under the Plasma Concentration-Time Profile From Time Zero to Time of the Last Quantifiable Concentration (AUClast) of Plasma PF-07321332

AUClast was area under the plasma concentration time-curve from zero (pre-dose) to the last measured concentration. The geometric coefficient of variation was expressed in percentage. (NCT05005312)
Timeframe: Day 1 at 0 (pre-dose for PF-07321332), 0.5, 1, 2, 3, 4, 6, 8, 10, and 12 hours, Day 2 at 24 and 36 hours, and Day 3 at 48 hours

Interventionmicrograms/milliliter/hour (ug*hr/mL) (Geometric Mean)
Normal Hepatic Function14.97
Moderate Hepatic Impairment14.86

[back to top]

Number of Participants With an Treatment Emergent Adverse Event (TEAE)

An AE was any untoward medical occurrence in a participant who received study drug without regard to possibility of causal relationship. Treatment-emergent adverse event (TEAE) means event between first dose of study treatment and up to 30 days after last dose that were absent before treatment or that worsened relative to pretreatment state. An SAE was an AE resulting in any of death; inpatient hospitalization; life-threatening experience; disability; congenital anomaly or deemed significant for any other reason. (NCT05005312)
Timeframe: Up to 2 months

,
InterventionParticipants (Count of Participants)
Participants with adverse events (All Causalities)Participants with adverse events (Treatment Related)
Moderate Hepatic Impairment43
Normal Hepatic Function30

[back to top]

Number of Participants With Abnormal Vital Signs

Criteria for vital signs abnormalities: increase or decrease from baseline in systolic blood pressure (SBP) >=30 mm Hg and increase or decrease from baseline in diastolic blood pressure (DBP) >=20 mm Hg; the value of SBP <90 mm Hg; the value of DBP <50 mm Hg; the value of pulse rate <40 bpm or >120 bpm. (NCT05005312)
Timeframe: Up to 2 months

,
InterventionParticipants (Count of Participants)
SBP: Value <90mmHgSBP: Change >= 30 mmHg increase from baselineSBP: Change >= 30 mmHg decrease from baselineDBP: Value <50 mmHgDBP: Change >= 20 mmHg increase from baselineDBP: Change >= 20 mmHg decrease from baselinePulse Rate: Value <40 bpmPulse Rate: Value >120 bpm
Moderate Hepatic Impairment01000000
Normal Hepatic Function00000000

[back to top]

Number of Participants With Treatment Emergent Adverse Events (TEAEs), Serious AEs and AEs Leading to Study and Study Drug Discontinuation

An AE was defined as any untoward medical occurrence in a participant or clinical study participant temporally associated with the use of study intervention, whether or not considered related to the study intervention. An SAE was defined as any untoward medical occurrence at any dose that resulted in any of the following outcomes: death; life-threatening ; required inpatient hospitalization or prolongation of existing hospitalization; persistent or significant disability/incapacity ; congenital anomaly/birth defect; or that was considered as an important medical event. TEAEs were defined as events that started on or after the study medication start date and time. AEs included both serious and all non-serious adverse events. AEs that led to study discontinuation and AEs that led to discontinuation of study intervention and then continued study were also reported in this outcome measure. (NCT05011513)
Timeframe: From start of study intervention (Day 1) up to Day 34

,
InterventionParticipants (Count of Participants)
TEAEsSAEsAEs led to discontinuation of studyAEs led to discontinue study intervention and continued study
Nirmatrelvir 300 mg + Ritonavir 100 mg1698016
Placebo1531315

[back to top]

Duration of Hospitalization and Intensive Care Unit (ICU) Stay Through Day 28

(NCT05011513)
Timeframe: From Day 1 to Day 28

,
InterventionDays (Mean)
HospitalizationICU
Nirmatrelvir 300 mg + Ritonavir 100 mg0.0490.000
Placebo0.1810.065

[back to top]

Change From Baseline in Logarithm to Base10 (Log10) Transformed Viral Load at Days 3, 5, 10 and 14

Nasal samples were collected to estimate the viral load in participants in terms of logarithm to base 10 (log10) copies per milliliter. (NCT05011513)
Timeframe: Baseline, Days 3, 5, 10 and 14

,
InterventionLog 10 copies per milliliter (Least Squares Mean)
Day 3Day 5Day 10Day 14
Nirmatrelvir 300 mg + Ritonavir 100 mg-2.302-3.669-4.873-5.464
Placebo-1.565-2.835-4.642-5.249

[back to top]

Time to Sustained Resolution of Overall COVID-19 Signs and Symptoms Through Day 28

Sustained resolution was defined as when targeted symptoms are scored as absent for 4 consecutive days. The first day of the 4 consecutive-day period was considered the first event date. (NCT05011513)
Timeframe: From Day 1 to Day 28

InterventionDays (Median)
Nirmatrelvir 300 mg + Ritonavir 100 mg15.000
Placebo16.000

[back to top] [back to top]

Percentage of Participants With Resting Peripheral Oxygen Saturation Greater Than or Equal to (>=) 95% at Day 1 and Day 5

Percentage of participants with a resting peripheral oxygen saturation >=95% were reported in this outcome measure. (NCT05011513)
Timeframe: Day 1 and Day 5

