Page last updated: 2024-11-08

darunavir

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

Description

Darunavir: An HIV PROTEASE INHIBITOR that is used in the treatment of AIDS and HIV INFECTIONS. Due to the emergence of ANTIVIRAL DRUG RESISTANCE when used alone, it is administered in combination with other ANTI-HIV AGENTS. [Medical Subject Headings (MeSH), National Library of Medicine, extracted Dec-2023]

darunavir : An N,N-disubstituted benzenesulfonamide bearing an unsubstituted amino group at the 4-position, used for the treatment of HIV infection. A second-generation HIV protease inhibitor, darunavir was designed to form robust interactions with the protease enzyme from many strains of HIV, including those from treatment-experienced patients with multiple resistance mutations to other protease inhibitors. [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 CID213039
CHEMBL ID1323
CHEBI ID367163
SCHEMBL ID118546
MeSH IDM0461844

Synonyms (130)

Synonym
HY-17040
tmc-41629
mc-114
uic-940t
(-)-darunavir
[(3as,4r,6ar)-2,3,3a,4,5,6a-hexahydrofuro[2,3-b]furan-4-yl] n-[(1s,2r)-3-[(4-aminophenyl)sulfonyl-isobutyl-amino]-1-benzyl-2-hydroxy-propyl]carbamate
tmc114
DRV ,
uic-96017
tmc 114
prezista(tm)
carbamic acid, [(1s,2r)-3-[[(4-aminophenyl)sulfonyl](2-methylpropyl)amino]-2-hydroxy-1-(phenylmethyl)propyl]-, (3r,3as,6ar)-hexahydrofuro[2,3-b]furan-3-yl ester
darunavir
tmc-114
prezista naive
darunavir (usan/inn)
D03656
206361-99-1
(3r,3as,6ar)-hexahydrofuro[2,3-b]furan-3-yl(1s,2r)-3-[[(4-aminophenyl)sulfonyl](isobutyl)amino]-1-benzyl-2-hydroxypropylcarbamate
darunavir (drv)
chembl1323 ,
(3r,3as,6ar)-hexahydrofuro[2,3-b]furan-3-yl n-[(2s,3r)-4-[(4-aminobenzene)(2-methylpropyl)sulfonamido]-3-hydroxy-1-phenylbutan-2-yl]carbamate
bdbm8125
uic-94017
2IDW
(3r,3as,6ar)-hexahydrofuro[2,3-b]furan-3-yl n-((1s,2r)-1-benzyl-2-hydroxy-3-(n1-isobutylsulfanilamido)propyl)carbamate
n-((1s,2r)-3-(((4-aminophenyl)sulfonyl)(2-methylpropyl)amino)-2-hydroxy-1-benzylpropyl)((1s,2r,5r)-4,6-dioxabicyclo(3.3.0)oct-2-yloxy)carboxamide
2F8G
2HS1
2F81
[(s)-3-[(4-amino-benzenesulfonyl)-isobutyl-amino]-2-hydroxy-1-((r)-phenylmethyl)-propyl]-carbamic acid (3r,3as,6ar)-(hexahydro-furo[2,3-b]furan-3-yl) ester
(3r,3as,6ar)-tetrahydro-2h-furo[2,3-b]furan-3-yl (2s,3r)-4-(4-amino-n-neopentylphenylsulfonamido)-3-hydroxy-1-phenylbutan-2-ylcarbamate
{(1s,2r)-3-[(4-amino-benzenesulfonyl)-isobutyl-amino]-1-benzyl-2-hydroxy-propyl}-carbamic acid (3r,3as,6ar)-(hexahydro-furo[2,3-b]furan-3-yl) ester
2IEN
(3r,3as,6ar)-tetrahydro-2h-furo[2,3-b]furan-3-yl (2s,3r)-4-(4-amino-n-isobutylphenylsulfonamido)-3-hydroxy-1-phenylbutan-2-ylcarbamate
DB01264
2F80
2HS2
NCGC00168773-01
darunavir [usan]
CHEBI:367163 ,
darunavirum
(3r,3as,6ar)-hexahydrofuro[2,3-b]furan-3-yl n-((1s,2r)-1-benzyl-2-hydroxy-3-(n(1)-isobutylsulfanilamido)propyl)carbamate
(3r,3as,6ar)-hexahydrofuro[2,3-b]furan-3-yl [(2s,3r)-4-{[(4-aminophenyl)sulfonyl](2-methylpropyl)amino}-3-hydroxy-1-phenylbutan-2-yl]carbamate
3D20
3D1Z
3CYW
3BVB
[(3as,4r,6ar)-2,3,3a,4,5,6a-hexahydrofuro[2,3-b]furan-4-yl] n-[(2s,3r)-4-[(4-aminophenyl)sulfonyl-(2-methylpropyl)amino]-3-hydroxy-1-phenylbutan-2-yl]carbamate
dtxsid0046779 ,
dtxcid8026779
tox21_112634
cas-206361-99-1
BCP9000587
(3r,3as,6ar)-hexahydrofuro(2,3-b)furan-3-yl n-((1s,2r)-1-benzyl-2-hydroxy-3-(n(sup 1)-isobutylsulfanilamido)propyl)carbamate
darunavirum [inn-latin]
tmc41629
darunavir [usan:inn:ban]
(3r,3as,6ar)-hexahydrofuro(2,3-b)furan-3-yl n-((1s,2r)-1-benzyl-2-hydroxy-3-(n1-isobutylsulfanilamido)propyl)carbamate
tmc 41629
unii-yo603y8113
carbamic acid, ((1s,2r)-3-(((4-aminophenyl)sulfonyl)(2-methylpropyl)amino)-2-hydroxy-1-(phenylmethyl)propyl)-, (3r,3as,6ar)-hexahydrofurano(2,3-b)furan-3-yl ester
hsdb 7788
yo603y8113 ,
((1s,2r)-3-(((4-aminophenyl)sulfonyl)(2-methylpropyl)amino)-2-hydroxy-1-(phenylmethyl)propyl)-carbamic acid (3r,3as,6ar)-hexahydrofurano(2,3-b)furan-3-yl ester
AKOS015966592
aids073035
drv & hsa
darunavir & alpha1-acid glycoprotein
drv & aag
BCP0726000058
darunavir [inn]
darunavir [orange book]
darunavir [mi]
darunavir [hsdb]
darunavir [ema epar]
(3r,3as,6ar)-hexahydrofuro(2,3-b)furan-3-yl ((1s,2r)-3-(((4-aminophenyl)sulfonyl)(isobutyl)amino)-1-benzyl-2-hydroxypropyl)carbamate
darunavir [vandf]
darunavir [mart.]
((1s,2r)-3-(((4-aminophenyl)sulfonyl)(2-methylpropyl)amino)-2-hydroxy-1-(phenylmethyl)propyl)-carbamic acid (3r,3as,6ar)-hexahydrofuro(2,3-b)furan-3-yl ester
darunavir [who-dd]
(3r,3as,6ar)-hexahydrofuro(2,3-b)furan-3-yl ((2s,3r)-4-((4-amino-n-isobutylphenyl)sulfonamido)-3-hydroxy-1-phenylbutan-2-yl)carbamate
3QOZ
4HLA
CS-0749
S5250
SCHEMBL118546
3PWM
3LZU
3GGU
3TKW
3LZS
3U7S
3LZV
3EKT
3T3C
3OGP
3S53
3SO9
3S54
CJBJHOAVZSMMDJ-HEXNFIEUSA-N
AC-26778
(3r,3as,6ar)-hexahydrofuro[2,3-b]furan-3-yl ((2s,3r)-4-(4-amino-n-isobutylphenylsulfonamido)-3-hydroxy-1-phenylbutan-2-yl)carbamate
AB01565837_02
mfcd09260006
3TTP
4LL3
(3r,3as,6ar)-hexahydrofuro[2,3-b]furan-3-yl n-[(2s,3r)-3-hydroxy-4-[n-(2-methylpropyl)(4-aminobenzene)sulfonamido]-1-phenylbutan-2-yl]carbamate
gtpl11243
[(3r,3as,6ar)-2,3,3a,4,5,6a-hexahydrofuro[5,4-b]furan-3-yl] n-[(2s,3r)-4-[(4-aminophenyl)sulfonyl-(2-methylpropyl)amino]-3-hydroxy-1-phenylbutan-2-yl]carbamate
darunavir, >=98% (hplc)
J-013483
HMS3715I13
(3r,3as,6ar)-hexahydrofuro[2,3-b]furan-3-yl (2s,3r)-4-(4-amino-n-isobutylphenylsulfonamido)-3-hydroxy-1-phenylbutan-2-ylcarbamate
Q3765251
EX-A4009
KS-1469
3-(4-amino-phenoxy)-pyrrolidine-1-carboxylicacidtert-butylester
(3r,3as,6ar)-hexahydrofuro[2,3-b]furan-3-yl n-[(1s,2r)-3-[[(4-aminophenyl)sulfonyl](2-methylpropyl)amino]-2-hydroxy-1-(phenylmethyl)propyl]carbamate
NCGC00388284-08
AMY373
CCG-269991
NCGC00388284-07
206361-99-1 (free)
derunavir
NCGC00388284-12
(3r,3as,6ar)-hexahydrofuro[2,3-b]furan-3-yl((2s,3r)-4-(4-amino-n-isobutylphenylsulfonamido)-3-hydroxy-1-phenylbutan-2-yl)carbamate
EN300-20600172
(3r,3as,6ar)-hexahydrofuro[2,3-b]furan-3-yl n-[(2s,3r)-3-hydroxy-4-[n-(2-methylpropyl)-4-aminobenzenesulfonamido]-1-phenylbutan-2-yl]carbamate
Z2301684748

Research Excerpts

Overview

Darunavir is a human immunodeficiency virus type 1 (HIV-1) protease inhibitor boosted with ritonavir (DRV/r) or cobicistat. It is an efficacious drug; however, pharmacokinetic variability has been reported.

ExcerptReferenceRelevance
"Darunavir is an anti-HIV protease inhibitor repurposed for SARS-CoV-2 treatment."( Comparative Population Pharmacokinetics of Darunavir in SARS-CoV-2 Patients vs. HIV Patients: The Role of Interleukin-6.
Biasizzo, J; Cojutti, PG; Della Siega, P; Fabris, M; Givone, F; Londero, A; Pea, F; Tascini, C, 2020
)
2.26
"Darunavir (DRV) is a potent antiviral drug used for treatment of infections with human immunodeficiency virus (HIV). "( Preparation and Characterization of Two Immunogens and Production of Polyclonal Antibody with High Affinity and Specificity for Darunavir.
Almehizia, AA; Darwish, IA; Herqash, RN; Radwan, AA, 2020
)
2.21
"Darunavir (DRV) is a highly potent protease inhibitor (PI) that is oftentimes effective when drug resistance has emerged against first-generation inhibitors."( Darunavir-Resistant HIV-1 Protease Constructs Uphold a Conformational Selection Hypothesis for Drug Resistance.
Bentz, K; Fanucci, GE; Hu, L; Liu, Z; Pham, L; Savin, DA; Tran, TT, 2020
)
2.72
"Darunavir is a human immunodeficiency virus type 1 (HIV-1) protease inhibitor boosted with ritonavir (DRV/r) or cobicistat."( 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
)
2.34
"Darunavir is an efficacious drug; however, pharmacokinetic variability has been reported. "( Risk factors contributing to a low darunavir plasma concentration.
Alffenaar, JC; Bierman, WFW; Daskapan, A; Kosterink, JGW; Stienstra, Y; Touw, DJ; van der Werf, TS, 2018
)
2.2
"Darunavir/ritonavir is a potent PI with a high genetic barrier and pharmacological robustness favourably investigated as monotherapy. "( Darunavir/ritonavir monotherapy at a low dose (600/100 mg/day) in HIV-1-infected individuals with suppressed HIV viraemia.
Assoumou, L; Caby, F; Calin, R; Katlama, C; Lê, MP; Marcelin, AG; Nguyen, T; Peytavin, G; Schneider, L; Seang, S; Soulie, C; Tubiana, R; Valantin, MA, 2018
)
3.37
"Darunavir (PrezistaTM) is a new protease inhibitor of HIV-1."( Drug-Drug Interactions Between PA-824 and Darunavir Based on Pharmacokinetics in Rats by LC-MS-MS.
Feng, T; Jing, J; Liu, X; Mi, L; Shen, X; Wang, L; Zhang, R; Zhang, S; Zhao, J; Zhou, N, 2018
)
1.47
"Darunavir (DRV) is an antiretroviral drug used to treat and prevent human immunodeficiency virus/acquired immune deficiency syndrome (HIV/AIDS), but whether DRV is protective against CPT-11-induced intestinal toxicity is unclear."( Darunavir alleviates irinotecan-induced intestinal toxicity in Vivo.
Jiang, W; Lan, T; Li, D; Ren, Y; Tian, J; Zhang, G; Zhang, X, 2018
)
2.64
"Darunavir is a second-generation protease inhibitor and is registered for the treatment of HIV-1 infection. "( Darunavir Population Pharmacokinetic Model Based on HIV Outpatient Data.
Alffenaar, JC; Bierman, WFW; Cattaneo, D; Daskapan, A; Gervasoni, C; Kosterink, JGW; Proost, JH; Resnati, C; Stienstra, Y; Touw, DJ; Tran, QTD; van der Werf, TS, 2019
)
3.4
"Darunavir/ritonavir is a substrate and inhibitor of CYP3A."( 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
)
1.36
"Darunavir is a potent protease inhibitor of HIV. "( 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
)
2.06
"Darunavir is a protease inhibitor that is administered with low-dose ritonavir to enhance its bioavailability. "( 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
)
2.07
"Darunavir is a synthetic non-peptidic protease inhibitor that has been shown to be extremely potent against wild-type HIV, and it is an important component of highly active antiretroviral treatment (HAART), which is considered as one of the most significant advances in the field of HIV therapy. "( A critical review of properties of darunavir and analytical methods for its determination.
Corrêa, JC; D'Arcy, DM; Salgado, HR; Serra, CH, 2014
)
2.12
"Darunavir is a relatively well-tolerated ARV, but concurrent ritonavir administration has several disadvantages (e.g., dose-related hyperlipidemia, gastrointestinal intolerance, drug-drug interactions) which may decrease patient adherence. "( The use of unboosted darunavir in the setting of ritonavir intolerance: three case reports.
Fulco, PP; Gomes, D; Nixon, D; Smith, K, 2016
)
2.2
"Darunavir (DRV) is a nonpeptidomimetic protease inhibitor approved for use with a ritonavir booster (DRV/r)."( 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
)
1.42
"Darunavir (DRV, TMC114) is a novel protease inhibitor administered in combination with low-dose ritonavir (DRV/r) and is highly active against both wild-type and multidrug-resistant HIV-1 strains. "( 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
)
2.08
"Darunavir is a promising new drug with good clinical efficacy data and safety profile."( New therapeutic agents in the management of HIV: an overview of darunavir for clinicians.
Hoy, J; Rotty, J, 2008
)
1.31
"Darunavir is a new protease inhibitor. "( [Darunavir in patients with advanced HIV and multiresistance. The POWER, DUET and BENCHMRK studies].
Arazo Garcés, P; Omiste Sanvicente, T, 2008
)
2.7
"Darunavir is a new HIV protease inhibitor that has been shown to be highly useful in HIV-infected patients. "( [Pharmacological interactions with darunavir].
Clotet, B; Moltó, J; Valle, M, 2008
)
2.07
"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,
)
1.84
"Darunavir (TMC114) is a new HIV protease inhibitor (PI)."( Effects of ritonavir-boosted darunavir vs. ritonavir-boosted atazanavir on lipid and glucose parameters in HIV-negative, healthy volunteers.
Diego Miralles, G; Lefebvre, E; Sekar, V; Tomaka, F; Van Baelen, B; Vandevoorde, A; Vangeneugden, T, 2009
)
2.09
"Darunavir is an oral nonpeptidic HIV-1 protease inhibitor (PI) that is used, together with a low boosting dose of ritonavir, as part of an antiretroviral therapy (ART) regimen in treatment-experienced and -naive patients with HIV-1 infection. "( Darunavir: a review of its use in the management of HIV infection in adults.
Keam, SJ; McKeage, K; Perry, CM, 2009
)
3.24
"Darunavir (DRV) is a nonpeptidic protease inhibitor (PI) approved for the treatment of human immunodeficiency virus (HIV) infection. "( P-glycoprotein mediates efflux transport of darunavir in human intestinal Caco-2 and ABCB1 gene-transfected renal LLC-PK1 cell lines.
Fujimoto, H; Ghosh, AK; Hamada, A; Higuchi, M; Koh, Y; Mitsuya, H; Saito, H; Tanoue, N; Watanabe, H, 2009
)
2.06
"Darunavir is a substrate for efflux and influx transporters in PBMC and intracellular concentrations can be manipulated using known inhibitors."( Intracellular 'boosting' of darunavir using known transport inhibitors in primary PBMC.
Back, D; Chandler, B; Hartkoorn, R; Janneh, O; Khoo, S; Kwan, WS; Owen, A, 2009
)
2.09
"Darunavir is a nonpeptidic HIV type 1 (HIV-1) protease inhibitor (PI) that binds with high affinity to the HIV-1 protease, including multi-drug resistant proteases. "( Darunavir: in treatment-experienced pediatric patients with HIV-1 infection.
McKeage, K; Scott, LJ, 2010
)
3.25
"Darunavir is a second-generation protease inhibitor designed to have antiviral efficacy against HIV-1 with multiple resistance mutations to protease inhibitors. "( In situ intestinal perfusion in knockout mice demonstrates inhibition of intestinal p-glycoprotein by ritonavir causing increased darunavir absorption.
Annaert, P; Augustijns, P; Holmstock, N; Mols, R, 2010
)
2.01
"Darunavir (TMC 114) is a protease inhibitor used in the therapy of HIV-1. "( 800 mg Darunavir tablets prepared by hot melt extrusion.
Baert, L; Rosier, J; Thommes, M,
)
2.03
"Darunavir is a new protease inhibitor (PI) that is co-administered with low-dose ritonavir and has demonstrated substantial efficacy in clinical trials of highly treatment-experienced patients when combined with an optimized background regimen (with or without enfuvirtide). "( Cost effectiveness of darunavir/ritonavir in highly treatment-experienced, HIV-1-infected adults in the USA.
Brogan, A; Martin, S; Mauskopf, J; Smets, E, 2010
)
2.12
"Darunavir is a new-generation nonpeptidic HIV protease inhibitor (PI), used with low doses of ritonavir for pharmacologic enhancement (boosting). "( Darunavir: an effective protease inhibitor for HIV-infected patients.
Phung, BC; Yeni, P, 2011
)
3.25
"Darunavir (DRV) is an HIV-1 protease inhibitor that is used together with a low boosting dose of ritonavir as part of an antiretroviral therapy (ART) regimen in treatment-experienced and naïve HIV-positive patients. "( A reduced dose of darunavir/ritonavir is effective in PI-experienced HIV-infected patients.
Bonora, S; Calcagno, A; Lanzafame, M; Lattuada, E; Vento, S,
)
1.91
"Darunavir is a synthetic nonpeptidic protease inhibitor which has been shown to be extremely potent against wild-type HIV as well as a large panel of PI-resistant clinical isolates and shows a high genetic barrier to the development of antiretroviral resistance. "( Darunavir: a critical review of its properties, use and drug interactions.
D'Arcy, DM; dos Reis Serra, CH; Nunes Salgado, HR; Ruela Corrêa, JC, 2012
)
3.26
"Darunavir (TMC114) is a second-generation, sulfonamide, nonpeptidic protease inhibitor (PI) with a unique, flexible, 3-dimensional structure that contributes to it high potency and slow selection of resistant virus. "( Darunavir: an overview of an HIV protease inhibitor developed to overcome drug resistance.
Hicks, CB; Taiwo, BO, 2007
)
3.23
"Darunavir is a new HIV PI that retains virological activity in the presence of multiple protease mutations. "( Darunavir: a second-generation protease inhibitor.
Busse, KH; Penzak, SR, 2007
)
3.23
"Darunavir (TMC114) is a newly developed HIV-1 protease inhibitor with potent antiviral activity against both wild-type and multidrug resistant HIV-1 strains. "( Clinical pharmacokinetics of darunavir.
Arastéh, K; Rittweger, M, 2007
)
2.07
"Darunavir (DRV) is a new protease inhibitor used to treat human immunodeficiency virus (HIV) type-1. "( The validation of plasma darunavir concentrations determined by the HPLC method for protease inhibitors.
Banno, K; Hirano, A; Kaneda, T; Kudaka, Y; Okumura, N; Takahashi, M, 2007
)
2.09
"Darunavir is a synthetic nonpeptidic analogue of amprenavir with enhanced activity against resistant virus that became available in 2006."( Darunavir: a nonpeptidic antiretroviral protease inhibitor.
McCoy, C, 2007
)
2.5
"Darunavir is a nonpeptidic protease inhibitor recently approved for the treatment of antiretroviral therapy-experienced patients. "( Darunavir.
El-Atrouni, WI; Temesgen, Z, 2007
)
3.23
"Darunavir (TMC114) is a nonpeptidic peptidomimetic HIV protease inhibitor (PI), with a high binding affinity and a close fit within the substrate envelope. "( Darunavir: in the treatment of HIV-1 infection.
Fenton, C; Perry, CM, 2007
)
3.23
"Darunavir (TMC114) is a new HIV protease inhibitor that has demonstrated substantial antiretroviral activity against wild-type HIV-1 virus and multidrug-resistant strains. "( Darunavir: pharmacokinetics and drug interactions.
Back, D; Hoetelmans, RM; Sekar, V, 2008
)
3.23

Effects

Darunavir has a low oral bioavailability (37%) due to its lipophilic nature. metabolism by cytochrome P450 enzymes and P-gp efflux. DarunavIR has a high genetic barrier to resistance.