,
InterventionPercentage of participants (Number)
Day 1Day 5
Nirmatrelvir 300 mg + Ritonavir 100 mg62.50094.671
Placebo67.85793.212

[back to top]

Plasma Concentration Versus Time Summary of PF-07321332

(NCT05011513)
Timeframe: Day 1: 1 hour post dose; Day 5: 0 minutes pre-dose

InterventionNanograms per milliliter (Mean)
Day 1 (1 hour post-dose)Day 5 (0 minutes pre-dose)
Nirmatrelvir 300 mg + Ritonavir 100 mg24373468

[back to top]

Time to Sustained Alleviation of Each COVID-19 Signs and Symptoms Through Day 28

Sustained alleviation of targeted COVID-19 signs/symptoms was defined as the event occurring on the first 4 consecutive days when all symptoms scored as moderate or severe at the time of enrollment were scored as mild or absent and those scored mild or absent at the time of enrollment were scored as absent. Missing severity at baseline was treated as mild. Time to sustained alleviation of all targeted COVID-19 signs and symptoms through Day 28, was calculated as time (days) from start of study intervention or placebo (Day 1) until sustained alleviation of all targeted COVID-19 associated signs and symptoms. In this outcome measure time to sustained alleviation is reported for each COVID-19 signs and symptoms. (NCT05011513)
Timeframe: From Day 1 to Day 28

,
InterventionDays (Median)
Muscle or body achesShortness of breath or difficulty breathingChills or shiveringCoughDiarrheaFeeling hot or feverishHeadacheNauseaStuffy or runny noseSore throatVomit
Nirmatrelvir 300 mg + Ritonavir 100 mg5.0005.0003.0007.0006.0003.0005.0004.0005.0004.0003.000
Placebo5.0006.0003.0008.0004.0004.0005.0004.0007.0005.0003.000

[back to top]

Time to Sustained Resolution of Each COVID-19 Signs and Symptoms Through Day 28

Sustained resolution was defined as when targeted symptoms are scored as absent for 4 consecutive days. The first day of the 4 consecutive-day period was considered the first event date. In this outcome measure time to sustained resolution is reported consolidated for each COVID-19 signs and symptoms. (NCT05011513)
Timeframe: From Day 1 to Day 28

,
InterventionDays (Median)
Muscle or body achesShortness of breath or difficulty breathingChills or shiveringCoughDiarrheaFeeling hot or feverishHeadacheNauseaStuffy or runny noseSore throatVomit
Nirmatrelvir 300 mg + Ritonavir 100 mg8.0007.0004.00011.0006.0004.0007.0006.0009.0006.0003.000
Placebo9.0008.0005.00012.0005.0005.0009.0005.00010.0008.0003.000

[back to top] [back to top]

Percentage of Participants With Death Through Week 24

Percentage of participants with death (all-cause) event were reported in this outcome measure. (NCT05011513)
Timeframe: From Day 1 to Week 24

InterventionPercentage of participants (Number)
Nirmatrelvir 300 mg + Ritonavir 100 mg0
Placebo0.2

[back to top]

Percentage of Participants With Progression to Worsening Status of COVID-19 Signs and Symptoms

Participants recorded a daily severity rating of their symptom severity over the past 24 hours based on a 4-point scale in which 0 was reported if no symptoms were present; 1 if mild; 2 if moderate; and 3 if severe. Vomiting and diarrhea was rated on a 4-point frequency scale where 0 is reported for no occurrence, 1 (mild) for 1 to 2 times, 2 (moderate) for 3 to 4 times, and 3 (severe) for 5 or greater. Progression to a worsening status for any targeted symptom was based up on increasing severity (i.e. the first time any targeted symptoms worsened after treatment relative to baseline). (NCT05011513)
Timeframe: From Day 1 to Day 28

InterventionPercentage of participants (Number)
Nirmatrelvir 300 mg + Ritonavir 100 mg75.463
Placebo78.515

[back to top]

Percentage of Participants With Severe Signs and Symptoms of COVID-19 Through Day 28

Participants recorded a daily severity rating of their symptom severity over the past 24 hours based on a 4-point scale in which 0 was reported if no symptoms were present; 1 if mild; 2 if moderate; and 3 if severe. A participant with severe score for any targeted symptoms post-baseline was counted as severe. Vomiting and diarrhea each was rated on a 4-point frequency scale where 0 was reported for no occurrence, 1 for 1 to 2 times, 2 for 3 to 4 times, and 3 for 5 or greater. Sense of smell and sense of taste each be rated on a 3-point Likert scale where 0 was reported if the sense of smell/taste was the same as usual, 1 if the sense of smell/taste was less than usual, and 2 for no sense of smell/taste. (NCT05011513)
Timeframe: From Day 1 to Day 28

InterventionPercentage of participants (Number)
Nirmatrelvir 300 mg + Ritonavir 100 mg19.136
Placebo21.643

[back to top]

Time to Sustained Alleviation of Overall COVID-19 Signs and Symptoms Through Day 28

Sustained alleviation of targeted COVID-19 signs/symptoms was defined as the event occurring on the first 4 consecutive days when all symptoms scored as moderate or severe at the time of enrollment were scored as mild or absent and those scored mild or absent at the time of enrollment were scored as absent. Missing severity at baseline was considered as mild. Time to sustained alleviation of all targeted COVID-19 signs and symptoms through Day 28, was calculated as time (days) from start of study intervention or placebo (Day 1) until sustained alleviation of all targeted COVID-19 associated signs and symptoms. In this outcome measure time to sustained alleviation is reported consolidated for overall COVID-19 signs and symptoms. (NCT05011513)
Timeframe: From Day 1 to Day 28

InterventionDays (Median)
Nirmatrelvir 300 mg + Ritonavir 100 mg12.000
Placebo13.000

[back to top]

Number of Participants With Laboratory Abnormalities

The haematological, clinical chemistry (serum) and urinalysis safety tests were assessed against the criteria specified in the sponsor reporting standards to determine if there were any clinically significant laboratory abnormalities. The assessment took into account whether each participant's baseline test result was within or outside the laboratory reference range for the particular laboratory parameter. Baseline was defined as the last planned predose measurement taken in each study period. (NCT05032950)
Timeframe: Baseline up to Day 28

,,
InterventionParticipants (Count of Participants)
HEMATOLOGY - Lymphocytes/Leukocytes (%) >1.2x ULNHEMATOLOGY - Neutrophils (10^3/mm^3) <0.8x LLNHEMATOLOGY - Neutrophils/Leukocytes (%) <0.8x LLNHEMATOLOGY - Eosinophils/Leukocytes (%) >1.2x ULNHEMATOLOGY - Monocytes/Leukocytes (%) >1.2x ULNHEMATOLOGY - Prothrombin Time (sec) >1.1x ULNCLINICAL CHEMISTRY - Thyrotropin (uIU/mL) <0.8x LLNCLINICAL CHEMISTRY - Fibrinogen (mg/dL) >1.25x BaselineURINALYSIS - URINE Hemoglobin (Scalar) ≥1
Midazolam 2 mg000010112
PF-07321332 300 mg/Ritonavir 100 mg + Midazolam 2 mg001110111
Ritonavir 100 mg + Midazolam 2 mg111121103

[back to top]

Apparent Volume of Distribution (Vz/F) of Midazolam When Administered Alone, With PF-07321332/Ritonavir, and With Ritonavir

Vz/F for midazolam following single dose administration with and without PF-07321332/ritonavir or ritonavir was calculated by Dose/(AUCinf • kel). (NCT05032950)
Timeframe: Midazolam: Day 1 Predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36 hours postdose; Midazolam+PF-07321332/ritonavir: Day 5 predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36, 48 and 72 hours postdose

InterventionLitre (Geometric Mean)
Midazolam 2 mg488.6
PF-07321332 300 mg/Ritonavir 100 mg + Midazolam 2 mg79.84
Ritonavir 100mg+ Midazolam 2mg76.43

[back to top]

Area Under the Plasma Concentration-time Profile From Time 0 to the Time of the Last Quantifiable Concentration (Clast) (AUClast) of Midazolam When Administered Alone and With PF-07321332/Ritonavir

AUClast for midazolam following single dose administration with and without PF-07321332/ritonavir was calculated by Linear/Log trapezoidal method. Natural log-transformed AUClast for Midazolam were analyzed using a mixed effect model with sequence, period and treatment as fixed effects and participant within sequence as a random effect. The ratios (PF-07321332/ritonavir + midazolam [test]/midazolam [reference] and 90% CIs) were expressed as percentages. (NCT05032950)
Timeframe: Midazolam: Day 1 Predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36 hours postdose; Midazolam+PF-07321332/ritonavir: Day 5 predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36, 48 and 72 hours postdose

Interventionng*hr/mL (Geometric Mean)
Midazolam 2 mg25.02
PF-07321332 300 mg/Ritonavir 100 mg + Midazolam 2 mg353.8

[back to top]

Apparent Clearance (CL/F) of Midazolam When Administered Alone, With PF-07321332/Ritonavir, and With Ritonavir

CL/F for midazolam following single dose administration with and without PF-07321332/ritonavir or ritonavir was calculated by Dose/AUCinf. (NCT05032950)
Timeframe: Midazolam: Day 1 Predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36 hours postdose; Midazolam+PF-07321332/ritonavir: Day 5 predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36, 48 and 72 hours postdose

InterventionLitre/hour (Geometric Mean)
Midazolam 2 mg76.57
PF-07321332 300 mg/Ritonavir 100 mg + Midazolam 2 mg5.500
Ritonavir 100mg+ Midazolam 2mg4.776

[back to top]

Area Under the Plasma Concentration-time Profile From Time 0 Extrapolated to Infinity Time (AUCinf) of Midazolam When Administered Alone and With PF-07321332/Ritonavir

AUCinf for midazolam following single dose administration with and without PF-07321332/ritonavir was calculated by AUClast + (Clast/kel), where Clast was the predicted plasma concentration at the last quantifiable time point estimated from the log-linear regression analysis. Natural log-transformed AUCinf for Midazolam were analyzed using a mixed effect model with sequence, period and treatment as fixed effects and participant within sequence as a random effect. The ratios (PF-07321332/ritonavir + midazolam [test]/midazolam [reference] and 90% CIs) were expressed as percentages. (NCT05032950)
Timeframe: Midazolam: Day 1 Predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36 hours postdose; Midazolam+PF-07321332/ritonavir: Day 5 predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36, 48 and 72 hours postdose