Darunavir has been approved for the treatment of HIV/AIDS patients harboring multidrug-resistant HIV-1 variants that do not respond to previously existing HAART regimens. The drug has a low oral bioavailability (37%) due to its lipophilic nature, metabolism by cytochrome P450 enzymes and P-gp efflux.

ExcerptReferenceRelevance
"Darunavir has a low oral bioavailability (37%) due to its lipophilic nature, metabolism by cytochrome P450 enzymes and P-gp efflux. "( Darunavir-Loaded Lipid Nanoparticles for Targeting to HIV Reservoirs.
Desai, J; Thakkar, H, 2018
)
3.37
"Darunavir has a high genetic barrier to resistance. "( HIV-1 resistance rarely observed in patients using darunavir once-daily regimens across clinical studies.
Brown, K; De Meyer, S; Lathouwers, E; Luo, D; Seyedkazemi, S; Wong, EY,
)
1.83
"Darunavir has recently been approved for the treatment of HIV/AIDS patients harboring multidrug-resistant HIV-1 variants that do not respond to previously existing HAART regimens."( Darunavir, a conceptually new HIV-1 protease inhibitor for the treatment of drug-resistant HIV.
Dawson, ZL; Ghosh, AK; Mitsuya, H, 2007
)
2.5
"Darunavir has been selected as an active pharmaceutical ingredient that is classified as a Class 2 BCS substance and exists in two commercially available forms: crystalline ethanolate and amorphous."( Development of novel darunavir amorphous solid dispersions with mesoporous carriers.
Buzanov, GA; Cheremisin, AM; Dain, IA; Demina, NB; Filatov, AR; Kozhukhova, EI; Retivov, VM; Shevlyagina, NV; Zakhoda, OY; Zolotov, SA; Zolotova, AS, 2021
)
1.66
"Darunavir has a low oral bioavailability (37%) due to its lipophilic nature, metabolism by cytochrome P450 enzymes and P-gp efflux. "( Darunavir-Loaded Lipid Nanoparticles for Targeting to HIV Reservoirs.
Desai, J; Thakkar, H, 2018
)
3.37
"Darunavir has a high genetic barrier to resistance. "( HIV-1 resistance rarely observed in patients using darunavir once-daily regimens across clinical studies.
Brown, K; De Meyer, S; Lathouwers, E; Luo, D; Seyedkazemi, S; Wong, EY,
)
1.83
"Darunavir (DRV) has bimodal activity against HIV-1 protease, enzymatic inhibition and protease dimerization inhibition, and has an extremely high genetic barrier against development of drug resistance. "( Mechanism of Darunavir (DRV)'s High Genetic Barrier to HIV-1 Resistance: A Key V32I Substitution in Protease Rarely Occurs, but Once It Occurs, It Predisposes HIV-1 To Develop DRV Resistance.
Aoki, M; Aoki-Ogata, H; Das, D; Ghosh, AK; Hayashi, H; Mitsuya, H; Takamatsu, Y, 2018
)
2.29
"Darunavir has been proven efficacious for antiretroviral-experienced HIV-1-infected patients in randomized trials. "( Virologic and immunologic effectiveness of darunavir-based salvage therapy in HIV-1-infected adults in a Brazilian clinical practice setting: results of a multicenter and retrospective cohort study.
Biscione, FM; Greco, DB; Machado, DP; Ribeiro, KM; Tupinambás, U; Westin, MR,
)
1.84
"Darunavir (DRV) has been confirmed to be an effective option for antiretroviral-naïve and experienced patients. "( Minimal effects of Darunavir on adipocyte differentiation and metabolism in 3T3-L1 cells.
Blanco, JR; Oteo, JA; Pérez-Martínez, L; Pérez-Matute, P, 2012
)
2.15
"Darunavir has to be coadministered with low-dose ritonavir and food to optimize its pharmacokinetics."( Darunavir: an overview of an HIV protease inhibitor developed to overcome drug resistance.
Hicks, CB; Taiwo, BO, 2007
)
2.5
"Darunavir has improved activity against resistant HIV isolates in patients with few treatment choices, particularly when enfuvirtide is added. "( Darunavir: a nonpeptidic antiretroviral protease inhibitor.
McCoy, C, 2007
)
3.23

Actions

ExcerptReferenceRelevance
"Darunavir AUC0-12 was lower with the increased dose during the second {[geometric mean ratio (GMR) of 0.62 (IQR 0.44-0.88); P = 0.055]} and third trimesters [GMR 0.64 (IQR 0.55-0.73); P = <0.001] compared with postpartum."( 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
)
2.72

Treatment

Darunavir/ritonavir treatment is safe and effective in treatment-experienced patients, irrespective of sex or race. No significant effect on expression of BCRP, MRP2 and P-glycoprotein implicated in efflux of different ARV drugs.

ExcerptReferenceRelevance
"Darunavir-containing treatment change episodes (TCEs) in patients previously failing PIs were selected from large European databases. "( Improved darunavir genotypic mutation score predicting treatment response for patients infected with HIV-1 subtype B and non-subtype B receiving a salvage regimen.
Brockmeyer, NH; Castagna, A; Churchill, D; Cozzi-Lepri, A; De Luca, A; De Wit, S; Descamps, D; Dunn, D; Flandre, P; Garcia, F; Günthard, HF; Imaz, A; Kordossis, T; Mussini, C; Obel, N; Perno, CF; Reiss, P; Roca, B; Santoro, MM; Schülter, E; Torti, C; van Sighem, A; Wensing, A; Wittkop, L; Zangerle, R; Zazzi, M, 2016
)
2.29
"Darunavir/ritonavir treatment is safe and effective in treatment-experienced patients, irrespective of sex or race."( Assessing darunavir/ritonavir-based therapy in a racially diverse population: 48-week outcomes from GRACE.
Currier, JS; Falcon, R; Garcia, F; Kumar, P; Mrus, J; Ryan, R; Smith, KY; Squires, K,
)
1.26
"Treatment with darunavir and dapivirine showed no significant effect on expression of BCRP, MRP2 and P-glycoprotein implicated in efflux of different ARV drugs."( Expression of Genes for Drug Transporters in the Human Female Genital Tract and Modulatory Effect of Antiretroviral Drugs.
Cuppone, AM; De Falco, G; Ekeruche-Makinde, J; Hijazi, K; Hold, GL; Iannelli, F; Kelly, CG; Pozzi, G; Shattock, R; Smith, K; Stincarelli, MA, 2015
)
0.76

Toxicity

Darunavir, rilpivirine and raltegravir do not induce toxic effects on Hep3B cells and primary rat neurons. This suggests a safer hepatic and neurological profile than that of efavirenz.

ExcerptReferenceRelevance
" 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
)
0.34
" 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
)
1.08
" 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
)
0.86
" 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
)
0.65
" 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
)
0.9
" 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,
)
0.4
" 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
)
0.63
" 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
)
0.92
" 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
)
0.66
" The most frequent adverse effect is rash (affecting 19% of patients), which is usually mild (grades 1 or 2) and does not lead to drug withdrawal."( [Safety and tolerability of etravirine].
Portilla, J, 2009
)
0.35
" 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
)
0.65
"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
)
2.05
"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,
)
0.38
"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,
)
0.57
" 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,
)
0.36
"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
)
0.63
"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
)
0.66
" 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
)
0.64
" 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,
)
0.37
"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
)
0.61
"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
)
0.61
"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
)
0.63
" However, data on the long-term exposure to these therapeutic options are needed, and a handful of case reports are emerging, reporting rare but potentially life-threatening adverse hepatic events in patients with hepatitis co-infection or taking other hepatotoxic drugs."( Hepatoxicity of new antiretrovirals: a systematic review.
Lacombe, K; Surgers, L, 2013
)
0.39
" Outcomes were incidence and health care costs of the following medically attended (International Classification of Diseases, Ninth Revision, Clinical Modification-coded or treated) adverse effects during the evaluation period: gastrointestinal, lipid abnormalities, diabetes/hyperglycemia, rash, and jaundice."( Comparative incidence and health care costs of medically attended adverse effects among U.S. Medicaid HIV patients on atazanavir- or darunavir-based antiretroviral therapy.
Chu, BC; Esker, S; Espindle, D; Hebden, T; Johnston, SS; Juday, T; Uy, J,
)
0.34
" Adverse event (AE) and treatment-related AE incidence was 91."( Safety and immunovirologic outcomes with maraviroc combination regimens in patients with a history of past treatment failures and virologic resistance in Brazil: an open-label, multicenter phase 3b study.
Bicudo, EL; Cassoli, LM; da Eira, M; de Andrade Neto, JL; Furtado, J; Leite, OH; Lewi, DS; Lima, MP; Lopes, MI; Machado, AA; Madruga, JV; Miranda, AF; Netto, EM; Pedro, Rde J; Portsmouth, S; Santini-Oliveira, M; Santos, BR; Tupinambas, U; Wajsbrot, DB, 2013
)
0.39
" 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
)
0.64
" 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
)
0.64
"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
)
0.64
"Growing evidence associates the non-nucleoside reverse transcriptase inhibitor efavirenz with several adverse events."( Lack of mitochondrial toxicity of darunavir, raltegravir and rilpivirine in neurons and hepatocytes: a comparison with efavirenz.
Alegre, F; Apostolova, N; Blas-García, A; Esplugues, JV; Funes, HA; Martínez, E; Polo, M, 2014
)
0.68
"Darunavir, rilpivirine and raltegravir do not induce toxic effects on Hep3B cells and primary rat neurons, which suggests a safer hepatic and neurological profile than that of efavirenz."( Lack of mitochondrial toxicity of darunavir, raltegravir and rilpivirine in neurons and hepatocytes: a comparison with efavirenz.
Alegre, F; Apostolova, N; Blas-García, A; Esplugues, JV; Funes, HA; Martínez, E; Polo, M, 2014
)
2.12
" 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
)
0.92
"3%) adverse events (AEs) were of mild or moderate severity with few grade 3/4 events, discontinuations, or temporary discontinuations/dose reductions due to AEs or serious AEs."( The maraviroc expanded access program - safety and efficacy data from an open-label study.
Craig, C; Heera, J; Lazzarin, A; Molina, JM; Mukwaya, G; Reynes, J; Sierra-Madero, JG; Valluri, S; van der Ryst, E,
)
0.13
"Cohorts contributed individual patient data on adverse events (AE) in those aged <18 years taking DRV and ATV, respectively, to 02/2014."( Safety of darunavir and atazanavir in HIV-infected children in Europe and Thailand.
, 2016
)
0.84
"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
)
0.65
" 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
)
0.67
" 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
)
0.7
" 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
)
0.74
" Numbers of discontinuations related to adverse events (11 [1%] of 763 patients in the study group vs four [1%] of 378 patients in the control group) and grade 3-4 adverse events (52 [7%] patients vs 31 [8%] patients) were similar between the two groups."( Efficacy and safety of switching from boosted protease inhibitors plus emtricitabine and tenofovir disoproxil fumarate regimens to single-tablet darunavir, cobicistat, emtricitabine, and tenofovir alafenamide at 48 weeks in adults with virologically suppr
Arribas, JR; Eron, JJ; Gathe, J; Hufkens, V; Lathouwers, E; Molina, JM; Negredo, E; Opsomer, M; Orkin, C; Petrovic, R; Van Landuyt, E; Vanveggel, S, 2018
)
0.68
"Dual therapy with RPV + DRVb proved to be effective and safe in patients with advanced HIV infection, long exposure to ART, low CD4 nadir, previous virologic failure, and/or history of ineffective ART."( Effectiveness and safety of dual therapy with rilpivirine and boosted darunavir in treatment-experienced patients with advanced HIV infection: a preliminary 24 week analysis (RIDAR study).
Arazo, P; Carmena, J; Crusells, MJ; de Jesus, SE; de la Torre, J; Galindo, MJ; Hidalgo-Tenorio, C; Lozano, F; Palacios, Z; Pasquau, J; Ríos, MJ; Samperiz, G; Santos, J; Tornero, C; Verdejo, G, 2019
)
0.75
" At week 48, there were no virological failures, three patients discontinued the regimen due to neuropsychiatric adverse events, two were lost to follow-up, and therefore the efficacy was 90% (95% CI, 82, 99%, intention-to-treat analysis)."( Efficacy and safety of dolutegravir plus boosted-darunavir dual therapy among highly treatment-experienced patients.
Casado, JL; Fontecha, M; Monsalvo, M; Rojo, A; Vivancos, MJ; Vizcarra, P, 2019
)
0.77
" Few discontinued due to adverse events (2% D/C/F/TAF arm)."( Week 96 efficacy and safety results of the phase 3, randomized EMERALD trial to evaluate switching from boosted-protease inhibitors plus emtricitabine/tenofovir disoproxil fumarate regimens to the once daily, single-tablet regimen of darunavir/cobicistat/
Brown, K; Cunningham, D; De Wit, S; Eron, JJ; Hufkens, V; Jezorwski, J; Lathouwers, E; Opsomer, M; Orkin, C; Petrovic, R; Post, FA; Pulido, F; Van Landuyt, E, 2019
)
0.7
" 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
)
0.89
"This current investigation was aimed to generate signals for adverse events (AEs) of darunavir-containing agents by data mining using the US Food and Drug Administration Adverse Event Reporting System (FAERS)."( Systematic analysis of safety profile for darunavir and its boosted agents using data mining in the FDA Adverse Event Reporting System database.
Chen, L; He, G; Jia, Y; Tian, X; Wang, K; Yao, Y, 2021
)
1.11
" 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
)
1.2
" This post hoc analysis evaluated gastrointestinal adverse events of interest (AEOIs; diarrhea, nausea, abdominal discomfort, flatulence [MedDRAv21]) through Wk96 among patients enrolled in the phase 3 AMBER (treatment-naïve) and EMERALD (virologically suppressed) studies of darunavir/cobicistat/emtricitabine/tenofovir alafenamide (D/C/F/TAF) 800/150/200/10 mg."( Low Incidence and Brief Duration of Gastrointestinal Adverse Events with Darunavir/Cobicistat/Emtricitabine/Tenofovir Alafenamide (D/C/F/TAF) Over 96 Weeks: Post hoc Analyses of AMBER and EMERALD.
Anderson, D; Baugh, B; Bejou, N; Campbell, J; Dunn, K; Luo, D; Seyedkazemi, S,
)
0.54
" Of the 3 withdrawals, 2 were attributed to drug-related adverse events."( Pharmacokinetics, Safety, and Tolerability of Once-Daily Darunavir With Cobicistat and Weekly Isoniazid/Rifapentine.
Adeojo, L; Brooks, KM; Bunn, HT; George, JM; Kovacs, JA; Kumar, P; Pau, AK; Peloquin, CA; Swaim, D, 2023
)
1.16

Pharmacokinetics

Darunavir Cmin was 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. The developed method was successfully applied to a pharmacokinetic study of (+) and (-) enantiomers of dar unavir on rat dried blood spots.

ExcerptReferenceRelevance
"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
)
2.06
" 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
)
0.86
"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
)
0.6
" 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
)
0.86
"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
)
0.84
"To investigate the potential for pharmacokinetic interactions between the protease inhibitors darunavir (DRV, TMC114) coadministered with low-dose ritonavir (darunavir/r), and atazanavir in HIV-negative, healthy volunteers."( Pharmacokinetics of darunavir (TMC114) and atazanavir during coadministration in HIV-negative, healthy volunteers.
De Marez, T; De Paepe, E; De Pauw, M; Hoetelmans, RM; Lefebvre, E; Sekar, VJ; Spinosa-Guzman, S; Vangeneugden, T, 2007
)
0.88
"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
)
0.56
" 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
)
0.66
" 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
)
0.63
"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
)
0.83
"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
)
2
" This article reviews all the pharmacokinetic and drug-drug interaction studies conducted to date for darunavir/r, providing guidance on how to co-administer darunavir/r with many other antiretroviral or non-antiretroviral medications commonly used in HIV-infected individuals."( Darunavir: pharmacokinetics and drug interactions.
Back, D; Hoetelmans, RM; Sekar, V, 2008
)
2
" 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
)
0.83
" 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
)
0.87
" 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
)
0.64
" 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
)
0.58
"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
)
0.58
" 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
)
0.82
" 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
)
0.61
" 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
)
0.61
" 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
)
0.59
"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,
)
1.84
"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
)
0.85
" (> 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
)
0.62
"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
)
0.85
" 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
)
0.61
"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
" 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
)
0.59
" 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
)
0.68
"Various formulations for combination of the anti-HIV protease inhibitor darunavir (DRV) and TMC41629, a pharmacokinetic booster for DRV, were studied."( Co-administration of darunavir and a new pharmacokinetic booster: formulation strategies and evaluation in dogs.
Baert, L; de Kock, H; Grooten, L; Rosier, J; Rouan, MC; Schueller, L; van 't Klooster, G; Van den Mooter, G; Van Gyseghem, E; Van Remoortere, P; Voorspoels, J, 2010
)
0.91
"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
)
0.8
"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
)
0.61
" 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
)
0.61
" 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
" 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
)
0.62
" 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
)
0.66
"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
)
0.81
" 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
)
1.01
"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
)
0.86
"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
)
0.9
" 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
)
1.22
"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
)
0.59
"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
)
0.59
" 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 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
)
0.88
" 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
)
0.89
"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
)
0.84
"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
)
0.83
"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
)
0.84
"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
)
0.64
" 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
)
0.64
" 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
)
0.74
"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
" 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
)
0.62
" 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
)
0.62
" They are characterized by highly cooperative dose-response curves that are not explained by current pharmacodynamic theory."( Multi-step inhibition explains HIV-1 protease inhibitor pharmacodynamics and resistance.
Bailey, JR; Chioma, S; Durand, CM; Laird, GM; Laskey, S; Moore, RD; Rabi, SA; Shan, L; Siliciano, RF, 2013
)
0.39
"Cobicistat (Tybost™) is a mechanism-based inhibitor of cytochrome P450 (CYP) 3A enzymes that is indicated in the EU as a pharmacokinetic enhancer (i."( Cobicistat: a review of its use as a pharmacokinetic enhancer of atazanavir and darunavir in patients with HIV-1 infection.
Deeks, ED, 2014
)
0.63
" The method was successfully applied to a single-dose pharmacokinetic study of darunavir boosted with ritonavir in Wistar rats."( LC-Q-TOF-MS/MS determination of darunavir and its metabolites in rat serum and urine: application to pharmacokinetics.
Guru Prasad, K; Nageswara Rao, R, 2014
)
0.91
" 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
)
1.64
" 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
)
0.82
" 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
)
0.88
" 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
)
0.92
" 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
)
0.68
"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
)
0.68
" Data from ARTEMIS and ODIN demonstrate non-inferiority of once-daily darunavir/ritonavir against a comparator and, together with pharmacokinetic data, have established the suitability of once-daily darunavir/ritonavir for treatment-naive and treatment-experienced patients with no darunavir resistance-associated mutations."( Pharmacokinetics and pharmacodynamics of boosted once-daily darunavir.
Brochot, A; Hoetelmans, RM; Kakuda, TN; Tomaka, FL; Van De Casteele, T; Vangeneugden, T, 2014
)
0.88
"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
)
0.88
"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
" Pharmacokinetic curves were recorded in the third trimester and post-partum."( Pharmacokinetics of total and unbound darunavir in HIV-1-infected pregnant women.
Burger, D; Colbers, A; Giaquinto, C; Gingelmaier, A; Hawkins, D; Ivanovic, J; Kabeya, K; Moltó, J; Sadiq, ST; Taylor, G; Van der Ende, M; Weizsäcker, K, 2015
)
0.69
"Darunavir AUC and Cmax were substantially decreased in pregnancy for both darunavir/ritonavir regimens."( Pharmacokinetics of total and unbound darunavir in HIV-1-infected pregnant women.
Burger, D; Colbers, A; Giaquinto, C; Gingelmaier, A; Hawkins, D; Ivanovic, J; Kabeya, K; Moltó, J; Sadiq, ST; Taylor, G; Van der Ende, M; Weizsäcker, K, 2015
)
2.13
"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
)
0.68
" 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
)
0.81
" The developed method was successfully applied to a pharmacokinetic study of (+) and (-) enantiomers of darunavir on rat dried blood spots."( Dried blood spot analysis of (+) and (-) darunavir enantiomers on immobilized amylose tris-(3, 5-dimethylphenylcarbamate) LC and its application to pharmacokinetics.
Arnipalli, MS; Nimmu, NV; Ramisetti, NR, 2015
)
0.9
"This article reviews the clinical pharmacology, pharmacodynamic and pharmacokinetic (PK) properties, clinical efficacy and tolerability, drug interactions, and dosing and administration of cobicistat."( Cobicistat: Review of a Pharmacokinetic Enhancer for HIV Infection.
Gauthier, TP; Schafer, JJ; Sherman, EM; Unger, NR; Worley, MV, 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
)
0.71
" 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
)
0.68
" 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
)
0.99
" 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
)
0.72
" 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,
)
0.64
"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
)
0.69
"Different concentration decay patterns were seen for atazanavir and darunavir, which may be partially explained by cobicistat half-life (longer with atazanavir than darunavir)."( Once-daily atazanavir/cobicistat and darunavir/cobicistat exposure over 72 h post-dose in plasma, urine and saliva: contribution to drug pharmacokinetic knowledge.
Amara, A; Boffito, M; Elliot, ER; Else, L; Higgs, C; Khoo, S; Moyle, G; Pagani, N; Schoolmeesters, A, 2017
)
0.96
" 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
)
0.86
"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
)
0.76
"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
)
0.46
" 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
)
0.46
"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
)
1
"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
)
1.02
" 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
)
0.8
" Full pharmacokinetic profiles were obtained on days 0 and 14 in the DRV cohort, and on days 0 and 7 in the ETR cohort."( Pharmacokinetics of darunavir/cobicistat and etravirine alone and co-administered in HIV-infected patients.
Challenger, E; Clotet, B; Curran, A; Khoo, S; Miranda, C; Moltó, J; Ribera, E; Santos, JR; Valle, M, 2018
)
0.8
"Etravirine co-administration decreased cobicistat AUC0-24, Cmax and C24 by 30%, 14% and 66%, respectively."( Pharmacokinetics of darunavir/cobicistat and etravirine alone and co-administered in HIV-infected patients.
Challenger, E; Clotet, B; Curran, A; Khoo, S; Miranda, C; Moltó, J; Ribera, E; Santos, JR; Valle, M, 2018
)
0.8
" The method was validated and successfully applied to everted sac and pharmacokinetic studies in rats."( HPLC Estimation, Ex vivo Everted Sac Permeability and In Vivo Pharmacokinetic Studies of Darunavir.
Sangave, PC; Suvarna, VM, 2018
)
0.7
" The method had been successfully applied to a pharmacokinetic study of fixed dose administration of PA-824, darunavir and their combination in rats."( Drug-Drug Interactions Between PA-824 and Darunavir Based on Pharmacokinetics in Rats by LC-MS-MS.
Feng, T; Jing, J; Liu, X; Mi, L; Shen, X; Wang, L; Zhang, R; Zhang, S; Zhao, J; Zhou, N, 2018
)
0.96
"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
)
0.92
" 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
)
0.77
"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
)
0.77
" These results suggest that ring placement and fit is an important species-specific study parameter that should be optimised prior to pharmacokinetic testing."( Impact of ring size and drug loading on the pharmacokinetics of a combination dapivirine-darunavir vaginal ring in cynomolgus macaques.
Boyd, P; Caldwell, A; Dereuddre-Bosquet, N; Desjardins, D; Kelly, C; Le Grand, R; Malcolm, RK; Murphy, DJ; Stimmer, L; van Roey, J, 2018
)
0.7
"This Phase I, open-label, 57 day, crossover, pharmacokinetic (PK) study, enrolled healthy volunteers aged 18-65 years, who were randomized to one of two groups."( Pharmacokinetics of dolutegravir with and without darunavir/cobicistat in healthy volunteers.
Amara, A; Bisdomini, E; Boffito, M; Cerrone, M; Elliot, ER; Else, L; Khoo, S; Owen, A, 2019
)
0.77
" The aim of this study was to develop and validate a darunavir population pharmacokinetic model based on data from daily practice."( Darunavir Population Pharmacokinetic Model Based on HIV Outpatient Data.
Alffenaar, JC; Bierman, WFW; Cattaneo, D; Daskapan, A; Gervasoni, C; Kosterink, JGW; Proost, JH; Resnati, C; Stienstra, Y; Touw, DJ; Tran, QTD; van der Werf, TS, 2019
)
2.21
" A pharmacokinetic model was developed using data from the largest data set using the iterative two-stage Bayesian procedure within the MWPharm software package."( Darunavir Population Pharmacokinetic Model Based on HIV Outpatient Data.
Alffenaar, JC; Bierman, WFW; Cattaneo, D; Daskapan, A; Gervasoni, C; Kosterink, JGW; Proost, JH; Resnati, C; Stienstra, Y; Touw, DJ; Tran, QTD; van der Werf, TS, 2019
)
1.96
"A robust population pharmacokinetic model was developed, which can support therapeutic drug monitoring of darunavir in daily outpatient settings."( Darunavir Population Pharmacokinetic Model Based on HIV Outpatient Data.
Alffenaar, JC; Bierman, WFW; Cattaneo, D; Daskapan, A; Gervasoni, C; Kosterink, JGW; Proost, JH; Resnati, C; Stienstra, Y; Touw, DJ; Tran, QTD; van der Werf, TS, 2019
)
2.17
"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
)
0.9
"Full pharmacokinetic profiling nested in a multicenter, observational, prospective cohort study."( Pharmacokinetic profiles of boosted darunavir, dolutegravir and lamivudine in aging people living with HIV.
Alves Saldanha, S; Battegay, M; Buclin, T; Cavassini, M; Courlet, P; Decosterd, LA; Guidi, M; Marzolini, C; Stader, F; Stoeckle, M, 2020
)
0.83
"Nineteen PLWH with a median age of 64 years participated in the full pharmacokinetic investigations."( Pharmacokinetic profiles of boosted darunavir, dolutegravir and lamivudine in aging people living with HIV.
Alves Saldanha, S; Battegay, M; Buclin, T; Cavassini, M; Courlet, P; Decosterd, LA; Guidi, M; Marzolini, C; Stader, F; Stoeckle, M, 2020
)
0.83
"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
)
0.79
"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
)
3.44
"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
)
1.04
" 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
)
1.27
"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
)
0.87
" 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
)
0.87
"A variety of diagnostic methods are available to validate the performance of population pharmacokinetic models."( Optimal sampling strategies for darunavir and external validation of the underlying population pharmacokinetic model.
Belkhir, L; Elens, L; Haufroid, V; Stillemans, G; Vandercam, B; Vincent, A, 2021
)
0.9
" These could be used to reliably estimate individual pharmacokinetic parameters, although with fewer samples, precision decreased and the number of outliers increased significantly."( Optimal sampling strategies for darunavir and external validation of the underlying population pharmacokinetic model.
Belkhir, L; Elens, L; Haufroid, V; Stillemans, G; Vandercam, B; Vincent, A, 2021
)
0.9
"Optimal sampling strategies derived from this model could be used in clinical practice to enhance therapeutic drug monitoring or to conduct additional pharmacokinetic studies."( Optimal sampling strategies for darunavir and external validation of the underlying population pharmacokinetic model.
Belkhir, L; Elens, L; Haufroid, V; Stillemans, G; Vandercam, B; Vincent, A, 2021
)
0.9
"Intensive steady-state 24-h pharmacokinetic profiles were performed after administration of 800 mg of darunavir and 150 mg of cobicistat orally in fixed dose combination once-daily during the second trimester, third trimester, and postpartum."( Pharmacokinetics of darunavir and cobicistat in pregnant and postpartum women with HIV.
Best, BM; Burchett, S; Capparelli, EV; Chakhtoura, N; Denson, K; Febo, IL; George, K; Mirochnick, M; Momper, JD; Paul, ME; Rungruengthanakit, K; Scott, GB; Shapiro, DE; Smith, E; Stek, A; Wang, J; Yang, DZ, 2021
)
1.16
" However, no real-world pharmacokinetic (PK) data are available in treatment-experienced patients with antiretroviral resistance receiving BIC/FTC/TAF plus a boosted protease inhibitor."( Brief Report: Pharmacokinetics of Bictegravir and Tenofovir in Combination With Darunavir/Cobicistat in Treatment-Experienced Persons With HIV.
Best, BM; Hill, L; Momper, JD; Patel, N; Salama, E, 2021
)
0.85
"This prospective, single-center, nonrandomized pharmacokinetic study enrolled adult treatment-experienced persons with HIV and creatinine clearance >30 mL/min receiving BIC/FTC/TAF + DRV/c as part of routine clinical care."( Brief Report: Pharmacokinetics of Bictegravir and Tenofovir in Combination With Darunavir/Cobicistat in Treatment-Experienced Persons With HIV.
Best, BM; Hill, L; Momper, JD; Patel, N; Salama, E, 2021
)
0.85
" The median (interquartile range [IQR]) BIC AUC0-tau,exp and Cmax values were 128."( Brief Report: Pharmacokinetics of Bictegravir and Tenofovir in Combination With Darunavir/Cobicistat in Treatment-Experienced Persons With HIV.
Best, BM; Hill, L; Momper, JD; Patel, N; Salama, E, 2021
)
0.85
"Because pharmacokinetic changes in antiretroviral drugs (ARV), due to their concurrent administration with food or nutritional products, have become a clinical challenge, it is necessary to monitor the therapeutic efficacy of ARV in people living with the human immunodeficiency virus (PLWH)."( Pharmacokinetic Outcomes of the Interactions of Antiretroviral Agents with Food and Supplements: A Systematic Review and Meta-Analysis.
Leelakanok, N; Methaneethorn, J; Siritientong, T; Thet, D, 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
)
1.13
" 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
" 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
" Peer-reviewed publications were included if they met all criteria: adults (≥ 18 years of age) living with or without HIV; report any pharmacokinetic parameter or plasma concentration of at least one of the following ARVs: tenofovir alafenamide fumarate (TAF); doravirine (DOR), rilpivirine (RIL) and etravirine (ETR); darunavir (DRV), tipranavir (TPV) and fostemsavir (FTR); dolutegravir (DTG), raltegravir (RAL), bictegravir (BIC) and elvitegravir (EVG); maraviroc (MVC); ibalizumab (IBA); cobicistat (COBI)."( Pharmacokinetics of Antiretroviral Drugs in Older People Living with HIV: A Systematic Review.
Cardoso, SW; Castro, T; Estrela, R; Grinsztejn, B; Oliveira, VG; Toledo, T; Torres, TS; Veloso, VG, 2023
)
1.08
"Among 97 studies included, 20 reported pharmacokinetic evaluation in older individuals (age ≥ 50 years)."( Pharmacokinetics of Antiretroviral Drugs in Older People Living with HIV: A Systematic Review.
Cardoso, SW; Castro, T; Estrela, R; Grinsztejn, B; Oliveira, VG; Toledo, T; Torres, TS; Veloso, VG, 2023
)
0.91

Compound-Compound Interactions

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.

ExcerptReferenceRelevance
" When brecanavir was tested in combination with nucleoside reverse transcriptase inhibitors, the antiviral activity of brecanavir was synergistic with the effects of stavudine and additive to the effects of zidovudine, tenofovir, dideoxycytidine, didanosine, adefovir, abacavir, lamivudine, and emtricitabine."( In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
Boone, L; Craig, C; Ferris, R; Furfine, E; Griffin, P; Hale, M; Hanlon, M; Harvey, R; Hazen, R; Kaldor, I; Miller, J; Ray, J; Samano, V; Spaltenstein, A; St Clair, M; Tung, R; Yates, P, 2007
)
0.34
" An extensive darunavir/r drug-drug interaction programme has been undertaken, covering a wide range of therapeutic areas."( Darunavir: pharmacokinetics and drug interactions.
Back, D; Hoetelmans, RM; Sekar, V, 2008
)
2.15
" 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
)
0.79
"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 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
)
0.81
" 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
)
0.59
"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,
)
0.37
" 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,
)
0.37
"Rodent models are less suitable for predicting drug-drug interactions at the level of the human intestinal mucosa, especially when nuclear receptors such as pregnane X receptor (PXR) are involved."( PXR/CYP3A4-humanized mice for studying drug-drug interactions involving intestinal P-glycoprotein.
Annaert, P; Augustijns, P; Baes, M; Gonzalez, FJ; Holmstock, N, 2013
)
0.39
" 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
" When ATV-LNPs were prepared with ritonavir (RTV), a metabolic and cellular membrane exporter inhibitor, and tenofovir (TFV), an HIV reverse-transcriptase inhibitor, stable, scalable, and reproducible anti-HIV drug combination LNPs were produced."( Evaluation of atazanavir and darunavir interactions with lipids for developing pH-responsive anti-HIV drug combination nanoparticles.
Duan, J; Freeling, JP; Ho, RJ; Koehn, J; Shu, C, 2014
)
0.69
"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
)
0.88
" 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
"Dolutegravir plasma trough concentrations were measured in 78 HIV-infected patients given the drug in combination with a protease inhibitor, a non-nucleoside reverse transcriptase inhibitor or abacavir/lamivudine."( Dolutegravir plasma concentrations according to companion antiretroviral drug: unwanted drug interaction or desirable boosting effect?
Cattaneo, D; Clementi, E; Cozzi, V; Galli, M; Gervasoni, C; Meraviglia, P; Minisci, D; Riva, A, 2017
)
0.46
" 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
)
0.73
"We report the cases of two treatment-experienced HIV-infected patients with complex antiretroviral regimens that showed significant drug-drug interactions with etravirine."( Optimizing concentrations of concomitant antiretrovirals by reducing etravirine doses: two case reports of complex drug-drug interactions.
Cabot, JF; Denault, JS; Langlois, H; Marcotte, S; Sheehan, NL, 2019
)
0.51
" Hence, potential drug-drug interactions (DDIs) may occur between these 2 drugs and antiretrovirals."( Drug-Drug Interactions Between Antiretrovirals and Carbamazepine/Oxcarbazepine: A Real-Life Investigation.
Atzori, C; Baldelli, S; Cattaneo, D; Cozzi, V; Filice, C; Fusi, M; Gervasoni, C; Micheli, V, 2020
)
0.56
"This was a Phase I, open-label, two cohort (n=18/cohort), fixed-sequence study in healthy females that evaluated the drug-drug interaction (DDI) between multiple-dose ATV+COBI or DRV+COBI and single-dose drospirenone/ethinyl estradiol (EE)."( Confirmation of the drug-drug interaction potential between cobicistat-boosted antiretroviral regimens and hormonal contraceptives.
Das, M; Kearney, BP; Ling, KH; Majeed, SR; West, S, 2019
)
0.51
": The risk of drug-drug interactions (DDIs) is elevated in aging people living with HIV (PLWH) because of highly prevalent age-related comorbidities leading to more comedications."( Aging does not impact drug--drug interaction magnitudes with antiretrovirals.
Battegay, M; Cavassini, M; Courlet, P; Decosterd, L; Marzolini, C; Saldanha, SA; Stader, F; Stoeckle, M, 2020
)
0.56
"Dolutegravir in combination with NRTIs was effective in treating patients with HIV-1 infection, including those with extensive NRTI resistance in whom no NRTIs were predicted to have activity."( Dolutegravir or Darunavir in Combination with Zidovudine or Tenofovir to Treat HIV.
Asienzo, J; Ategeka, G; Balyegisawa, A; Castelnuovo, B; Hakim, J; Hoppe, A; Kaimal, A; Kambugu, A; Kiragga, A; Kityo, C; Mirembe, G; Mugerwa, H; Musaazi, J; Paton, NI; Siika, A; Tukamushabe, P; Walimbwa, S, 2021
)
0.97
" 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
)
1.17
"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

Bioavailability

Darunavir has a low oral bioavailability and poor intestinal absorption. It is increased when it is coadministered in combination with low-dose (100mg) ritonavir.