Interventionng*hr/mL (Geometric Mean)
Midazolam 2 mg26.13
PF-07321332 300 mg/Ritonavir 100 mg + Midazolam 2 mg363.9

[back to top]

AUCinf of Midazolam When Administered Alone and With Ritonavir

AUCinf for midazolam following single dose administration with and without ritonavir was calculated by AUClast + (Clast/kel), where Clast was the predicted plasma concentration at the last quantifiable time point estimated from the log-linear regression analysis. Natural log-transformed AUCinf for Midazolam were analyzed using a mixed effect model with sequence, period and treatment as fixed effects and participant within sequence as a random effect. The ratios (ritonavir + midazolam [test]/midazolam [reference] and 90% CIs) were expressed as percentages. (NCT05032950)
Timeframe: Midazolam: Day 1 Predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36 hours postdose; Midazolam/ritonavir: Day 5 predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36, 48 and 72 hours postdose

Interventionng*hr/mL (Geometric Mean)
Midazolam 2 mg26.13
Ritonavir 100 mg + Midazolam 2 mg418.6

[back to top]

AUClast of Midazolam When Administered Alone and With Ritonavir

AUClast for midazolam following single dose administration with and without ritonavir was calculated by Linear/Log trapezoidal method. Natural log-transformed AUClast for Midazolam were analyzed using a mixed effect model with sequence, period and treatment as fixed effects and participant within sequence as a random effect. The ratios (ritonavir + midazolam [test]/midazolam [reference] and 90% CIs) were expressed as percentages. (NCT05032950)
Timeframe: Midazolam: Day 1 Predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36 hours postdose; Midazolam/ritonavir: Day 5 pre-dose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36, 48 and 72 hours post-dose

Interventionng*hr/mL (Geometric Mean)
Midazolam 2 mg25.02
Ritonavir 100 mg + Midazolam 2 mg408.8

[back to top]

Cmax of Midazolam When Administered Alone and With Ritonavir

Cmax for midazolam following single dose administration with and without ritonavir was observed directly form data. Natural log-transformed Cmax for Midazolam were analyzed using a mixed effect model with sequence, period and treatment as fixed effects and participant within sequence as a random effect. The ratios (ritonavir + midazolam [test]/midazolam [reference] and 90% CIs) were expressed as percentages. (NCT05032950)
Timeframe: Midazolam: Day 1 Predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36 hours postdose; Midazolam/ritonavir: Day 5 predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36, 48 and 72 hours postdose

Interventionng/mL (Geometric Mean)
Midazolam 2 mg9.812
Ritonavir 100 mg + Midazolam 2 mg38.03

[back to top]

Maximum Plasma Concentration (Cmax) of Midazolam When Administered Alone and With PF-07321332/Ritonavir

Cmax for midazolam following single dose administration with and without PF-07321332/ritonavir was observed directly from data. Natural log-transformed Cmax for Midazolam were analyzed using a mixed effect model with sequence, period and treatment as fixed effects and participant within sequence as a random effect. The ratios (PF-07321332/ritonavir + midazolam [test]/midazolam [reference] and 90% CIs) were expressed as percentages. (NCT05032950)
Timeframe: Midazolam: Day 1 Predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36 hours postdose; Midazolam+PF-07321332/ritonavir: Day 5 predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36, 48 and 72 hours postdose

Interventionng/mL (Geometric Mean)
Midazolam 2 mg9.812
PF-07321332 300 mg/Ritonavir 100 mg + Midazolam 2 mg36.18

[back to top]

Number of Participants With Vital Signs Abnormalities

Baseline was the last predose recording in each study period. Only post baseline values are included in this analysis (NCT05032950)
Timeframe: Baseline up to Day 28

InterventionParticipants (Count of Participants)
Midazolam 2 mg0
PF-07321332 300 mg/Ritonavir 100 mg + Midazolam 2 mg1
Ritonavir 100 mg + Midazolam 2 mg0

[back to top]

Terminal Half-life (t1/2) of Midazolam When Administered Alone, With PF-07321332/Ritonavir, and With Ritonavir

t½ for midazolam following single dose administration with and without PF-07321332/ritonavir or ritonavir was calculated by Loge(2)/kel, where kel was the terminal phase rate constant calculated by a linear regression of the log-linear concentration-time curve. Only those data points judged to describe the terminal log-linear. (NCT05032950)
Timeframe: Midazolam: Day 1 Predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36 hours postdose; Midazolam+PF-07321332/ritonavir: Day 5 predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36, 48 and 72 hours postdose

InterventionHour (Mean)
Midazolam 2 mg4.988
PF-07321332 300 mg/Ritonavir 100 mg + Midazolam 2 mg10.47
Ritonavir 100 mg + Midazolam 2 mg11.54

[back to top]

Time for Cmax (Tmax) of Midazolam When Administered Alone, With PF-07321332/Ritonavir, and With Ritonavir

Tmax for midazolam following single dose administration with and without PF-07321332/ritonavir or ritonavir was calculated by observed directly from data as time of first occurrence. (NCT05032950)
Timeframe: Midazolam: Day 1 Predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36 hours postdose; Midazolam+PF-07321332/ritonavir: Day 5 predose, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36, 48 and 72 hours postdose