ExcerptReferenceRelevance
" Darunavir is well absorbed, and the bioavailability of darunavir increases by 30% when given with food."( Darunavir: a second-generation protease inhibitor.
Busse, KH; Penzak, SR, 2007
)
2.69
" 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
)
0.85
" We initially introduced new structural templates, particulary nonpeptidic conformationally constrained P 2 ligands that would efficiently mimic peptide binding in the S 2 subsite of the protease and provide enhanced bioavailability to the inhibitor."( Design of HIV protease inhibitors targeting protein backbone: an effective strategy for combating drug resistance.
Chapsal, BD; Ghosh, AK; Mitsuya, H; Weber, IT, 2008
)
0.35
" The bioavailability of oral darunavir is increased when it is coadministered in combination with low-dose (100mg) ritonavir."( Darunavir: in the treatment of HIV-1 infection.
Fenton, C; Perry, CM, 2007
)
2.07
" The relative bioavailability of darunavir versus the reference tablet (F(rel)) was 155% with kappa-carrageenan pellets and 2% with MCC pellets without ritonavir, while 78% and 9%, respectively, in presence of ritonavir."( Improved bioavailability of darunavir by use of kappa-carrageenan versus microcrystalline cellulose as pelletisation aid.
Baert, L; Geldof, M; Kleinebudde, P; Rosier, J; Schueller, L; Thommes, M; van 't Klooster, G, 2009
)
0.93
" 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
" Coadministration with ritonavir (RTV) improves the oral bioavailability of DRV."( P-glycoprotein mediates efflux transport of darunavir in human intestinal Caco-2 and ABCB1 gene-transfected renal LLC-PK1 cell lines.
Fujimoto, H; Ghosh, AK; Hamada, A; Higuchi, M; Koh, Y; Mitsuya, H; Saito, H; Tanoue, N; Watanabe, H, 2009
)
0.61
" 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
)
0.59
" 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
)
0.6
"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
)
0.94
" Finally, data on a fixed-dose combination of 800/150 mg of darunavir/cobicistat once daily are presented, showing comparable darunavir bioavailability to that obtained with 800/100 mg of darunavir/ritonavir once daily."( Pharmacokinetics and pharmacodynamics of boosted once-daily darunavir.
Brochot, A; Hoetelmans, RM; Kakuda, TN; Tomaka, FL; Van De Casteele, T; Vangeneugden, T, 2014
)
0.89
"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
)
0.89
"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
)
0.67
"The current study was aimed to investigate the potential of solid self-nanoemulsifying drug delivery system (S-SNEDDS) composed of Capmul MCM C8 (oil), Tween 80 (surfactant) and Transcutol P (co-surfactant) in improving the dissolution and oral bioavailability of darunavir."( Solid self-nanoemulsifying drug delivery system (S-SNEDDS) of darunavir for improved dissolution and oral bioavailability: In vitro and in vivo evaluation.
Bandari, S; Dhurke, R; Eedara, BB; Inugala, S; Jukanti, R; Kandadi, P; Sunkavalli, S, 2015
)
0.84
"Darunavir is effective against wild-type and PI-resistant HIV, and has an oral bioavailability of 37%."( In-vivo bioavailability and lymphatic uptake evaluation of lipid nanoparticulates of darunavir.
Bartakke, US; Bhalekar, MR; Dube, A; Kshirsagar, SJ; Madgulkar, AR; Upadhaya, PG, 2016
)
2.1
" 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
)
0.75
" 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
)
0.89
"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
)
0.94
"The present investigation aimed to study the effect of particle size of solid lipid nanoparticles (SLNs) on oral bioavailability of darunavir."( Effect of particle size on oral bioavailability of darunavir-loaded solid lipid nanoparticles.
Desai, J; Thakkar, H, 2016
)
0.89
"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
)
0.95
"Darunavir has a low oral bioavailability (37%) due to its lipophilic nature, metabolism by cytochrome P450 enzymes and P-gp efflux."( Darunavir-Loaded Lipid Nanoparticles for Targeting to HIV Reservoirs.
Desai, J; Thakkar, H, 2018
)
3.37
"The aim of the present study was to improve bioavailability of an important antiretroviral drug, Darunavir (DRV), which has low water solubility and poor intestinal absorption through solid dispersion (SD) approach incorporating polymer with P-glycoprotein inhibitory potential."( Role of P-Glycoprotein Inhibitors in the Bioavailability Enhancement of Solid Dispersion of Darunavir.
Ahmad, S; Ali, J; Baboota, S; Bari, NK; Fazil, M; Khan, S; Kumar, V; Nabi, B; Rehman, S; Singh, R, 2017
)
0.89
" Oral bioavailability studies revealed better absorption of DRV when given with food."( Role of P-Glycoprotein Inhibitors in the Bioavailability Enhancement of Solid Dispersion of Darunavir.
Ahmad, S; Ali, J; Baboota, S; Bari, NK; Fazil, M; Khan, S; Kumar, V; Nabi, B; Rehman, S; Singh, R, 2017
)
0.68
"It is concluded that SD of DRV with the incorporation of Kolliphor TPGS was potential and promising approach in increasing bioavailability of DRV as well as minimizing its extrusion via P-glycoprotein efflux transporters."( Role of P-Glycoprotein Inhibitors in the Bioavailability Enhancement of Solid Dispersion of Darunavir.
Ahmad, S; Ali, J; Baboota, S; Bari, NK; Fazil, M; Khan, S; Kumar, V; Nabi, B; Rehman, S; Singh, R, 2017
)
0.68
"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
)
0.71
"The present work aimed to formulate Darunavir loaded lipid nanoemulsion to increase its oral bioavailability and enhance brain uptake."( Enhanced oral bioavailability and brain uptake of Darunavir using lipid nanoemulsion formulation.
Desai, J; Thakkar, H, 2019
)
1.04
"Physiological changes during pregnancy can affect pharmacokinetics and reduce a mother's bioavailability of antiretroviral drugs, potentially altering their pharmacological activity."( Low plasmatic concentration of intensified antiretroviral therapy in a pregnant woman: a case report.
Be, G; Chiesi, S; Lanzafame, M; Lattuada, E; Piacentini, D; Rizzardo, S; Tacconelli, E, 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
"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
)
0.78

Dosage Studied

This is the first study to compare darunavir CSF concentrations in patients taking the once-daily or the twice-daily dosage. The article also discusses the methods for preparation of darUnavir, its physical-chemical properties and analytical methods.

ExcerptRelevanceReference
"The pharmacology, pharmacokinetics, drug interactions, clinical efficacy, adverse events, dosage and administration, and place in therapy of darunavir are reviewed."( Darunavir: a second-generation protease inhibitor.
Busse, KH; Penzak, SR, 2007
)
1.98
" The recommended adult dosage of darunavir is 600 mg (two tablets) combined with ritonavir 100 mg every 12 hours with food."( Darunavir: a second-generation protease inhibitor.
Busse, KH; Penzak, SR, 2007
)
2.06
" The approved dosage of darunavir is 600 mg in combination with ritonavir 100mg twice daily."( Clinical pharmacokinetics of darunavir.
Arastéh, K; Rittweger, M, 2007
)
0.94
" Also discussed are the published clinical experience with darunavir, its adverse events, drug interactions, pharmacoeconomics, and dosing and administration."( Darunavir: a nonpeptidic antiretroviral protease inhibitor.
McCoy, C, 2007
)
2.03
" In conclusion, the implantable adapted Codman 3000 constant-flow infusion pump customized to anti-HIV therapy allows sustained release of anti-HIV medication and may represent an opportunity to reduce the pill burden and complexity of dosing schemes associated with common anti-HIV therapy."( Development of an implantable infusion pump for sustained anti-HIV drug administration.
Baert, L; Borghys, H; Clessens, E; Klooster, Gv; Macbride, D; Rosier, J; Schueller, L; Tardy, Y; Van Den Mooter, G; Van Gyseghem, E; Van Remoortere, P; Wigerinck, P, 2008
)
0.35
" 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
)
0.64
"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
)
0.88
" 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
)
1.26
" 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,
)
0.6
" 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
)
0.62
" 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
"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
)
0.63
" 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
)
0.86
" 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
)
0.59
" 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
)
0.68
"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
)
0.97
" The aim of this study was to formulate 800 mg of Darunavir in a single unit dosage form, with suitable mechanical properties and dissolution behavior, using a corotating twin screw extruder."( 800 mg Darunavir tablets prepared by hot melt extrusion.
Baert, L; Rosier, J; Thommes, M,
)
0.84
" 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
)
0.58
" 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
)
0.61
"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
)
0.89
" 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
)
0.37
" 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
)
0.88
"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
)
0.82
"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
)
0.63
"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
)
1
"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
)
0.7
"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
)
0.96
"Subjects with HIV infection were selected based on the use of darunavir-containing regimens with a twice-daily dosing schedule and availability of stored CSF and matched plasma."( Darunavir is predominantly unbound to protein in cerebrospinal fluid and concentrations exceed the wild-type HIV-1 median 90% inhibitory concentration.
Best, BM; Capparelli, E; Clifford, DB; Collier, AC; Croteau, D; Ellis, RJ; Gelman, BB; Grant, I; Letendre, S; Marra, CM; McArthur, J; McCutchan, JA; Morgello, S; Rossi, SS; Simpson, DM, 2013
)
2.07
" 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
)
0.59
" They are characterized by highly cooperative dose-response curves that are not explained by current pharmacodynamic theory."( Multi-step inhibition explains HIV-1 protease inhibitor pharmacodynamics and resistance.
Bailey, JR; Chioma, S; Durand, CM; Laird, GM; Laskey, S; Moore, RD; Rabi, SA; Shan, L; Siliciano, RF, 2013
)
0.39
"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
" 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
)
0.63
" 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
)
0.82
" 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
)
0.92
" 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
)
0.68
" Thus, once-daily dosing can lead to improved patient adherence to medication and, consequently, sustained virological suppression and reduction in the risk of emergence of drug resistance."( Pharmacokinetics and pharmacodynamics of boosted once-daily darunavir.
Brochot, A; Hoetelmans, RM; Kakuda, TN; Tomaka, FL; Van De Casteele, T; Vangeneugden, T, 2014
)
0.64
" This behavior of darunavir is problematic because the dosage form is exposed to different ambient conditions around the world, since HIV/AIDS is prevalent globally."( A critical review of properties of darunavir and analytical methods for its determination.
Corrêa, JC; D'Arcy, DM; Salgado, HR; Serra, CH, 2014
)
1.01
"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
)
1.06
"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
)
0.68
" 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
)
0.68
"This article reviews the clinical pharmacology, pharmacodynamic and pharmacokinetic (PK) properties, clinical efficacy and tolerability, drug interactions, and dosing and administration of cobicistat."( Cobicistat: Review of a Pharmacokinetic Enhancer for HIV Infection.
Gauthier, TP; Schafer, JJ; Sherman, EM; Unger, NR; Worley, MV, 2015
)
0.42
"With potent durability through 48 weeks, a tolerability profile comparable to other first- and second-line antiretroviral therapies, and a convenient dosing schedule with low daily pill burden in fixed-dose combination tablets, cobicistat is a potential addition to the management of HIV infection as a PK enhancer."( Cobicistat: Review of a Pharmacokinetic Enhancer for HIV Infection.
Gauthier, TP; Schafer, JJ; Sherman, EM; Unger, NR; Worley, MV, 2015
)
0.42
" 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
)
0.43
"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
)
0.69
" Elvitegravir, cobicistat and darunavir concentrations at the end of the dosing interval ( C 24 ) were quantified using a validated LC with tandem MS method."( Pharmacokinetic interactions between cobicistat-boosted elvitegravir and darunavir in HIV-infected patients.
Benmarzouk-Hidalgo, OJ; Espinosa, N; Fernandez-Magdaleno, T; Gutierrez-Valencia, A; Llaves, S; Lopez-Cortes, LF; Viciana, P, 2017
)
0.98
"The results provide evidence of similar elvitegravir and darunavir C 24 concentrations when these drugs are co-administered as co-formulated elvitegravir, cobicistat, emtricitabine and tenofovir disoproxil fumarate plus 800 mg darunavir or dosed separately."( Pharmacokinetic interactions between cobicistat-boosted elvitegravir and darunavir in HIV-infected patients.
Benmarzouk-Hidalgo, OJ; Espinosa, N; Fernandez-Magdaleno, T; Gutierrez-Valencia, A; Llaves, S; Lopez-Cortes, LF; Viciana, P, 2017
)
0.93
" This boosting effect of atazanavir could be used to optimize dolutegravir dosing in particular clinical settings."( Dolutegravir plasma concentrations according to companion antiretroviral drug: unwanted drug interaction or desirable boosting effect?
Cattaneo, D; Clementi, E; Cozzi, V; Galli, M; Gervasoni, C; Meraviglia, P; Minisci, D; Riva, A, 2017
)
0.46
"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,
)
0.78
"A rapid, simple and sensitive high-performance liquid chromatography (HPLC) method with ultraviolet detection has been developed for quantification of darunavir and raltegravir in their pharmaceutical dosage form."( A rapid validated UV-HPLC method for the simultaneous determination of the antiretroviral compounds darunavir and raltegravir in their dosage form.
Estan-Cerezo, G; García-Monsalve, A; Navarro-Ruiz, A; Rodríguez-Lucena, FJ; Soriano-Irigaray, L, 2017
)
0.87
"Determination of the darunavir and raltegravir in their dosage form was done with a maximum deviation of 4%."( A rapid validated UV-HPLC method for the simultaneous determination of the antiretroviral compounds darunavir and raltegravir in their dosage form.
Estan-Cerezo, G; García-Monsalve, A; Navarro-Ruiz, A; Rodríguez-Lucena, FJ; Soriano-Irigaray, L, 2017
)
0.99
" 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
)
2.81
" 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
)
1.03
"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
)
1.03
" Clinical pharmacology and trial data are reviewed, and the safety, efficacy and dosing of darunavir during pregnancy is discussed."( Darunavir for use in pregnant women with HIV.
Kashuba, A; Pope, R, 2017
)
2.12
"Regimen switching in virally suppressed HIV-1-infected patients may be considered to reduce pill burden or dosing frequency, decrease short- or long-term toxicity, prevent or manage drug-drug interactions, and/or decrease cost."( Dolutegravir Dual Therapy as Maintenance Treatment in HIV-Infected Patients: A Review.
Boswell, R; Foisy, MM; Hughes, CA, 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
)
0.92
" 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
)
2.18
"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
)
1.19
" 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
)
0.9
" 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
)
0.9
" 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
)
0.83
" 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
)
1.1
" 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
)
0.89
" Two Chemo-metric assisted UV-spectrophotometric methods were developed for simultaneous estimation of DRV and CBS in tablet dosage form, namely; partial least square (PLS) and Classical least square method (CLS)."( Chemo-metric assisted UV-spectrophotometric methods for simultaneous estimation of Darunavir ethanolate and Cobicistat in binary mixture and their tablet formulation.
Devi Singh, V; Kumar Singh, V, 2021
)
0.85
" The article also discusses the methods for preparation of darunavir, its physical-chemical properties, analytical methods for its determination, pharmacological properties, and dosing information."( Darunavir: A comprehensive profile.
Al-Majed, AA; Alsaif, NA; Alzaid, A; Bakheit, AH; Darwish, IA; Herqash, RN, 2021
)
2.31
"Standard darunavir/cobicistat dosing during pregnancy results in significantly lower exposure during pregnancy, which may increase the risk of virologic failure and perinatal transmission."( Pharmacokinetics of darunavir and cobicistat in pregnant and postpartum women with HIV.
Best, BM; Burchett, S; Capparelli, EV; Chakhtoura, N; Denson, K; Febo, IL; George, K; Mirochnick, M; Momper, JD; Paul, ME; Rungruengthanakit, K; Scott, GB; Shapiro, DE; Smith, E; Stek, A; Wang, J; Yang, DZ, 2021
)
1.36
" 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
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Roles (2)

RoleDescription
HIV protease inhibitorAn inhibitor of HIV protease, an enzyme required for production of proteins needed for viral assembly.
antiviral drugA substance used in the prophylaxis or therapy of virus diseases.
[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 (3)

ClassDescription
furofuranOrganic heterobicyclic compounds containing a two furan rings ortho-fused to each other.
carbamate esterAny ester of carbamic acid or its N-substituted derivatives.
sulfonamideAn amide of a sulfonic acid RS(=O)2NR'2.
[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 (116)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
TDP1 proteinHomo sapiens (human)Potency13.33590.000811.382244.6684AID686978
cytochrome P450 family 3 subfamily A polypeptide 4Homo sapiens (human)Potency0.19000.01237.983543.2770AID1645841
EWS/FLI fusion proteinHomo sapiens (human)Potency1.99230.001310.157742.8575AID1259252; AID1259253; AID1259255; AID1259256
pregnane X nuclear receptorHomo sapiens (human)Potency14.12540.005428.02631,258.9301AID1346985
estrogen nuclear receptor alphaHomo sapiens (human)Potency21.29290.000229.305416,493.5996AID743075
GVesicular stomatitis virusPotency7.56370.01238.964839.8107AID1645842
cytochrome P450 2D6Homo sapiens (human)Potency26.83700.00108.379861.1304AID1645840
nuclear receptor subfamily 1, group I, member 2Rattus norvegicus (Norway rat)Potency22.38720.10009.191631.6228AID1346983
potassium voltage-gated channel subfamily H member 2 isoform dHomo sapiens (human)Potency35.48130.01789.637444.6684AID588834
gemininHomo sapiens (human)Potency16.78890.004611.374133.4983AID624297
Interferon betaHomo sapiens (human)Potency7.56370.00339.158239.8107AID1645842
HLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)Potency7.56370.01238.964839.8107AID1645842
Inositol hexakisphosphate kinase 1Homo sapiens (human)Potency7.56370.01238.964839.8107AID1645842
cytochrome P450 2C9, partialHomo sapiens (human)Potency7.56370.01238.964839.8107AID1645842
[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, Pol PolyproteinHuman immunodeficiency virus 1Ki0.00290.00000.00290.0066AID977610
Chain A, Pol PolyproteinHuman immunodeficiency virus 1Ki0.00290.00000.00290.0066AID977610
Chain B, Pol PolyproteinHuman immunodeficiency virus 1Ki0.00290.00000.00290.0066AID977610
Chain A, Pol PolyproteinHuman immunodeficiency virus 1Ki0.00290.00000.00290.0066AID977610
Chain B, Pol PolyproteinHuman immunodeficiency virus 1Ki0.00290.00000.00290.0066AID977610
Chain A, HIV-1 ProteaseHuman immunodeficiency virus 1Ki0.00410.00330.00410.0049AID977610
Chain B, HIV-1 ProteaseHuman immunodeficiency virus 1Ki0.00410.00330.00410.0049AID977610
Chain A, proteaseHuman immunodeficiency virus 1Ki0.00410.00330.00410.0049AID977610
Chain B, proteaseHuman immunodeficiency virus 1Ki0.00410.00330.00410.0049AID977610
Chain A, ProteaseHuman immunodeficiency virus 1Ki0.00120.00100.00120.0013AID977610
Chain B, ProteaseHuman immunodeficiency virus 1Ki0.00120.00100.00120.0013AID977610
Chain A, ProteaseHuman immunodeficiency virus 1Ki0.00120.00100.00120.0013AID977610
Chain B, ProteaseHuman immunodeficiency virus 1Ki0.00120.00100.00120.0013AID977610
Chain A, ProteaseHuman immunodeficiency virus 1Ki0.00120.00100.00120.0013AID977610
Chain B, ProteaseHuman immunodeficiency virus 1Ki0.00120.00100.00120.0013AID977610
Chain A, HIV-1 ProteaseHuman immunodeficiency virus 1Ki0.00790.00160.00790.0170AID977610
Chain B, HIV-1 ProteaseHuman immunodeficiency virus 1Ki0.00790.00160.00790.0170AID977610
Chain A, HIV-1 ProteaseHuman immunodeficiency virus 1Ki0.00790.00160.00790.0170AID977610
Chain B, HIV-1 ProteaseHuman immunodeficiency virus 1Ki0.00790.00160.00790.0170AID977610
Chain A, HIV-1 ProteaseHuman immunodeficiency virus 1Ki0.00790.00160.00790.0170AID977610
Chain B, HIV-1 ProteaseHuman immunodeficiency virus 1Ki0.00790.00160.00790.0170AID977610
Chain A, HIV-1 ProteaseHuman immunodeficiency virus type 1 (BH5 ISOLATE)Ki0.00790.00160.00790.0170AID977610
Chain B, HIV-1 ProteaseHuman immunodeficiency virus type 1 (BH5 ISOLATE)Ki0.00790.00160.00790.0170AID977610
Chain A, HIV-1 ProteaseHuman immunodeficiency virus type 1 (BH5 ISOLATE)Ki0.00790.00160.00790.0170AID977610
Chain B, HIV-1 ProteaseHuman immunodeficiency virus type 1 (BH5 ISOLATE)Ki0.00790.00160.00790.0170AID977610
Chain A, HIV-1 ProteaseHuman immunodeficiency virus type 1 (BH5 ISOLATE)Ki0.00790.00160.00790.0170AID977610
Chain B, HIV-1 ProteaseHuman immunodeficiency virus type 1 (BH5 ISOLATE)Ki0.00790.00160.00790.0170AID977610
Chain A, ProteaseHuman immunodeficiency virus type 1 (BRU ISOLATE)Ki0.00190.00020.00190.0037AID977610
Chain B, ProteaseHuman immunodeficiency virus type 1 (BRU ISOLATE)Ki0.00190.00020.00190.0037AID977610
Chain A, ProteaseHuman immunodeficiency virus 1Ki0.00020.00020.00160.0032AID977610
Chain B, ProteaseHuman immunodeficiency virus 1Ki0.00020.00020.00160.0032AID977610
Chain A, ProteaseHuman immunodeficiency virus 2Ki0.00020.00020.00160.0032AID977610
Chain B, ProteaseHuman immunodeficiency virus 2Ki0.00020.00020.00160.0032AID977610
Chain A, ProteaseHuman immunodeficiency virus 1Ki0.00020.00020.00160.0032AID977610
Chain B, ProteaseHuman immunodeficiency virus 1Ki0.00020.00020.00160.0032AID977610
Chain A, ProteaseHuman immunodeficiency virus 1Ki0.00020.00020.00160.0032AID977610
Chain B, ProteaseHuman immunodeficiency virus 1Ki0.00020.00020.00160.0032AID977610
Chain A, ProteaseHuman immunodeficiency virus 1Ki0.00020.00020.00160.0032AID977610
Chain B, ProteaseHuman immunodeficiency virus 1Ki0.00020.00020.00160.0032AID977610
Chain A, HIV-1 proteaseHuman immunodeficiency virus 1IC50 (µMol)0.00280.00280.00280.0028AID977608
Chain B, HIV-1 proteaseHuman immunodeficiency virus 1IC50 (µMol)0.00280.00280.00280.0028AID977608
Chain A, HIV-1 proteaseHuman immunodeficiency virus 1IC50 (µMol)0.00280.00280.00280.0028AID977608
Chain B, HIV-1 proteaseHuman immunodeficiency virus 1IC50 (µMol)0.00280.00280.00280.0028AID977608
Chain A, HIV-1 proteaseHuman immunodeficiency virus type 1 (BRU ISOLATE)Ki0.00010.00010.00010.0001AID977610
Chain B, HIV-1 proteaseHuman immunodeficiency virus type 1 (BRU ISOLATE)Ki0.00010.00010.00010.0001AID977610
Chain A, Pol polyproteinHuman immunodeficiency virus 1Ki0.00190.00020.00190.0037AID977610
Chain B, Pol polyproteinHuman immunodeficiency virus 1Ki0.00190.00020.00190.0037AID977610
Gag-Pol polyproteinHuman immunodeficiency virus type 1 (BRU ISOLATE)Ki0.00130.00000.08283.3000AID1796146; AID1797105; AID1799362
ATP-dependent translocase ABCB1Homo sapiens (human)IC50 (µMol)43.04000.00022.318510.0000AID1604115; AID1604116; AID1604117; AID1604118; AID416864
Replicase polyprotein 1abSevere acute respiratory syndrome-related coronavirusKi518.00000.00753.00839.1100AID1805801
Replicase polyprotein 1abSevere acute respiratory syndrome coronavirus 2Ki518.00000.00001.63079.0000AID1805801
Potassium voltage-gated channel subfamily E member 1Homo sapiens (human)IC50 (µMol)183.17330.12004.048010.0000AID1207362; AID1207394
Alpha-1B adrenergic receptorRattus norvegicus (Norway rat)IC50 (µMol)100.00000.00021.874210.0000AID416864
Alpha-1D adrenergic receptorRattus norvegicus (Norway rat)IC50 (µMol)100.00000.00021.270410.0000AID416864
Alpha-1A adrenergic receptorRattus norvegicus (Norway rat)IC50 (µMol)100.00000.00001.819410.0000AID416864
Potassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)IC50 (µMol)183.17330.12004.048010.0000AID1207362; AID1207394
Potassium voltage-gated channel subfamily H member 2Homo sapiens (human)IC50 (µMol)107.19490.00091.901410.0000AID1207456; AID1207488; AID1207516
Voltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)IC50 (µMol)1,584.89000.00032.25459.6000AID1207274
Sodium channel protein type 5 subunit alphaHomo sapiens (human)IC50 (µMol)69.90530.00033.64849.2000AID1207302; AID1207334
Protease Human immunodeficiency virus 1IC50 (µMol)0.06300.00010.22487.3200AID1657083; AID1665063; AID1687689; AID1767117; AID1891872; AID1904900
Protease Human immunodeficiency virus 1Ki0.01780.00000.04433.1000AID1295708; AID1295710; AID160442; AID1658399; AID1669454; AID1778333; AID238198; AID260513; AID260514; AID260515; AID260516; AID260517; AID260518; AID269303; AID297695; AID297696; AID297697; AID297698; AID343015; AID343016; AID343017; AID343018; AID343019; AID343020; AID343021; AID390728; AID390729; AID390730; AID697744; AID697916; AID697917
Gag-Pol polyproteinHIV-1 M:K_96CM-MP535Ki0.00010.00000.00010.0002AID1800303
Potassium voltage-gated channel subfamily D member 3Homo sapiens (human)IC50 (µMol)100.00001.40005.35009.3000AID1207422
Protease Human immunodeficiency virus 1IC50 (µMol)0.00140.00000.81769.8500AID1194842; AID1491222; AID1504971; AID1520035; AID1544553; AID1575211; AID668818
Protease Human immunodeficiency virus 1Ki0.00130.00000.02841.1000AID1227235; AID1241687; AID1252235; AID1295707; AID1295709; AID1295711; AID1295712; AID1295713; AID1295714; AID1295715; AID1295716; AID1295717; AID1295718; AID1295721; AID1380922; AID1409306; AID1516788; AID1516789; AID1525498; AID1589132; AID1607525; AID1669455; AID1669456; AID297694; AID311574; AID328062; AID343014; AID390725; AID390726; AID390727; AID391263; AID396262; AID415236; AID443165; AID446190; AID459072; AID537765; AID667088; AID690521; AID690522; AID697918
[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)
Chain A, Protease (Retropepsin)Human immunodeficiency virus 1Kd3.20003.20003.20003.2000AID977611
Chain B, Protease (Retropepsin)Human immunodeficiency virus 1Kd3.20003.20003.20003.2000AID977611
Chain A, Protease (Retropepsin)Human immunodeficiency virus 1Kd3.20003.20003.20003.2000AID977611
Chain B, Protease (Retropepsin)Human immunodeficiency virus 1Kd3.20003.20003.20003.2000AID977611
Chain A, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain B, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain A, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain B, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain A, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain B, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain A, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain B, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain A, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain B, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain A, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain B, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain A, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain B, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain A, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain B, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain A, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain B, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain A, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain B, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain A, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain B, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain A, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain B, ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00060.0013AID977611
Chain A, HIV-1 proteaseHuman immunodeficiency virus 1Kd0.00000.00000.00000.0001AID977611
Chain B, HIV-1 proteaseHuman immunodeficiency virus 1Kd0.00000.00000.00000.0001AID977611
Chain A, HIV-1 proteaseHuman immunodeficiency virus 1Kd0.00000.00000.00000.0001AID977611
Chain B, HIV-1 proteaseHuman immunodeficiency virus 1Kd0.00000.00000.00000.0001AID977611
Chain A, HIV-1 ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00000.0001AID977611
Chain B, HIV-1 ProteaseHIV-1 M:B_ARV2/SF2Kd0.00000.00000.00000.0001AID977611
Chain A, ProteaseHuman immunodeficiency virus 1Kd0.00080.00080.00080.0008AID977611
Chain B, ProteaseHuman immunodeficiency virus 1Kd0.00080.00080.00080.0008AID977611
Chain A, ProteaseHuman immunodeficiency virus type 1 (BRU ISOLATE)Kd5.40005.40005.40005.4000AID977611
Chain B, ProteaseHuman immunodeficiency virus type 1 (BRU ISOLATE)Kd5.40005.40005.40005.4000AID977611
Gag-Pol polyproteinHuman immunodeficiency virus type 1 (NEW YORK-5 ISOLATE)EC50 (µMol)0.00030.00020.00380.0175AID1800111
Protease Human immunodeficiency virus 1EC50 (µMol)0.06240.00070.69422.7300AID1350492; AID1350507
Protease Human immunodeficiency virus 1Kd0.00000.00000.61178.1500AID1307690
[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)
Protease Human immunodeficiency virus 1ID500.00140.00140.85491.7000AID242819
Protease Human immunodeficiency virus 1K0.04100.03950.30460.9600AID738152
Protease Human immunodeficiency virus 1K0.00000.00000.00020.0004AID738146
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (175)

Processvia Protein(s)Taxonomy
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)
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)
symbiont-mediated perturbation of host ubiquitin-like protein modificationReplicase polyprotein 1abSevere acute respiratory syndrome-related coronavirus
epithelial cell maturationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
sensory perception of soundPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
male gonad developmentPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
vestibular nucleus developmentPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
secretory granule organizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cellular response to cAMPPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cellular response to acidic pHPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cellular response to light stimulusPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
potassium ion transmembrane transportPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cardiac muscle cell action potential involved in contractionPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cardiac muscle cell contractionPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane repolarizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane repolarization during action potentialPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
negative regulation of protein targeting to membranePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
positive regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
regulation of delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
negative regulation of delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
gastrin-induced gastric acid secretionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
glucose metabolic processPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
heart developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
sensory perception of soundPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
rhythmic behaviorPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of heart contractionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of blood pressurePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
positive regulation of heart ratePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
iodide transportPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
erythrocyte differentiationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
intracellular chloride ion homeostasisPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
response to insulinPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
social behaviorPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
corticosterone secretionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
inner ear morphogenesisPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
inner ear developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
intestinal absorptionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
detection of mechanical stimulus involved in sensory perception of soundPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
potassium ion homeostasisPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cardiac muscle contractionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
auditory receptor cell developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of membrane repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of atrial cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
positive regulation of cardiac muscle contractionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of gastric acid secretionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
stomach developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
renal absorptionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
renal sodium ion absorptionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cellular response to cAMPPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cellular response to xenobiotic stimulusPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
potassium ion transmembrane transportPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cellular response to epinephrine stimulusPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
adrenergic receptor signaling pathwayPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cardiac muscle cell contractionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane repolarization during action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
atrial cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cochlea developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane repolarization during atrial cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
positive regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
non-motile cilium assemblyPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
potassium ion import across plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of heart rate by hormonePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of membrane potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
positive regulation of DNA-templated transcriptionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion homeostasisPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cardiac muscle contractionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cellular response to xenobiotic stimulusPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane depolarization during action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
negative regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
positive regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
negative regulation of potassium ion export across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion import across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
immune system developmentVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
positive regulation of cytosolic calcium ion concentrationVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
heart developmentVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
regulation of cardiac muscle contraction by regulation of the release of sequestered calcium ionVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
embryonic forelimb morphogenesisVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
camera-type eye developmentVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
positive regulation of adenylate cyclase activityVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
positive regulation of muscle contractionVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
calcium ion transport into cytosolVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
cardiac conductionVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
calcium ion transmembrane transport via high voltage-gated calcium channelVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
calcium ion transmembrane transportVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
cardiac muscle cell action potential involved in contractionVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
membrane depolarization during cardiac muscle cell action potentialVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
membrane depolarization during AV node cell action potentialVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
cell communication by electrical coupling involved in cardiac conductionVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
regulation of heart rate by cardiac conductionVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
regulation of ventricular cardiac muscle cell action potentialVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
membrane depolarization during atrial cardiac muscle cell action potentialVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
calcium ion import across plasma membraneVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
regulation of heart rateSodium channel protein type 5 subunit alphaHomo sapiens (human)
cardiac conduction system developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
cardiac ventricle developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
brainstem developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
sodium ion transportSodium channel protein type 5 subunit alphaHomo sapiens (human)
positive regulation of sodium ion transportSodium channel protein type 5 subunit alphaHomo sapiens (human)
response to denervation involved in regulation of muscle adaptationSodium channel protein type 5 subunit alphaHomo sapiens (human)
telencephalon developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
cerebellum developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
sodium ion transmembrane transportSodium channel protein type 5 subunit alphaHomo sapiens (human)
odontogenesis of dentin-containing toothSodium channel protein type 5 subunit alphaHomo sapiens (human)
positive regulation of action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
positive regulation of epithelial cell proliferationSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
cardiac muscle contractionSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of atrial cardiac muscle cell membrane depolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of atrial cardiac muscle cell membrane repolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of ventricular cardiac muscle cell membrane depolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
cellular response to calcium ionSodium channel protein type 5 subunit alphaHomo sapiens (human)
cardiac muscle cell action potential involved in contractionSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of cardiac muscle cell contractionSodium channel protein type 5 subunit alphaHomo sapiens (human)
ventricular cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
atrial cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
SA node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
AV node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
bundle of His cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during AV node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during SA node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during Purkinje myocyte cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during bundle of His cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
AV node cell to bundle of His cell communicationSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of heart rate by cardiac conductionSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during atrial cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of sodium ion transmembrane transportSodium channel protein type 5 subunit alphaHomo 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)
potassium ion transportPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
protein homooligomerizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
membrane repolarizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
action potentialPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (82)

Processvia Protein(s)Taxonomy
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)
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)
3'-5'-RNA exonuclease activityReplicase polyprotein 1abSevere acute respiratory syndrome-related coronavirus
RNA-dependent RNA polymerase activityReplicase polyprotein 1abSevere acute respiratory syndrome-related coronavirus
cysteine-type endopeptidase activityReplicase polyprotein 1abSevere acute respiratory syndrome-related coronavirus
mRNA 5'-cap (guanine-N7-)-methyltransferase activityReplicase polyprotein 1abSevere acute respiratory syndrome-related coronavirus
mRNA (nucleoside-2'-O-)-methyltransferase activityReplicase polyprotein 1abSevere acute respiratory syndrome-related coronavirus
5'-3' RNA helicase activityReplicase polyprotein 1abSevere acute respiratory syndrome-related coronavirus
K63-linked deubiquitinase activityReplicase polyprotein 1abSevere acute respiratory syndrome-related coronavirus
K48-linked deubiquitinase activityReplicase polyprotein 1abSevere acute respiratory syndrome-related coronavirus
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
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
potassium channel regulator activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
telethonin bindingPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
protein-containing complex bindingPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
transmembrane transporter bindingPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
calmodulin bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
phosphatidylinositol-4,5-bisphosphate bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
protein phosphatase 1 bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
outward rectifier potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
protein kinase A catalytic subunit bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
protein kinase A regulatory subunit bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
transmembrane transporter bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in atrial cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
scaffold protein bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
transcription cis-regulatory region bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
inward rectifier potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
ubiquitin protein ligase bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
identical protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
protein homodimerization activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
C3HC4-type RING finger domain bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
scaffold protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
high voltage-gated calcium channel activityVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
voltage-gated calcium channel activity involved in cardiac muscle cell action potentialVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
voltage-gated calcium channel activityVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
protein bindingVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
calmodulin bindingVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
high voltage-gated calcium channel activityVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
metal ion bindingVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
alpha-actinin bindingVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
voltage-gated calcium channel activity involved in cardiac muscle cell action potentialVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
voltage-gated calcium channel activity involved in AV node cell action potentialVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
voltage-gated sodium channel activitySodium channel protein type 5 subunit alphaHomo sapiens (human)
protein bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
calmodulin bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
fibroblast growth factor bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
enzyme bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
protein kinase bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
protein domain specific bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
ankyrin bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
ubiquitin protein ligase bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
transmembrane transporter bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
nitric-oxide synthase bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in AV node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in bundle of His cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in Purkinje myocyte action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in SA node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
scaffold protein bindingSodium channel protein type 5 subunit alphaHomo 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)
A-type (transient outward) potassium channel activityPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
transmembrane transporter bindingPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
metal ion bindingPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (59)

Processvia Protein(s)Taxonomy
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)
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)
double membrane vesicle viral factory outer membraneReplicase polyprotein 1abSevere acute respiratory syndrome-related coronavirus
double membrane vesicle viral factory outer membraneReplicase polyprotein 1abSevere acute respiratory syndrome coronavirus 2
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
lysosomePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cell surfacePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
apical plasma membranePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
Z discPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane raftPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
early endosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
endoplasmic reticulumPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cytoplasmPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
lysosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
early endosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
late endosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
endoplasmic reticulumPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
basolateral plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
apical plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
transport vesiclePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cytoplasmic vesicle membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
neuron projectionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
neuronal cell bodyPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane raftPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
ciliary basePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
lumenal side of membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
basolateral part of cellPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
monoatomic ion channel complexPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cell surfacePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
perinuclear region of cytoplasmPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
inward rectifier potassium channel complexPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cytoplasmVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
plasma membraneVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
postsynaptic densityVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
membraneVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
Z discVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
dendriteVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
perikaryonVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
postsynaptic density membraneVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
L-type voltage-gated calcium channel complexVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
voltage-gated calcium channel complexVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
caveolaSodium channel protein type 5 subunit alphaHomo sapiens (human)
nucleoplasmSodium channel protein type 5 subunit alphaHomo sapiens (human)
nucleolusSodium channel protein type 5 subunit alphaHomo sapiens (human)
endoplasmic reticulumSodium channel protein type 5 subunit alphaHomo sapiens (human)
plasma membraneSodium channel protein type 5 subunit alphaHomo sapiens (human)
caveolaSodium channel protein type 5 subunit alphaHomo sapiens (human)
cell surfaceSodium channel protein type 5 subunit alphaHomo sapiens (human)
intercalated discSodium channel protein type 5 subunit alphaHomo sapiens (human)
membraneSodium channel protein type 5 subunit alphaHomo sapiens (human)
lateral plasma membraneSodium channel protein type 5 subunit alphaHomo sapiens (human)
Z discSodium channel protein type 5 subunit alphaHomo sapiens (human)
T-tubuleSodium channel protein type 5 subunit alphaHomo sapiens (human)
sarcolemmaSodium channel protein type 5 subunit alphaHomo sapiens (human)
perinuclear region of cytoplasmSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel complexSodium channel protein type 5 subunit alphaHomo 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 membranePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
sarcolemmaPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
GABA-ergic synapsePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
postsynaptic specialization membranePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
dendritic spinePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
neuronal cell bodyPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
postsynaptic membranePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (680)