InterventionHour (Median)
Midazolam 2 mg1.00
PF-07321332 300 mg/Ritonavir 100 mg + Midazolam 2 mg1.00
Ritonavir 100mg+ Midazolam 2mg1.02

[back to top]

Number of Participants With Electrocardiogram (ECG) Abnormalities

Baseline and changes from baseline in PR, QT, QRS, heart rate and QTcF were summarized by treatment and time postdose. Baseline was defined as the average of the triplicate predose recordings in each study period. ECG endpoints and changes from baseline (QTcF, PR, QRS), over all measurements taken postdose, were also summarized descriptively by treatment using categories as defined in the Criteria for Safety Values of Potential Clinical Concern appendix of the protocol and for QTc values corresponding to ICH E14 thresholds, which are: QTcF (msec): 450500; QTcF (msec) increase from baseline: 3060 (NCT05032950)
Timeframe: Baseline up to Day 28

,,
InterventionParticipants (Count of Participants)
PR INTERVAL, AGGREGATE (MSEC) - Value≥300PR INTERVAL, AGGREGATE (MSEC) - %Change≥25/50%QRS DURATION, AGGREGATE (MSEC) - Value≥140QRS DURATION, AGGREGATE (MSEC) - %Change≥50%QTCF INTERVAL, AGGREGATE (MSEC) - 450QTCF INTERVAL, AGGREGATE (MSEC) - 480QTCF INTERVAL, AGGREGATE (MSEC) - Value>500QTCF INTERVAL, AGGREGATE (MSEC) - 30QTCF INTERVAL, AGGREGATE (MSEC) - Change>60
Midazolam 2 mg000000000
PF-07321332 300 mg/Ritonavir 100 mg + Midazolam 2 mg000000000
Ritonavir 100 mg + Midazolam 2 mg000000000

[back to top]

Number of Participants With Treatment-emergent Adverse Events (TEAEs)

An adverse event was considered a TEAE if the event started during the effective duration of treatment. All events that start on or after the first dosing day and time/ start time, if collected, but before the last dose plus the lag time (28 days) were flagged as TEAEs. The algorithm did consider any events that started prior to the first dose date. Any events occurring following start of treatment or increasing in severity were counted as treatment emergent. Events that occur in a non-treatment period (for example, Washout or Follow-up) were counted as treatment emergent and attributed to the previous treatment taken. (NCT05032950)
Timeframe: Baseline up to Day 28

,,
InterventionParticipants (Count of Participants)
Participants with adverse events (All Causalities)Participants with adverse events (Treatment related)Participants with serious adverse eventsParticipants with severe adverse events
Midazolam 2 mg4400
PF-07321332 300 mg/Ritonavir 100 mg + Midazolam 2 mg9900
Ritonavir 100 mg + Midazolam 2 mg7700

[back to top]

Plasma Concentration Versus Time Summary of Nirmatrelvir (PF-07321332)

(NCT05047601)
Timeframe: Day 1: 1 hour post dose; Day 5: 2 hours pre-dose

,
InterventionNanograms per milliliter (Mean)
Day 1 (1 hour post-dose)Day 5 (2 hours pre-dose)
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 10 Days14721657
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 5 Days14891688

[back to top] [back to top]

Percentage of Participants Who Developed Symptomatic RT-PCR or RAT Confirmed SARS-CoV-2 Infection Through Day 14: Among Participants With Negative RT-PCR at Baseline

Percentage of participants who developed symptomatic Reverse Transcriptase Polymerase Chain Reaction (RT-PCR) or Rapid Antigen Test (RAT) confirmed SARS-Cov-2 infection were reported in this outcome measure. Index case was defined as participants with symptomatic COVID-19. (NCT05047601)
Timeframe: From Day 1 to Day 14

InterventionPercentage of participants (Number)
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 5 Days2.607
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 10 Days2.410
Placebo3.929

[back to top]

Percentage of Participants Who Developed Symptomatic RT-PCR or RAT Confirmed SARS-CoV-2 Infection Through Day 14: Among Participants With Negative RT-PCR at Baseline With Increased Risk of Severe COVID-19 Illness

Percentage of participants who had a symptomatic RT-PCR or RAT confirmed SARS-Cov-2 infection were reported in this outcome measure. The risk factors associated with severe covid-19 illness included age greater than or equal to 60 years, body mass index greater than 25, social history of smoking and presence of comorbidities. Index case was defined as participants with symptomatic COVID-19. (NCT05047601)
Timeframe: From Day 1 to Day 14

InterventionPercentage of participants (Number)
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 5 Days2.871
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 10 Days2.645
Placebo3.465

[back to top]

Percentage of Participants With Asymptomatic RT-PCR or RAT Confirmed SARS-CoV-2 Infection Through Day 14: Among Participants With Negative RT-PCR at Baseline

Percentage of participants who had asymptomatic RT-PCR or RAT confirmed SARS-CoV-2 infection through day 14 among participants with negative RT-PCR at baseline were reported in this outcome measure. Index case was defined as participants with symptomatic COVID-19. (NCT05047601)
Timeframe: From Day 1 to Day 14