Assay IDTitleYearJournalArticle
AID977610Experimentally measured binding affinity data (Ki) for protein-ligand complexes derived from PDB2013Antimicrobial agents and chemotherapy, Oct, Volume: 57, Issue:10
P2' benzene carboxylic acid moiety is associated with decrease in cellular uptake: evaluation of novel nonpeptidic HIV-1 protease inhibitors containing P2 bis-tetrahydrofuran moiety.
AID375199Antiviral activity against wild type HIV1 isolate ERS104pre infected in PHA-stimulated PBMC assessed as inhibition of p24 gap protein2009Journal of medicinal chemistry, Jul-09, Volume: 52, Issue:13
Design of HIV-1 protease inhibitors with pyrrolidinones and oxazolidinones as novel P1'-ligands to enhance backbone-binding interactions with protease: synthesis, biological evaluation, and protein-ligand X-ray studies.
AID1525498Inhibition of HIV1 protease using RE(Edans)SGIFLETSK(Dabcyl)R as substrate by fluorescence method2019Journal of medicinal chemistry, 11-14, Volume: 62, Issue:21
Overview of Recent Strategic Advances in Medicinal Chemistry.
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.
AID1230029Antiviral activity against multidrug resistant HIV1 MDR/G containing protease mutant infected in human MT4 cells assessed as inhibition of p24 Gag protein production relative to wild type2015Journal of medicinal chemistry, Jul-09, Volume: 58, Issue:13
Structure-based design of potent HIV-1 protease inhibitors with modified P1-biphenyl ligands: synthesis, biological evaluation, and enzyme-inhibitor X-ray structural studies.
AID1658400Antiviral activity against VSV-G pseudotyped HIV1 NL4-3 in human TZM-bl cells infected with virus particles extracted from human SupT1 cells infected with HIV1 NL4-3 at 10 uM incubated for 48 hrs assessed as inhibition of late stage viral infection by luc2020ACS medicinal chemistry letters, Jun-11, Volume: 11, Issue:6
Novel HIV-1 Protease Inhibitors with Morpholine as the P2 Ligand to Enhance Activity against DRV-Resistant Variants.
AID1465007Ratio of IC50 for darunavir-resistant HIV1 harboring protease L10I/I15V/K20R/L24I/V32I/M36I/M46L/L63P/V82A/L89M mutant infected in human MT4 cells to IC50 for wild type HIV1 NL4-3 infected in human MT4 cells2017Bioorganic & medicinal chemistry letters, 11-01, Volume: 27, Issue:21
Design, synthesis, X-ray studies, and biological evaluation of novel macrocyclic HIV-1 protease inhibitors involving the P1'-P2' ligands.
AID649700Antiviral activity against HIV1 MDR/TM harboring protease L10I, K14R, R41K, M46L, I54V, L63P, A71V, V82A, L90M, I93L mutant infected in PHA-PBM cells assessed as inhibition of p24 Gag protein production2012Bioorganic & medicinal chemistry letters, Mar-15, Volume: 22, Issue:6
Substituent effects on P2-cyclopentyltetrahydrofuranyl urethanes: design, synthesis, and X-ray studies of potent HIV-1 protease inhibitors.
AID1482920Antiviral activity against indinavir-resistant HIV1 NL4-3 harboring protease L10F/L24I/M46I/I54V/L63P/A71V/G73S/V82T mutant infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by chemiluminescent enzyme immunoassay2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID390725Inhibition of wild type HIV1 protease by uncompetitive binding2008Journal of medicinal chemistry, Oct-23, Volume: 51, Issue:20
Solution kinetics measurements suggest HIV-1 protease has two binding sites for darunavir and amprenavir.
AID260516Binding affinity to HIV1 protease V82A2006Journal of medicinal chemistry, Feb-23, Volume: 49, Issue:4
Effectiveness of nonpeptide clinical inhibitor TMC-114 on HIV-1 protease with highly drug resistant mutations D30N, I50V, and L90M.
AID564040Antiviral activity against HIV1 expressing protease L10F/M46I/I54V/V82A mutant infected in human MT4 cells selected at 5 uM of Lopinavir by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
AID1604131Antiviral activity against HIV1 LAI infected in human 12D7 cells overexpressing p-gp assessed as reduction in P24 level at 1:1 ratio of test compound to abacavir incubated for 3 days followed by followed by replacement of fresh medium containing compound 2020Journal of medicinal chemistry, 03-12, Volume: 63, Issue:5
Potential Tools for Eradicating HIV Reservoirs in the Brain: Development of Trojan Horse Prodrugs for the Inhibition of P-Glycoprotein with Anti-HIV-1 Activity.
AID1633412Antiviral activity against wild-type HIV1 NL4-3 infected infected in human MT2 cells assessed as reduction in intracellular RNA at 1 uM and measured after 30 hrs post infection by Alu-LTR PCR protocol based method2019ACS medicinal chemistry letters, Apr-11, Volume: 10, Issue:4
Synthesis and Evaluation of Bifunctional Aminothiazoles as Antiretrovirals Targeting the HIV-1 Nucleocapsid Protein.
AID1241687Inhibition of HIV1 protease2015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
Design of HIV-1 Protease Inhibitors with Amino-bis-tetrahydrofuran Derivatives as P2-Ligands to Enhance Backbone-Binding Interactions: Synthesis, Biological Evaluation, and Protein-Ligand X-ray Studies.
AID1725423Antiviral activity against HIV1 DRVR P51 infected in human MT4 cells incubated for 7 days by fully automated chemiluminescent enzyme immunoassay2020ACS medicinal chemistry letters, Oct-08, Volume: 11, Issue:10
Design, Synthesis, and X-ray Studies of Potent HIV-1 Protease Inhibitors with P2-Carboxamide Functionalities.
AID236642Maximum plasma concentration in rat after 80 mg/kg oral administration2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Discovery and selection of TMC114, a next generation HIV-1 protease inhibitor.
AID396255Ratio of EC50 for HIV1 HXB2 in presence of 40% fetal bovine serum to EC50 for HIV1 HXB2 in presence of 10% fetal bovine serum2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
AID297694Inhibition of wild type HIV1 protease2007Journal of medicinal chemistry, Sep-06, Volume: 50, Issue:18
Potent new antiviral compound shows similar inhibition and structural interactions with drug resistant mutants and wild type HIV-1 protease.
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.
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.
AID1207302Inhibition of fast sodium current (INa) in Chinese Hamster Ovary (CHO) K1 cells transfected with human Nav1.5 measured using IonWorks Quattro automated patch clamp platform
AID519786Antiviral activity against HIV 2 subtype A clinical isolate expressing 10I-17D-40D-43I-45K/R-46V-54M-64I/V-69K/R-71V/I-90M protease gene sequence from HIV2 infected patient plasma and PBMC obtained at T1 during compound treatment measured after 13 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
AID673370Inhibition of Human immunodeficiency virus protease M10 mutant expressed in Escherichia coli using ArgGlu(EDANS)SerGlnAsnTyrProIleValGlnLys(DABCYL)Arg as substrate preincubated with compound for 0.5 to 1 min measured by fluorometric analysis at pH 6.52012Bioorganic & medicinal chemistry letters, Aug-01, Volume: 22, Issue:15
Enamino-oxindole HIV protease inhibitors.
AID1904900Inhibition of HIV-1 protease using Arg-Glu (EDANS)-Ser-Gln-Asn-Tyr-Pro-lle-Val-Gln-Lys (DABCYL)-Arg as substrate preincubated for 20 to 30 mins followed by substrate addition and measured by FRET assay2022European journal of medicinal chemistry, Apr-05, Volume: 233Design, synthesis and biological evaluation of protease inhibitors containing morpholine cores with remarkable potency against both HIV-1 subtypes B and C.
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.
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.
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.
AID1350493Antiviral activity against wild-type HIV1 pNL4-3 infected in CMVdeltaR8.91 transfected 293T cells after 48 hrs by lentiviral vector-based luciferase reporter gene assay2018Journal of medicinal chemistry, 06-28, Volume: 61, Issue:12
Identification of Highly Potent Human Immunodeficiency Virus Type-1 Protease Inhibitors against Lopinavir and Darunavir Resistant Viruses from Allophenylnorstatine-Based Peptidomimetics with P2 Tetrahydrofuranylglycine.
AID673373Antiviral activity against Human immunodeficiency virus 1 HXB2 expressing protease M9 mutant infected in human MT4 cells after 5 days by MTT assay2012Bioorganic & medicinal chemistry letters, Aug-01, Volume: 22, Issue:15
Enamino-oxindole HIV protease inhibitors.
AID537767Antiviral activity against wild type Human immunodeficiency virus 12010Journal of medicinal chemistry, Nov-11, Volume: 53, Issue:21
Structure-based design, synthesis, and structure-activity relationship studies of HIV-1 protease inhibitors incorporating phenyloxazolidinones.
AID658567Resistance factor, ratio of EC50 for multidrug-resistant HIV1 106-PR infected in HEK293T cells to wildtype HIV1 infected in HEK293T cells2012Journal of natural products, Mar-23, Volume: 75, Issue:3
Library-based discovery and characterization of daphnane diterpenes as potent and selective HIV inhibitors in Daphne gnidium.
AID649698Antiviral activity against HIV1 MDR/C harboring protease L10I, I15V, K20R, L24I, M36I, M46L, I54V, I62V, L63P, K70Q, V82A, and L89 mutant infected in PHA-PBM cells assessed as inhibition of p24 Gag protein production2012Bioorganic & medicinal chemistry letters, Mar-15, Volume: 22, Issue:6
Substituent effects on P2-cyclopentyltetrahydrofuranyl urethanes: design, synthesis, and X-ray studies of potent HIV-1 protease inhibitors.
AID249508Effective concentration of against HIV Wild Type (IIIB) strain2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Discovery and selection of TMC114, a next generation HIV-1 protease inhibitor.
AID519780Antiviral activity against HIV 2 subtype H expressing 10I-34E-40P-41Y-60H-63N-70T-73G-82F-89L-92E protease gene sequence from HIV2 infected patient plasma and PBMC obtained at T1 during compound treatment measured after 13 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
AID237956Percentage of remaining compound after 30 min incubation in human liver microsomes was determined as metabolic stability2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Discovery and selection of TMC114, a next generation HIV-1 protease inhibitor.
AID297697Inhibition of HIV1 protease V82A mutant2007Journal of medicinal chemistry, Sep-06, Volume: 50, Issue:18
Potent new antiviral compound shows similar inhibition and structural interactions with drug resistant mutants and wild type HIV-1 protease.
AID260513Binding affinity to HIV1 protease2006Journal of medicinal chemistry, Feb-23, Volume: 49, Issue:4
Effectiveness of nonpeptide clinical inhibitor TMC-114 on HIV-1 protease with highly drug resistant mutations D30N, I50V, and L90M.
AID396259Antiviral activity against HIV1 drug resistant mutant isolates from protease inhibitor treated HIV patient2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
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.
AID697916Inhibition of HIV1 protease L10I, L63P, A71V, G73S, I84V, L90M mutant by FRET assay2012Journal of medicinal chemistry, Jul-26, Volume: 55, Issue:14
Design, synthesis, and biological and structural evaluations of novel HIV-1 protease inhibitors to combat drug resistance.
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.
AID343015Inhibition of HIV1 recombinant protease D30N/N88D mutant expressed in Escherichia coli by spectrophotometric assay2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
Inorganic polyhedral metallacarborane inhibitors of HIV protease: a new approach to overcoming antiviral resistance.
AID396251Ratio of EC50 for HIV1 HXB2 in presence of 10% human serum to EC50 for HIV1 HXB2 in presence of 10% fetal bovine serum2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
AID1482911Antiviral activity against wild type HIV1 NL4-3 infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by chemiluminescent enzyme immunoassay2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
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.
AID237560Area under concentration time curve value in dog after 80 mg/kg oral administration2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Discovery and selection of TMC114, a next generation HIV-1 protease inhibitor.
AID668809Antiviral activity against Human immunodeficiency virus 1 isolate M1 expressing protease L10I, M46I, I64V, I84V, L90M, I93L mutant infected in human MT4 cells2011ACS medicinal chemistry letters, Jun-09, Volume: 2, Issue:6
Disubstituted Bis-THF Moieties as New P2 Ligands in Nonpeptidal HIV-1 Protease Inhibitors.
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.
AID236938Half-life in rat after 80 mg/kg oral administration2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Discovery and selection of TMC114, a next generation HIV-1 protease inhibitor.
AID391263Inhibition of HIV1 protease2008Journal 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.
AID297698Inhibition of HIV1 protease I84V mutant2007Journal of medicinal chemistry, Sep-06, Volume: 50, Issue:18
Potent new antiviral compound shows similar inhibition and structural interactions with drug resistant mutants and wild type HIV-1 protease.
AID328065Inhibition of HIV1 protease dimerization in COS7 cells at 1 uM assessed as decrease in luciferase response ratio2007The Journal of biological chemistry, Sep-28, Volume: 282, Issue:39
Potent inhibition of HIV-1 replication by novel non-peptidyl small molecule inhibitors of protease dimerization.
AID1380929Resistance index, ratio of EC50 for antiviral activity against LPV resistant HIV1 harboring protease L10F/M46/I54V/V82A mutant infected in human MT4 cells to EC50 for antiviral activity against wild type HIV1 NL4-3 infected in human MT4 cells2018Journal of medicinal chemistry, 05-24, Volume: 61, Issue:10
Design and Synthesis of Highly Potent HIV-1 Protease Inhibitors Containing Tricyclic Fused Ring Systems as Novel P2 Ligands: Structure-Activity Studies, Biological and X-ray Structural Analysis.
AID1778343Antiviral activity against HIV1 subtype C isolate 119292021European journal of medicinal chemistry, Aug-05, Volume: 220Design and evaluation of novel piperidine HIV-1 protease inhibitors with potency against DRV-resistant variants.
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.
AID1380928Resistance index, ratio of EC50 for antiviral activity against ATV resistant HIV1 harboring protease L23I/E34Q/K43I/M46I/I50L/G51A/L63P/A71V/V82A/T91A mutant infected in human MT4 cells to EC50 for antiviral activity against wild type HIV1 NL4-3 infected 2018Journal of medicinal chemistry, 05-24, Volume: 61, Issue:10
Design and Synthesis of Highly Potent HIV-1 Protease Inhibitors Containing Tricyclic Fused Ring Systems as Novel P2 Ligands: Structure-Activity Studies, Biological and X-ray Structural Analysis.
AID668816Selectivity index, ratio of CC50 for human MT4 cells to EC50 for Human immunodeficiency virus 1 3B2011ACS medicinal chemistry letters, Jun-09, Volume: 2, Issue:6
Disubstituted Bis-THF Moieties as New P2 Ligands in Nonpeptidal HIV-1 Protease Inhibitors.
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.
AID519787Antiviral activity against HIV 2 subtype A clinical isolate expressing 14H-40D-70K-72R/K-91T/S protease gene sequence from HIV2 infected patient plasma and PBMC obtained before compound treatment measured after 13 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
AID446201Selectivity ratio of IC50 for multidrug-resistant HIV1 isolate C to IC50 for wild type HIV1 isolate ERS104pre2009Journal of medicinal chemistry, Dec-10, Volume: 52, Issue:23
Design, synthesis, protein-ligand X-ray structure, and biological evaluation of a series of novel macrocyclic human immunodeficiency virus-1 protease inhibitors to combat drug resistance.
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.
AID564037Antiviral activity against HIV1 expressing protease L10I/G48V/I54V/A71V/I84V/L90M mutant infected in human MT4 cells selected at 5 uM of saquinavir by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
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.
AID668818Inhibition of Human immunodeficiency virus 1 3B protease2011ACS medicinal chemistry letters, Jun-09, Volume: 2, Issue:6
Disubstituted Bis-THF Moieties as New P2 Ligands in Nonpeptidal HIV-1 Protease Inhibitors.
AID1504972Antiviral activity against HIV1 infected in GFP expressing human MT4 cells assessed as inhibition of viral replication after 24 hrs in presence of 50% normal human serum2017ACS medicinal chemistry letters, Dec-14, Volume: 8, Issue:12
Design and Synthesis of Piperazine Sulfonamide Cores Leading to Highly Potent HIV-1 Protease Inhibitors.
AID770191Antiviral activity against wild type HIV1 ERS104pre infected in human PHA-PBMC cells assessed as inhibition of p24 Gag protein production2013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Highly potent HIV-1 protease inhibitors with novel tricyclic P2 ligands: design, synthesis, and protein-ligand X-ray studies.
AID1460920Inhibition of HIV1 protease using fluorogenic substrate by continuous fluorometric assay2017Bioorganic & medicinal chemistry, 10-01, Volume: 25, Issue:19
Design of novel HIV-1 protease inhibitors incorporating isophthalamide-derived P2-P3 ligands: Synthesis, biological evaluation and X-ray structural studies of inhibitor-HIV-1 protease complex.
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.
AID1544553Inhibition of wild type HIV1 protease expressed in Escherichia coli using Arg-Glu (EDANS)-Ser-Gln-Asn-Tyr-Pro-Ile-Val-Gln-Lys(DABCYL)-Arg as substrate preincubated for 20 to 30 mins followed by substrate addition and measured for 10 mins by FRET assay2019Bioorganic & medicinal chemistry letters, 06-15, Volume: 29, Issue:12
Synthesis and biological evaluation of new HIV-1 protease inhibitors with purine bases as P2-ligands.
AID1891874Selectivity index, ratio of CC50 for HEK293T cells to IC50 for inhibition of HIV1 protease expressed in Escherichia coli2022Bioorganic & medicinal chemistry, 06-15, Volume: 64A kind of HIV-1 protease inhibitors containing phenols with antiviral activity against DRV-resistant variants.
AID1241692Antiviral activity against darunavir-resistant HIV1 P51 infected in human MT4 cells by XTT assay2015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
Design of HIV-1 Protease Inhibitors with Amino-bis-tetrahydrofuran Derivatives as P2-Ligands to Enhance Backbone-Binding Interactions: Synthesis, Biological Evaluation, and Protein-Ligand X-ray Studies.
AID237561Area under concentration time curve value in rat after 80 mg/kg oral administration2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Discovery and selection of TMC114, a next generation HIV-1 protease inhibitor.
AID459084Ratio of IC50 for multi drug-resistant HIV1 isolate of subtype MM to IC50 for wild type HIV1 isolate ERS104pre2010Bioorganic & medicinal chemistry letters, Feb-01, Volume: 20, Issue:3
Synthesis and biological evaluation of novel allophenylnorstatine-based HIV-1 protease inhibitors incorporating high affinity P2-ligands.
AID1778336Selectivity index, ratio of CC50 for cytotoxicity against HEK293T cells to IC50 of antiviral activity against HIV12021European journal of medicinal chemistry, Aug-05, Volume: 220Design and evaluation of novel piperidine HIV-1 protease inhibitors with potency against DRV-resistant variants.
AID160442Inhibitory activity against HIV-1 protease1998Bioorganic & medicinal chemistry letters, Mar-17, Volume: 8, Issue:6
Potent HIV protease inhibitors incorporating high-affinity P2-ligands and (R)-(hydroxyethylamino)sulfonamide isostere.
AID1409310Antiviral activity against amprenavir-resistant HIV1 infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by automated chemiluminescent enzyme immunoassay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Design and Synthesis of Potent HIV-1 Protease Inhibitors Containing Bicyclic Oxazolidinone Scaffold as the P2 Ligands: Structure-Activity Studies and Biological and X-ray Structural Studies.
AID1658402Antiviral activity against VSV-G pseudotyped darunavir-resistant HIV1 NL4-3 in human SupT1 cells infected with supernatants of virus particles extracted from human 293T cells infected with darunavir-resistant HIV1 NL4-3 incubated for 48 hrs assessed as in2020ACS medicinal chemistry letters, Jun-11, Volume: 11, Issue:6
Novel HIV-1 Protease Inhibitors with Morpholine as the P2 Ligand to Enhance Activity against DRV-Resistant Variants.
AID1516792Antiviral activity against multidrug-resistant HIV1 NL4-3 VSL20 infected in human TZM-bl cells preincubated for 18 hrs followed by viral infection and measured after 48 hrs by luciferase reporter gene assay2019Journal of medicinal chemistry, 09-12, Volume: 62, Issue:17
HIV-1 Protease Inhibitors Incorporating Stereochemically Defined P2' Ligands To Optimize Hydrogen Bonding in the Substrate Envelope.
AID416853Inhibition of human MDR1-dependent accumulation of calcein-AM expressed in MDCK2 cells at 50 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.
AID416866Effect on tenofovir disoproxil fumarate metabolism in ritonavir booster drug treated healthy human assessed as change in plasma AUC of tenofovir at 300 mg, po, BID co-administered with 300 mg once daily dose of tenofovir disoproxil fumarate2007Antimicrobial 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.
AID521548Antiviral activity against Human immunodeficiency virus type 1 (BRU ISOLATE) after 13 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
AID396250Antiviral activity against HIV1 HXB2 in human MT4 cells assessed as inhibition of viral-induced viral cytopathic effect in presence of 10% fetal bovine serum and 40% human serum by MTS assay2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
AID525493Cmin in HIV-infected patient2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
Antimalarial asexual stage-specific and gametocytocidal activities of HIV protease inhibitors.
AID564046Antiviral activity against multidrug-resistant HIV1 isolate TM containing L10I, K14R, R41K, M46L, I54V, L63P, A71V, V82A, L90M, and I93L mutant infected in human PHA-PBMC cells by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
AID1516788Inhibition of HIV1 NL4-3 protease I84V mutant expressed in Escherichia coli TAP-106 cells using EDANS/DABCYL-labelled 10-amino acid containing natural MA/CA cleavage site as substrate preincubated for 1 hr followed by substrate addition and measured for 62019Journal of medicinal chemistry, 09-12, Volume: 62, Issue:17
HIV-1 Protease Inhibitors Incorporating Stereochemically Defined P2' Ligands To Optimize Hydrogen Bonding in the Substrate Envelope.
AID564064Ratio of EC50 for HIV1 expressing protease L10I/L24I/M46I/V82I/I84V mutant infected in human MT4 cells selected after 50 passages of GRL-216 to EC50 for HIV1 NL4-3 infected in human MT4 cells2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
AID1665068Antiviral activity against VSV-G pseudotyped wild type HIV1 NL4-3 infected in human TZM-b1 cells at 100 nM infected with supernatants from virus-infected human SUPT1 cells after 48 hrs by luciferase assay relative to control2020Bioorganic & medicinal chemistry, 08-15, Volume: 28, Issue:16
Design and biological evaluation of novel HIV-1 protease inhibitors with isopropanol as P1' ligand to enhance binding with S1' subsite.
AID564039Antiviral activity against HIV1 expressing protease L10F/M46I/I50V/A71V/I84V/L90M mutant infected in human MT4 cells selected at 5 uM of amprenavir by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
AID668814Selectivity index, ratio of EC50 for Human immunodeficiency virus 1 isolate M3 expressing protease L10I, K20R, M36I, G48V, I62V, A71V, V82A, I93L mutant to EC50 for Human immunodeficiency virus 1 3B expressing wild-type protease2011ACS medicinal chemistry letters, Jun-09, Volume: 2, Issue:6
Disubstituted Bis-THF Moieties as New P2 Ligands in Nonpeptidal HIV-1 Protease Inhibitors.
AID613964Ratio of IC50 for Human immunodeficiency virus 1 MDR/TM to IC50 for wild-type Human immunodeficiency virus 1 ERS104pre2011Journal of medicinal chemistry, Aug-25, Volume: 54, Issue:16
Design of HIV-1 protease inhibitors with C3-substituted hexahydrocyclopentafuranyl urethanes as P2-ligands: synthesis, biological evaluation, and protein-ligand X-ray crystal structure.
AID260517Binding affinity to HIV1 protease I84V2006Journal of medicinal chemistry, Feb-23, Volume: 49, Issue:4
Effectiveness of nonpeptide clinical inhibitor TMC-114 on HIV-1 protease with highly drug resistant mutations D30N, I50V, and L90M.
AID521547Antiviral activity against Human immunodeficiency virus type 2 (ISOLATE ROD) after 13 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
AID1482927Ratio of IC50 for tipranavir-resistant HIV1 NL4-3 harboring protease L10I/L33I/M36I/M46I/I54V/K55R/I62V/L63P/A71V/G73S/V82T/L90M/I93L mutant infected in human MT4 cells to IC50 for wild type HIV1 NL4.3 infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
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.
AID613957Antiviral activity against multidrug-resistant Human immunodeficiency virus 1 MDR/C harboring protease L10I, I15 V, K20R, L24I, M36I, M46L, I54V, I62 V, L63P, K70Q, V82A, L89 M mutation infected in PHA-stimulated human PBMC assessed as inhibition of p24 G2011Journal of medicinal chemistry, Aug-25, Volume: 54, Issue:16
Design of HIV-1 protease inhibitors with C3-substituted hexahydrocyclopentafuranyl urethanes as P2-ligands: synthesis, biological evaluation, and protein-ligand X-ray crystal structure.
AID566847Antiviral activity against HIV-1 MDR/B infected in human PHA-PBM cells assessed as inhibition of p24 Gag protein production2011Journal of medicinal chemistry, Jan-27, Volume: 54, Issue:2
Design and synthesis of potent HIV-1 protease inhibitors incorporating hexahydrofuropyranol-derived high affinity P(2) ligands: structure-activity studies and biological evaluation.
AID726412Selectivity ratio of EC50 for Human immunodeficiency virus 1 3B clinical isolate harboring L10I/I13V/M46I/I50V/L63P/L76V protease mutant to EC50 for wild type Human immunodeficiency virus 1 3B2013Bioorganic & medicinal chemistry letters, Jan-01, Volume: 23, Issue:1
Design and synthesis of HIV-1 protease inhibitors for a long-acting injectable drug application.
AID564063Antiviral activity against HIV1 expressing protease L10F/M46M,I/Q61Q mutant infected in human MT4 cells selected at 1 uM of GRL-396 by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
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.
AID446196Antiviral activity against multidrug-resistant HIV1 isolate G infected in PHA-stimulated human PBMC assessed as inhibition of p24 gap protein production2009Journal of medicinal chemistry, Dec-10, Volume: 52, Issue:23
Design, synthesis, protein-ligand X-ray structure, and biological evaluation of a series of novel macrocyclic human immunodeficiency virus-1 protease inhibitors to combat drug resistance.
AID564032Cytotoxicity against human MT2 cells after 7 days by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
AID415283Antiviral activity against HIV1 bearing protease gene with P25 mutation infected in human MT4 cells after 5 days by MTT assay2009Journal 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.
AID311576Antiviral activity against HIV1 in MT4 cells2007Bioorganic & medicinal chemistry, Dec-15, Volume: 15, Issue:24
Darunavir, a conceptually new HIV-1 protease inhibitor for the treatment of drug-resistant HIV.
AID417037Effect on tenofovir disoproxil fumarate metabolism in ritonavir booster drug treated healthy human assessed as change in plasma Cmin of tenofovir at 300 mg, po, BID co-administered with 300 mg once daily dose of tenofovir disoproxil fumarate2007Antimicrobial 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.
AID1516789Inhibition of HIV1 NL4-3 protease I50V/A71V mutant expressed in Escherichia coli TAP-106 cells using EDANS/DABCYL-labelled 10-amino acid containing natural MA/CA cleavage site as substrate preincubated for 1 hr followed by substrate addition and measured 2019Journal of medicinal chemistry, 09-12, Volume: 62, Issue:17
HIV-1 Protease Inhibitors Incorporating Stereochemically Defined P2' Ligands To Optimize Hydrogen Bonding in the Substrate Envelope.
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.
AID1482919Antiviral activity against saquinavir-resistant HIV1 NL4-3 harboring protease L10I/N37D/G48V/I54V/L63P/G73C/I84V/L90M mutant infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by chemiluminescent enzyme immunoassa2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID697740Antiviral activity against wild type HIV1 clade C isolated form HIV1 patient infected in HEK293 cells assessed as reduction in viral replication incubated for 48 hrs by luciferase reporter based assay2012Journal of medicinal chemistry, Jul-26, Volume: 55, Issue:14
Design, synthesis, and biological and structural evaluations of novel HIV-1 protease inhibitors to combat drug resistance.
AID446197Antiviral activity against multidrug-resistant HIV1 isolate TM infected in PHA-stimulated human PBMC assessed as inhibition of p24 gap protein production2009Journal of medicinal chemistry, Dec-10, Volume: 52, Issue:23
Design, synthesis, protein-ligand X-ray structure, and biological evaluation of a series of novel macrocyclic human immunodeficiency virus-1 protease inhibitors to combat drug resistance.
AID566848Antiviral activity against HIV-1 MDR/C infected in human PHA-PBM cells assessed as inhibition of p24 Gag protein production2011Journal of medicinal chemistry, Jan-27, Volume: 54, Issue:2
Design and synthesis of potent HIV-1 protease inhibitors incorporating hexahydrofuropyranol-derived high affinity P(2) ligands: structure-activity studies and biological evaluation.
AID249506Effective concentration against PI-Resistant HIV strain (M4)2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Discovery and selection of TMC114, a next generation HIV-1 protease inhibitor.
AID697917Inhibition of HIV1 protease L10I, G48V, I54V, L63P and V82A mutant by FRET assay2012Journal of medicinal chemistry, Jul-26, Volume: 55, Issue:14
Design, synthesis, and biological and structural evaluations of novel HIV-1 protease inhibitors to combat drug resistance.
AID738146Binding affinity to HIV1 protease expressed in Escherichia coli BL21 (DE3) assessed as dissociation constant by isothermal titration calorimetric analysis2013Journal of medicinal chemistry, May-23, Volume: 56, Issue:10
Extreme multidrug resistant HIV-1 protease with 20 mutations is resistant to novel protease inhibitors with P1'-pyrrolidinone or P2-tris-tetrahydrofuran.
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.
AID1482907Ratio of IC50 for saquinavir-resistant HIV1 NL4-3 harboring protease L10I/N37D/G48V/I54V/L63P/G73C/I84V/L90M mutant infected in human MT4 cells to IC50 for wild type HIV1 NL4.3 infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID1241693Resistance index, ratio of EC50 for darunavir-resistant HIV1 P10 infected in human MT4 cells by XTT assay to EC50 for HIV1 NL4-3 infected in human MT4 cells by XTT assay2015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
Design of HIV-1 Protease Inhibitors with Amino-bis-tetrahydrofuran Derivatives as P2-Ligands to Enhance Backbone-Binding Interactions: Synthesis, Biological Evaluation, and Protein-Ligand X-ray Studies.
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.
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.
AID1594347Inhibition of GFP-tagged HIV1 protease transfected in HEK293 cells at 10 to 30 uM incubated for 24 hrs by FACS analysis2019Bioorganic & medicinal chemistry, 05-01, Volume: 27, Issue:9
New heteroaryl carbamates: Synthesis and biological screening in vitro and in mammalian cells of wild-type and mutant HIV-protease inhibitors.
AID391264Antiviral activity against HIV1 LAI in human MT2 cells assessed as inhibition of virus-induced cytopathic effect2008Journal 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.
AID236973Time to reach maximum concentration in rat after 80 mg/kg oral administration2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Discovery and selection of TMC114, a next generation HIV-1 protease inhibitor.
AID1607532Selectivity ratio of IC50 for antiviral activity against lopinavir resistant HIV1 harboring protease L10F/V32I/M46I/I47A/A71V/I84V mutant infected in human MT4 cells to IC50 for antiviral activity against wild-type HIV1 NL4-32020Journal of medicinal chemistry, 05-14, Volume: 63, Issue:9
Structure-Based Design of Highly Potent HIV-1 Protease Inhibitors Containing New Tricyclic Ring P2-Ligands: Design, Synthesis, Biological, and X-ray Structural Studies.
AID519542Antiviral activity against HIV1 B26 infected in human MT4 cells harboring protease L33F, K45I, M46I, I50V, I54V, A71V, and V82F mutation derived from viral passages with Lopinavir assessed as reduction in viral cytopathogenicity treated 1 hr post infectio2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
Characterization of a novel human immunodeficiency virus type 1 protease inhibitor, A-790742.
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.
AID519782Antiviral activity against HIV 2 subtype A clinical isolate expressing 5L/F-14Y/H-17G/D-43T-54I/M-62V/A-70R/K-71I protease gene sequence from HIV2 infected patient plasma and PBMC obtained at T1 during compound treatment measured after 13 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
AID537765Inhibition of wild type HIV1 protease by FRET2010Journal of medicinal chemistry, Nov-11, Volume: 53, Issue:21
Structure-based design, synthesis, and structure-activity relationship studies of HIV-1 protease inhibitors incorporating phenyloxazolidinones.
AID690522Inhibition of HIV1 protease2012Journal of medicinal chemistry, Jun-28, Volume: 55, Issue:12
Dual inhibitors for aspartic proteases HIV-1 PR and renin: advancements in AIDS-hypertension-diabetes linkage via molecular dynamics, inhibition assays, and binding free energy calculations.
AID566846Antiviral activity against wild type HIV-1 ERS104 pre infected in human PHA-PBM cells assessed as inhibition of p24 Gag protein production2011Journal of medicinal chemistry, Jan-27, Volume: 54, Issue:2
Design and synthesis of potent HIV-1 protease inhibitors incorporating hexahydrofuropyranol-derived high affinity P(2) ligands: structure-activity studies and biological evaluation.
AID390730Inhibition of HIV1 protease V32I mutant by competitive binding2008Journal of medicinal chemistry, Oct-23, Volume: 51, Issue:20
Solution kinetics measurements suggest HIV-1 protease has two binding sites for darunavir and amprenavir.
AID375207Ratio of EC50 for multidrug-resistant HIV1 isolate G to EC50 for wild type HIV1 isolate ERS104pre2009Journal of medicinal chemistry, Jul-09, Volume: 52, Issue:13
Design of HIV-1 protease inhibitors with pyrrolidinones and oxazolidinones as novel P1'-ligands to enhance backbone-binding interactions with protease: synthesis, biological evaluation, and protein-ligand X-ray studies.
AID459085Ratio of IC50 for multi drug-resistant HIV1 isolate of subtype JSL to IC50 for wild type HIV1 isolate ERS104pre2010Bioorganic & medicinal chemistry letters, Feb-01, Volume: 20, Issue:3
Synthesis and biological evaluation of novel allophenylnorstatine-based HIV-1 protease inhibitors incorporating high affinity P2-ligands.
AID375202Antiviral activity against multidrug-resistant HIV1 isolate C infected in PHA-stimulated PBMC assessed as inhibition of p24 gap protein2009Journal of medicinal chemistry, Jul-09, Volume: 52, Issue:13
Design of HIV-1 protease inhibitors with pyrrolidinones and oxazolidinones as novel P1'-ligands to enhance backbone-binding interactions with protease: synthesis, biological evaluation, and protein-ligand X-ray studies.
AID673368Inhibition of Human immunodeficiency virus protease M8 mutant expressed in Escherichia coli using ArgGlu(EDANS)SerGlnAsnTyrProIleValGlnLys(DABCYL)Arg as substrate preincubated with compound for 0.5 to 1 min measured by fluorometric analysis at pH 6.52012Bioorganic & medicinal chemistry letters, Aug-01, Volume: 22, Issue:15
Enamino-oxindole HIV protease inhibitors.
AID1520036Antiviral activity against VSV-G pseudotyped HIV1 transfected in human 293T cells co-transfected with NL4-3 HIV1 supernatant infected in human SupT1 cells treated with compound for 48 hrs assessed as inhibition of viral infection at 10 uM after 48 hrs by 2020European journal of medicinal chemistry, Jan-01, Volume: 185Preliminary SAR and biological evaluation of potent HIV-1 protease inhibitors with pyrimidine bases as novel P2 ligands to enhance activity against DRV-resistant HIV-1 variants.
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.
AID519789Antiviral activity against HIV 2 subtype A clinical isolate expressing 10V/I-40D-43I-56V-70K-82F-84V-89V-90M protease gene sequence from HIV2 infected patient plasma and PBMC obtained at T2 during compound treatment measured after 3 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
AID1409308Antiviral activity against Atazanavir-resistant HIV1 infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by automated chemiluminescent enzyme immunoassay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Design and Synthesis of Potent HIV-1 Protease Inhibitors Containing Bicyclic Oxazolidinone Scaffold as the P2 Ligands: Structure-Activity Studies and Biological and X-ray Structural Studies.
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.
AID1207274Inhibition of long-lasting type calcium current (ICaL) in HEK293 cells (alpha1C/beta2a/alpha2delta1) cells measured using IonWorks Barracuda automated patch clamp platform
AID1665069Resistance index, ratio of antiviral activity against VSV-G pseudotyped wild type HIV1 NL4-3 infected in human TZM-b1 cells infected with supernatants from virus-infected human SUPT1 cells to antiviral activity against darunavir-resistant HIV1 NL4-3 infec2020Bioorganic & medicinal chemistry, 08-15, Volume: 28, Issue:16
Design and biological evaluation of novel HIV-1 protease inhibitors with isopropanol as P1' ligand to enhance binding with S1' subsite.
AID396262Inhibition of HIV1 protease2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
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.
AID537774Resistance index, ratio of EC50 for multidrug resistant Human immunodeficiency virus 1 MDRC4 to EC50 for wild type Human immunodeficiency virus 12010Journal of medicinal chemistry, Nov-11, Volume: 53, Issue:21
Structure-based design, synthesis, and structure-activity relationship studies of HIV-1 protease inhibitors incorporating phenyloxazolidinones.
AID613955Antiviral activity against wild type Human immunodeficiency virus 1 ERS104pre infected in PHA-stimulated human PBMC assessed as inhibition of p24 Gag protein production2011Journal of medicinal chemistry, Aug-25, Volume: 54, Issue:16
Design of HIV-1 protease inhibitors with C3-substituted hexahydrocyclopentafuranyl urethanes as P2-ligands: synthesis, biological evaluation, and protein-ligand X-ray crystal structure.
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.
AID343024Ratio of Ki for HIV1 recombinant protease A71V/V82T/I84V mutant to Ki for wild-type HIV1 BH10 protease2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