InterventionPercentage of participants (Number)
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 5 Days2.014
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 10 Days1.928
Placebo3.095

[back to top] [back to top]

Percentage of Participants With Death Event Through Day 38: Among Participants With Negative RT-PCR at Baseline

Percentage of participants with death (all-cause) event were reported in this outcome measure. (NCT05047601)
Timeframe: From Day 1 to Day 38

InterventionPercentage of participants (Number)
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 5 Days0
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 10 Days0
Placebo0

[back to top]

Percentage of Participants With no, Mild, Moderate, or Severe Signs and Symptoms Attributed to COVID-19 Through Day 28: Among Participants With Negative RT-PCR at Baseline

Participants were categorized according to severity of signs and symptoms as no, mild, moderate, severe in this outcome measure. The 12 signs and symptoms included stuffy or runny nose, sore throat, shortness of breath or difficulty breathing, cough, low energy or tiredness, muscle or body aches, headache, chills or shivering, feeling hot or feverish, nausea, vomiting, diarrhea. Participants recorded their daily severity rating of their symptoms over the past 24 hours based on a 4-point scale in which 0 was reported if no symptoms were present; 1 if mild; 2 if moderate; and 3 if severe. (NCT05047601)
Timeframe: From Day 1 to Day 28

,,
InterventionPercentage of participants (Number)
NoMildModerateSevereMissing
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 10 Days83.3738.1935.0602.0481.325
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 5 Days81.5177.8206.8722.1331.659
Placebo81.6677.6197.1432.7380.833

[back to top]

Percentage of Participants With Symptomatic RT-PCR or RAT Confirmed SARS-CoV-2 Infection Through Day 14: Among Participants With Negative, Positive or Missing RT-PCR at Baseline

Percentage of participants with a negative, positive, or missing RT-PCR result at baseline, who had a symptomatic SARS-CoV-2 infection confirmed by RAT or RT-PCR through Day 14 were reported in this outcome measure. Index case was defined as participants with symptomatic COVID-19. (NCT05047601)
Timeframe: From Day 1 to Day 14

InterventionPercentage of Participants (Number)
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 5 Days3.712
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 10 Days4.848
Placebo5.269

[back to top]

Percentage of Participants With Symptomatic RT-PCR or RAT Confirmed SARS-CoV-2 Infection Through Day 14: Among Participants With Positive RT-PCR at Baseline

Percentage of participants with a positive RT-PCR result at baseline who had a symptomatic SARS-CoV-2 infection confirmed by RAT or RT-PCR through Day 14 were reported in this outcome measure. Index case was defined as participants with symptomatic COVID-19. (NCT05047601)
Timeframe: From Day 1 to Day 14

InterventionPercentage of participants (Number)
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 5 Days28.947
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 10 Days45.833
Placebo37.931

[back to top]

Viral Load in Nasal Samples Over Time: Among Participants With Negative RT-PCR at Baseline

Nasal samples were collected to estimate the viral load in terms of logarithm to base 10 (log10) copies per milliliter in participants with negative RT-PCR at baseline and were reported in this outcome measure. (NCT05047601)
Timeframe: From Day 1 to Day 14

,,
InterventionLog 10 copies per milliliter (Mean)
Day 1Day 2Day 3Day 4Day 5Day 6Day 7Day 8Day 9Day 10Day 11Day 12Day 13Day 14
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 10 Days0.0350.0530.0480.0570.0740.0900.0530.0810.0620.0640.0530.0510.0520.035
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 5 Days0.0420.0780.0900.0810.0790.0740.0880.0960.0940.0800.1030.1060.0850.108
Placebo0.0380.1080.1470.1650.2170.1890.1860.1590.1350.1330.1150.1030.1170.146

[back to top]

Time to RT-PCR or RAT Confirmed SARS-CoV-2 Infection Through Day 14: Among Participants With Negative RT-PCR at Baseline

Number of days between first dose and confirmation of the SARS-CoV-2 infection by RT-PCR or RAT was reported in this outcome measure. (NCT05047601)
Timeframe: From Day 1 to Day 14

InterventionDays (Median)
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 5 DaysNA
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 10 DaysNA
PlaceboNA

[back to top]

Number of Days of Hospitalization and Intensive Care Unit (ICU) Stay: Among Participants With Negative RT-PCR at Baseline

This outcome measure has been presented in terms of participants according to number of days of hospitalization and in ICU as 0 days and more than or equal to 1 day. (NCT05047601)
Timeframe: From Day 1 to Day 28

,,
InterventionParticipants (Count of Participants)
ICU Visits: 0 DayICU Visits: More than or equal to 1 dayHospitalization Visits: 0 DayHospitalization Visits: More than or equal to 1 day
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 10 Days83008300
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 5 Days84408440
Placebo84008391

[back to top]

Number of Participants With Treatment Emergent Adverse Events (TEAEs), Serious AEs and AEs Leading to Study and Study Drug Discontinuation

An AE was defined as any untoward medical occurrence in a participant temporally associated with the use of study intervention, whether or not considered related to the study intervention. An SAE was defined as any untoward medical occurrence at any dose that resulted in any of the following outcomes: death; life-threatening ; required inpatient hospitalization or prolongation of existing hospitalization; persistent or significant disability/incapacity ; congenital anomaly/birth defect; or that was considered as an important medical event. TEAEs were defined as events that started on or after the study medication start date and time. AEs included both serious and all non-serious adverse events. AEs that led to study discontinuation and AEs that led to discontinuation of study intervention and then continued study were also reported in this outcome measure. (NCT05047601)
Timeframe: From start of study intervention (Day 1) up to end of safety follow-up (Day 38)

,,
InterventionParticipants (Count of Participants)
TEAEsSAEsAEs led to discontinuation of studyAEs led to discontinue study intervention and continued study
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 10 Days2121011
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 5 Days2183010
Placebo1952014

[back to top]

Viral Load in Nasal Samples Over Time: Among Participants With Positive RT-PCR at Baseline

Nasal samples were collected to estimate the viral load in terms of logarithm to base 10 (log10) copies per milliliter in participants with positive RT-PCR at baseline and were reported in this outcome measure. (NCT05047601)
Timeframe: From Day 1 to Day 14

,,
InterventionLog 10 copies per milliliter (Mean)
Day 1Day 2Day 3Day 4Day 5Day 6Day 7Day 8Day 9Day 10Day 11Day 12Day 13Day 14
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 10 Days4.4702.7242.0511.5141.4130.9970.9130.9420.7660.5410.6030.6700.3130.345
Nirmatrelvir (PF-07321332) 300 mg + Ritonavir 100 mg 5 Days4.8703.2862.8802.6001.4701.0651.1991.2121.1690.8190.6230.5320.4130.284
Placebo4.8373.1043.2552.7212.9942.4661.4781.0721.1030.9650.7070.4360.3610.358

[back to top]

Apparent Volume of Distribution (Vz/F) of PF-07321332

Volume of distribution is defined as the theoretical volume in which the total amount of drug would need to be uniformly distributed to produce the desired plasma concentration of a drug. Apparent volume of distribution after oral dose (Vz/F) is influenced by the fraction absorbed. Vz/F was calculated as dose/(AUCinf*kel). kel was the terminal phase rate constant calculated by a linear regression of the loglinear concentration-time curve. (NCT05129475)
Timeframe: 0 (pre-dose), 0.5, 1, 1.5, 2, 2.5, 3, 4, 6, 8, 12, 16, 24 and 48 hours post-dose on Day 1 of Period 1 and 2

InterventionLiters (Geometric Mean)
PF-07321332 300 mg/Ritonavir 100 mg Fed68.77
PF-07321332 300 mg/Ritonavir 100 mg Fasted96.88

[back to top]

Area Under the Plasma Concentration-Time Profile From Time Zero to Extrapolated Infinite Time (AUCinf) of PF-07321332

AUCinf was calculated by AUClast + (Clast/kel). AUClast was the area under the plasma concentration-time profile from time zero to the time of the last quantifiable concentration (Clast). Clast was the predicted plasma concentration at the last quantifiable time point from the log-linear regression analysis and kel was the terminal phase rate constant calculated by a linear regression of the log-linear concentration-time curve. (NCT05129475)
Timeframe: 0 (pre-dose), 0.5, 1, 1.5, 2, 2.5, 3, 4, 6, 8, 12, 16, 24 and 48 hours post-dose on Day 1 of Period 1 and 2

InterventionNanogram*hour per milliliter (Geometric Mean)
PF-07321332 300 mg/Ritonavir 100 mg Fed44050
PF-07321332 300 mg/Ritonavir 100 mg Fasted36810

[back to top]

Area Under the Plasma Concentration-Time Profile From Time Zero to the Time of the Last Quantifiable Concentration (AUClast) of PF-07321332

Area under the plasma concentration-time profile from time zero to the time of the last quantifiable concentration was determined by linear/log trapezoidal method and reported in this outcome measure. (NCT05129475)
Timeframe: 0 (pre-dose), 0.5, 1, 1.5, 2, 2.5, 3, 4, 6, 8, 12, 16, 24 and 48 hours post-dose on Day 1 of Period 1 and 2

InterventionNanogram*hour per milliliter (Geometric Mean)
PF-07321332 300 mg/Ritonavir 100 mg Fed43360
PF-07321332 300 mg/Ritonavir 100 mg Fasted35860

[back to top]

Maximum Observed Plasma Concentration (Cmax) of PF-07321332

Cmax was defined as maximum observed plasma concentration. It was observed directly from data. (NCT05129475)
Timeframe: 0 (pre-dose), 0.5, 1, 1.5, 2, 2.5, 3, 4, 6, 8, 12, 16, 24 and 48 hours post-dose on Day 1 of Period 1 and 2

InterventionNanogram per milliliter (Geometric Mean)
PF-07321332 300 mg/Ritonavir 100 mg Fed5951
PF-07321332 300 mg/Ritonavir 100 mg Fasted3696

[back to top]

Number of Participants Meeting Pre-Specified Criteria for 12-Lead Electrocardiogram (ECG) Values

A 12-lead ECG was obtained using an ECG machine that automatically calculated the heart rate and measured PR, QT, and QTc intervals and QRS complex. All scheduled ECGs were performed after the participant had rested quietly for at least 5 minutes in a supine position. The pre specified criteria for ECG values included absolute value of QTCF (Fridericia's correction formula): >=450 to less than or equal to (<=) 480 milliseconds, >480 to <=500 milliseconds, >500 milliseconds, increase from baseline >30 - <=60, increase from baseline >60. PR interval: >=300 milliseconds, increase from baseline: baseline >200 milliseconds and max. increase >=25% and baseline <=200 milliseconds and max. increase >=50%; QRS duration: >=140 milliseconds, increase from baseline >=50%. (NCT05129475)
Timeframe: Day 1 (pre-dose) of Period 1 up to Day 3 of Period 2 (maximum of 7 days)

InterventionParticipants (Count of Participants)
PF-07321332 300 mg/Ritonavir 100 mg Fed0
PF-07321332 300 mg/Ritonavir 100 mg Fasted0

[back to top]

Number of Participants Meeting Pre-Specified Criteria for Vital Signs

Vital signs evaluations included supine blood pressure (BP) and pulse rate. The pre specified criteria for vital signs included systolic blood pressure (BP) minimum (min.) less than (<) 90 millimeter of mercury (mmHg); systolic BP change from baseline maximum (max.) decrease >= 30 mmHg, max. increase >=30 mmHg; diastolic BP min. <50mmHg; diastolic BP change from baseline max. decrease >=20, max. increase >=20; supine pulse rate min. <40 beats per minute (bpm) and max. >120 bpm. (NCT05129475)
Timeframe: Day 1 (pre-dose) of Period 1 up to Day 3 of Period 2 (maximum of 7 days)

InterventionParticipants (Count of Participants)
PF-07321332 300 mg/Ritonavir 100 mg Fed0
PF-07321332 300 mg/Ritonavir 100 mg Fasted0

[back to top]

Number of Participants With Clinical Laboratory Abnormalities

Clinical laboratory abnormalities included following criteria: a) hematology evaluation included eosinophils/leukocytes greater than (>) 1.2* upper limit of normal (ULN), monocytes/leukocytes >1.2*ULN; b) clinical chemistry evaluation included urobilinogen greater than or equal to (>=) 1, fibrinogen >1.25*baseline; c) urinalysis evaluation included urine hemoglobin >=1. (NCT05129475)
Timeframe: Day -1 of Period 1 up to Day 3 of Period 2 (maximum of 8 days)

InterventionParticipants (Count of Participants)
PF-07321332 300 mg/Ritonavir 100 mg Fed4
PF-07321332 300 mg/Ritonavir 100 mg Fasted4

[back to top]

Terminal Elimination Half-life (t1/2) of PF-07321332

t1/2 was calculated by Loge(2)/kel. kel was the terminal phase rate constant calculated by a linear regression of the loglinear concentration-time curve. (NCT05129475)
Timeframe: 0 (pre-dose), 0.5, 1, 1.5, 2, 2.5, 3, 4, 6, 8, 12, 16, 24 and 48 hours post-dose on Day 1 of Period 1 and 2

InterventionHours (Mean)
PF-07321332 300 mg/Ritonavir 100 mg Fed7.390
PF-07321332 300 mg/Ritonavir 100 mg Fasted8.673

[back to top]

Apparent Clearance (CL/F) of PF-07321332 From Plasma

Clearance of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. Clearance obtained after oral dose (apparent clearance) is influenced by the fraction of the dose absorbed. CL/F was calculated as dose/AUCinf. (NCT05129475)
Timeframe: 0 (pre-dose), 0.5, 1, 1.5, 2, 2.5, 3, 4, 6, 8, 12, 16, 24 and 48 hours post-dose on Day 1 of Period 1 and 2

InterventionLiters per hour (Geometric Mean)
PF-07321332 300 mg/Ritonavir 100 mg Fed6.812
PF-07321332 300 mg/Ritonavir 100 mg Fasted8.148

[back to top]

Time to Reach Maximum Observed Plasma Concentration (Tmax) of PF-07321332

Tmax was defined as the time to reach maximum observed plasma concentration of PF-07321332. (NCT05129475)
Timeframe: 0 (pre-dose), 0.5, 1, 1.5, 2, 2.5, 3, 4, 6, 8, 12, 16, 24 and 48 hours post-dose on Day 1 of Period 1 and 2

InterventionHours (Median)
PF-07321332 300 mg/Ritonavir 100 mg Fed2.50
PF-07321332 300 mg/Ritonavir 100 mg Fasted2.29

[back to top]

Number of Participants With Treatment-Emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs)

An adverse event (AE) was any untoward medical occurrence in a participant, temporally associated with the use of study intervention, whether or not considered related to the study intervention. An SAE was an AE which resulted in any of the following outcomes or deemed significant for any other reason: death; initial or prolonged inpatient hospitalization; life-threatening experience (immediate risk of dying); persistent or significant disability/incapacity; congenital anomaly. Treatment-emergent were events between first dose of study drug and up to maximum of 35 days after last dose that were absent before treatment or that worsened relative to pretreatment state. (NCT05129475)
Timeframe: Day 1 of dosing in the study up to maximum of 35 days after last dose of study drug (approximately maximum of 40 days)

,
InterventionParticipants (Count of Participants)
TEAEsSAEs
PF-07321332 300 mg/Ritonavir 100 mg Fasted50
PF-07321332 300 mg/Ritonavir 100 mg Fed60

[back to top]