Inorganic polyhedral metallacarborane inhibitors of HIV protease: a new approach to overcoming antiviral resistance.
AID1350492Inhibition of DRV-resistant protease V32I/L33F/I54M/V82I mutant in HIV1 transfected in 293T cells after 48 hrs by lentiviral vector-based luciferase reporter gene assay2018Journal of medicinal chemistry, 06-28, Volume: 61, Issue:12
Identification of Highly Potent Human Immunodeficiency Virus Type-1 Protease Inhibitors against Lopinavir and Darunavir Resistant Viruses from Allophenylnorstatine-Based Peptidomimetics with P2 Tetrahydrofuranylglycine.
AID1520039Antiviral activity against darunavir-resistant HIV1 NL4-3 infected in human SupT1 cells infected with supernatants from virus-infected human TZM-bl cells after 48 hrs by luciferase reporter assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Preliminary SAR and biological evaluation of potent HIV-1 protease inhibitors with pyrimidine bases as novel P2 ligands to enhance activity against DRV-resistant HIV-1 variants.
AID1516791Antiviral activity against multidrug-resistant HIV1 NL4-3 SLK19 infected in human TZM-bl cells preincubated for 18 hrs followed by viral infection and measured after 48 hrs by luciferase reporter gene assay2019Journal of medicinal chemistry, 09-12, Volume: 62, Issue:17
HIV-1 Protease Inhibitors Incorporating Stereochemically Defined P2' Ligands To Optimize Hydrogen Bonding in the Substrate Envelope.
AID1295709Binding affinity to HIV1 protease G48V mutant2016Journal of medicinal chemistry, Apr-14, Volume: 59, Issue:7
Anti-HIV Drug Discovery and Development: Current Innovations and Future Trends.
AID1607529Antiviral activity against lopinavir resistant HIV1 harboring protease L10F/V32I/M46I/I47A/A71V/I84V mutant infected in human MT4 cells assessed as inhibition of p24 Gag production2020Journal of medicinal chemistry, 05-14, Volume: 63, Issue:9
Structure-Based Design of Highly Potent HIV-1 Protease Inhibitors Containing New Tricyclic Ring P2-Ligands: Design, Synthesis, Biological, and X-ray Structural Studies.
AID519791Antiviral activity against HIV 2 subtype B clinical isolate expressing 14Y-19P-33I-61N-71I-75M-84V-90M protease gene sequence from HIV2 infected patient plasma and PBMC obtained at T1 during compound treatment measured after 13 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
AID770186Ratio of EC50 for wild type HIV1 ERS104pre to EC50 for multidrug-resistant HIV1 G harboring L10I/V11I/T12E/I15V/L19I/R41K/M46L/L63P/A71T/V82A/L90M protease-encoding region mutant2013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Highly potent HIV-1 protease inhibitors with novel tricyclic P2 ligands: design, synthesis, and protein-ligand X-ray studies.
AID1607525Inhibition of HIV-1 protease2020Journal of medicinal chemistry, 05-14, Volume: 63, Issue:9
Structure-Based Design of Highly Potent HIV-1 Protease Inhibitors Containing New Tricyclic Ring P2-Ligands: Design, Synthesis, Biological, and X-ray Structural Studies.
AID328066Effect on dimerized HIV1 protease in COS7 cells assessed as determination of CFPA/B ratio after 5 days2007The Journal of biological chemistry, Sep-28, Volume: 282, Issue:39
Potent inhibition of HIV-1 replication by novel non-peptidyl small molecule inhibitors of protease dimerization.
AID390727Inhibition of wild type HIV1 protease by competitive binding2008Journal of medicinal chemistry, Oct-23, Volume: 51, Issue:20
Solution kinetics measurements suggest HIV-1 protease has two binding sites for darunavir and amprenavir.
AID390726Inhibition of wild type HIV1 protease by enzyme inhibition2008Journal of medicinal chemistry, Oct-23, Volume: 51, Issue:20
Solution kinetics measurements suggest HIV-1 protease has two binding sites for darunavir and amprenavir.
AID1891878Antiviral activity against DRV-resistant HIV1 NL4-3 VSV-G pseudotyped virus infected in human SUP-T1 cells assessed as inhibition of late stage viral production pretreated for 48 hrs followed by viral infection and measured after 48 hrs by luciferase assa2022Bioorganic & medicinal chemistry, 06-15, Volume: 64A kind of HIV-1 protease inhibitors containing phenols with antiviral activity against DRV-resistant variants.
AID459072Inhibition of HIV1 protease2010Bioorganic & medicinal chemistry letters, Feb-01, Volume: 20, Issue:3
Synthesis and biological evaluation of novel allophenylnorstatine-based HIV-1 protease inhibitors incorporating high affinity P2-ligands.
AID343022Ratio of Ki for HIV1 recombinant protease D30N/N88D mutant to Ki for wild-type HIV1 BH10 protease2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
Inorganic polyhedral metallacarborane inhibitors of HIV protease: a new approach to overcoming antiviral resistance.
AID519794Antiviral activity against HIV 2 subtype B clinical isolate expressing 12Q-14R-17G/D-19P-61N-62I-92A protease gene sequence from HIV2 infected patient plasma and PBMC obtained before compound treatment measured after 13 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
AID1604115Inhibition of P-gp in human 12D7-MDR cells using calcein-AM as substrate after 30 mins by flow cytometric analysis2020Journal of medicinal chemistry, 03-12, Volume: 63, Issue:5
Potential Tools for Eradicating HIV Reservoirs in the Brain: Development of Trojan Horse Prodrugs for the Inhibition of P-Glycoprotein with Anti-HIV-1 Activity.
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.
AID1516793Antiviral activity against multidrug-resistant HIV1 NL4-3 KY24 infected in human TZM-bl cells preincubated for 18 hrs followed by viral infection and measured after 48 hrs by luciferase reporter gene assay2019Journal of medicinal chemistry, 09-12, Volume: 62, Issue:17
HIV-1 Protease Inhibitors Incorporating Stereochemically Defined P2' Ligands To Optimize Hydrogen Bonding in the Substrate Envelope.
AID770190Antiviral activity against multidrug-resistant HIV1 C harboring L10I/I15V/K20R/L24I/M36I/M46L/I54V/I62V/L63P/K70Q/V82A/L89M protease-encoding region mutant infected in human PHA-PBMC cells assessed as inhibition of p24 Gag protein production2013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Highly potent HIV-1 protease inhibitors with novel tricyclic P2 ligands: design, synthesis, and protein-ligand X-ray studies.
AID1241690Antiviral activity against darunavir-resistant HIV1 P10 infected in human MT4 cells by XTT assay2015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
Design of HIV-1 Protease Inhibitors with Amino-bis-tetrahydrofuran Derivatives as P2-Ligands to Enhance Backbone-Binding Interactions: Synthesis, Biological Evaluation, and Protein-Ligand X-ray Studies.
AID1409311Antiviral activity against darunavir-resistant HIV1 at passage 21 infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by automated chemiluminescent enzyme immunoassay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Design and Synthesis of Potent HIV-1 Protease Inhibitors Containing Bicyclic Oxazolidinone Scaffold as the P2 Ligands: Structure-Activity Studies and Biological and X-ray Structural Studies.
AID1520040Ratio of EC50 for antiviral activity against darunavir-resistant HIV1 NL4-3 infected in human SupT1 cells infected with supernatants from virus-infected human TZM-bl cells to EC50 for antiviral activity against wild-type HIV1 NL4-3 infected in human SupT12020European journal of medicinal chemistry, Jan-01, Volume: 185Preliminary SAR and biological evaluation of potent HIV-1 protease inhibitors with pyrimidine bases as novel P2 ligands to enhance activity against DRV-resistant HIV-1 variants.
AID673377Antiviral activity against Human immunodeficiency virus 1 HXB2 expressing protease M5 mutant infected in human MT4 cells after 5 days by MTT assay2012Bioorganic & medicinal chemistry letters, Aug-01, Volume: 22, Issue:15
Enamino-oxindole HIV protease inhibitors.
AID1891872Inhibition of HIV1 protease expressed in Escherichia coli using Arg-Glu (EDANS)-Ser-Gln-Asn-Tyr-Pro-Ile-Val-Gln-Lys(DABCYL)-Arg as substrate preincubated for 20 to 30 mins followed by substrate addition and measured for 10 mins by FRET assay2022Bioorganic & medicinal chemistry, 06-15, Volume: 64A kind of HIV-1 protease inhibitors containing phenols with antiviral activity against DRV-resistant variants.
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.
AID1904905Antiviral activity against DRV resistant HIV12022European journal of medicinal chemistry, Apr-05, Volume: 233Design, synthesis and biological evaluation of protease inhibitors containing morpholine cores with remarkable potency against both HIV-1 subtypes B and C.
AID343027Ratio of Ki for HIV1 recombinant protease L10I/L24I/L33F/M46L/154V/L63P/A71V/V82A/I84V mutant to Ki for wild-type HIV1 BH10 protease expressed in Escherichia coli2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
Inorganic polyhedral metallacarborane inhibitors of HIV protease: a new approach to overcoming antiviral resistance.
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.
AID1520035Inhibition of HIV1 protease expressed in Escherichia coli using Arg-Glu (EDANS)-Ser-Gln-Asn-Tyr-Pro-Ile-Val-Gln-Lys(DABCYL)-Arg as substrate preincubated for 20 to 30 mins followed by substrate addition and measured for 10 mins by FRET assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Preliminary SAR and biological evaluation of potent HIV-1 protease inhibitors with pyrimidine bases as novel P2 ligands to enhance activity against DRV-resistant HIV-1 variants.
AID586623Antiviral activity against 8gpNS with amino acid 116 insertion removed HIV1 infected in HEK293T cells assessed as inhibition of viral replication after 48 hrs by luciferase assay2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Three residues in HIV-1 matrix contribute to protease inhibitor susceptibility and replication capacity.
AID649703Ratio of EC50 for HIV1 MDR/G harboring protease L10I, V11I, T12E, I15V, L19I, R41K, M46L, L63P, A71T, V82A, and L90M mutant to EC50 for HIV1 ERS104 harboring wild type protease2012Bioorganic & medicinal chemistry letters, Mar-15, Volume: 22, Issue:6
Substituent effects on P2-cyclopentyltetrahydrofuranyl urethanes: design, synthesis, and X-ray studies of potent HIV-1 protease inhibitors.
AID446205Selectivity ratio of IC50 for multidrug-resistant HIV1 isolate JSL to IC50 for wild type HIV1 isolate ERS104pre2009Journal of medicinal chemistry, Dec-10, Volume: 52, Issue:23
Design, synthesis, protein-ligand X-ray structure, and biological evaluation of a series of novel macrocyclic human immunodeficiency virus-1 protease inhibitors to combat drug resistance.
AID1658399Inhibition of wild type HIV1 protease expressed in Escherichia coli using Arg-Glu (EDANS)-Ser-Gln-Asn-Tyr-Pro-Ile-Val-Gln-Lys(DABCYL)-Arg as substrate preincubated for 20 to 30 mins followed by substrate addition and measured for 10 mins by FRET assay2020ACS medicinal chemistry letters, Jun-11, Volume: 11, Issue:6
Novel HIV-1 Protease Inhibitors with Morpholine as the P2 Ligand to Enhance Activity against DRV-Resistant Variants.
AID1295707Binding affinity to wild type HIV1 protease2016Journal of medicinal chemistry, Apr-14, Volume: 59, Issue:7
Anti-HIV Drug Discovery and Development: Current Innovations and Future Trends.
AID1295711Binding affinity to HIV1 protease D30N mutant2016Journal of medicinal chemistry, Apr-14, Volume: 59, Issue:7
Anti-HIV Drug Discovery and Development: Current Innovations and Future Trends.
AID519783Antiviral activity against HIV 2 subtype A clinical isolate expressing 14H-60K/N-65E protease gene sequence from HIV2 infected patient plasma and PBMC obtained before compound treatment measured after 13 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
AID1230027Antiviral activity against multidrug resistant HIV1 MDR/B containing protease mutant infected in human MT4 cells assessed as inhibition of p24 Gag protein production relative to wild type2015Journal of medicinal chemistry, Jul-09, Volume: 58, Issue:13
Structure-based design of potent HIV-1 protease inhibitors with modified P1-biphenyl ligands: synthesis, biological evaluation, and enzyme-inhibitor X-ray structural studies.
AID1516794Selectivity ratio of EC50 for antiviral activity against multidrug-resistant HIV1 NL4-3 SLK19 infected in human TZM-bl cells to EC50 for antiviral activity against wild-type HIV1 NL4-3 infected in human TZM-bl cells2019Journal of medicinal chemistry, 09-12, Volume: 62, Issue:17
HIV-1 Protease Inhibitors Incorporating Stereochemically Defined P2' Ligands To Optimize Hydrogen Bonding in the Substrate Envelope.
AID396245Antiviral activity against HIV1 HXB2 in human MT4 cells assessed as inhibition of viral-induced viral cytopathic effect in presence of 10% fetal bovine serum by MTS assay2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
AID519781Antiviral activity against HIV 2 subtype A clinical isolate expressing 14H-17D-43T-68N/D protease gene sequence from HIV2 infected patient plasma and PBMC obtained before compound treatment measured after 13 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
AID673374Antiviral activity against Human immunodeficiency virus 1 HXB2 expressing protease M10 mutant infected in human MT4 cells after 5 days by MTT assay2012Bioorganic & medicinal chemistry letters, Aug-01, Volume: 22, Issue:15
Enamino-oxindole HIV protease inhibitors.
AID343020Inhibition of HIV1 recombinant protease L10I/L24I/L33F/M46L/154V/L63P/A71V/V82A/I84V mutant expressed in Escherichia coli by spectrophotometric assay2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
Inorganic polyhedral metallacarborane inhibitors of HIV protease: a new approach to overcoming antiviral resistance.
AID375201Antiviral activity against multidrug-resistant HIV1 isolate MM infected in PHA-stimulated PBMC assessed as inhibition of p24 gap protein2009Journal of medicinal chemistry, Jul-09, Volume: 52, Issue:13
Design of HIV-1 protease inhibitors with pyrrolidinones and oxazolidinones as novel P1'-ligands to enhance backbone-binding interactions with protease: 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.
AID1482918Ratio of IC50 for darunavir-resistant HIV1 derived from 51 passages harboring protease L10I/I15V/K20R/L24I/V32I/L33F/M36I/M46L/I54M/L63P/K70Q/V82I/I84V/L89M mutant infected in human MT4 cells to IC50 for wild type HIV1 NL4.3 infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID396247Antiviral activity against HIV1 HXB2 in human MT4 cells assessed as inhibition of viral-induced viral cytopathic effect in presence of 10% fetal bovine serum and 10% human serum by MTS assay2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
AID738153Binding affinity to HIV1 protease Q7K, L10F, I13V, I15V, D30V, V32I, L33F, E35D, M36I, S37N, I47V, I54L, Q58E, I62V, L63P, A71V, I84V, N88D, L89T, L90M mutant expressed in Escherichia coli BL21 (DE3) assessed as association constant by isothermal titratio2013Journal of medicinal chemistry, May-23, Volume: 56, Issue:10
Extreme multidrug resistant HIV-1 protease with 20 mutations is resistant to novel protease inhibitors with P1'-pyrrolidinone or P2-tris-tetrahydrofuran.
AID1633413Antiviral activity against wild-type HIV1 NL4-3 infected infected in human MT2 cells assessed as reduction in extracellular RNA at 1 uM and measured after 30 hrs post infection by Alu-LTR PCR protocol based method2019ACS medicinal chemistry letters, Apr-11, Volume: 10, Issue:4
Synthesis and Evaluation of Bifunctional Aminothiazoles as Antiretrovirals Targeting the HIV-1 Nucleocapsid Protein.
AID1904906Antiviral activity against HIV1 subtype C isolate ZM2462022European journal of medicinal chemistry, Apr-05, Volume: 233Design, synthesis and biological evaluation of protease inhibitors containing morpholine cores with remarkable potency against both HIV-1 subtypes B and C.
AID396248Antiviral activity against HIV1 HXB2 in human MT4 cells assessed as inhibition of viral-induced viral cytopathic effect in presence of 10% fetal bovine serum and 20% human serum by MTS assay2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
AID726415Antiviral activity against Human immunodeficiency virus 1 3B clinical isolate harboring L10I/I13V/M46I/I50V/L63P/L76V protease mutant infected in human MT4 cells assessed as inhibition of viral replication2013Bioorganic & medicinal chemistry letters, Jan-01, Volume: 23, Issue:1
Design and synthesis of HIV-1 protease inhibitors for a long-acting injectable drug application.
AID1482926Ratio of IC50 for atazanavir-resistant HIV1 NL4-3 harboring protease L23I/E34Q/K43I/M46I/I50L/G51A/L63P/A71V/V82A/T91A mutant infected in human MT4 cells to IC50 for wild type HIV1 NL4.3 infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
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.
AID1409306Inhibition of HIV1 wild type NL4-3 protease expressed in Escherichia coli Rosetta (DE3) pLysS using Abz-Thr-Ile-Nle-Phe-(pNO2)-Gln-Arg-NH2 as substrate preincubated for 5 to 10 mins followed by substrate addition measured after 1 hr by fluorescence assay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Design and Synthesis of Potent HIV-1 Protease Inhibitors Containing Bicyclic Oxazolidinone Scaffold as the P2 Ligands: Structure-Activity Studies and Biological and X-ray Structural Studies.
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.
AID1520034Cytotoxicity against human 293T cells measured after 24 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Preliminary SAR and biological evaluation of potent HIV-1 protease inhibitors with pyrimidine bases as novel P2 ligands to enhance activity against DRV-resistant HIV-1 variants.
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.
AID446199Antiviral activity against multidrug-resistant HIV1 isolate JSL infected in PHA-stimulated human PBMC assessed as inhibition of p24 gap protein production2009Journal of medicinal chemistry, Dec-10, Volume: 52, Issue:23
Design, synthesis, protein-ligand X-ray structure, and biological evaluation of a series of novel macrocyclic human immunodeficiency virus-1 protease inhibitors to combat drug resistance.
AID396246Antiviral activity against HIV1 HXB2 in human MT4 cells assessed as inhibition of viral-induced viral cytopathic effect in presence of 40% fetal bovine serum by MTS assay2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
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).
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.
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.
AID1604117Inhibition of P-gp in human CMEC/D3 cells using calcein-AM as substrate after 30 mins by flow cytometric analysis2020Journal of medicinal chemistry, 03-12, Volume: 63, Issue:5
Potential Tools for Eradicating HIV Reservoirs in the Brain: Development of Trojan Horse Prodrugs for the Inhibition of P-Glycoprotein with Anti-HIV-1 Activity.
AID415238Antiviral activity against protease inhibitor resistant HIV1 isolates2009Journal 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.
AID1658398Antiviral activity against wild type HIV-12020ACS medicinal chemistry letters, Jun-11, Volume: 11, Issue:6
Novel HIV-1 Protease Inhibitors with Morpholine as the P2 Ligand to Enhance Activity against DRV-Resistant Variants.
AID770188Antiviral activity against multidrug-resistant HIV1 TM harboring L10I/K14R/R41K/M46L/I54V/L63P/A71V/V82A/L90M/I93L protease-encoding region mutant infected in human PHA-PBMC cells assessed as inhibition of p24 Gag protein production2013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Highly potent HIV-1 protease inhibitors with novel tricyclic P2 ligands: design, synthesis, and protein-ligand X-ray studies.
AID1607528Antiviral activity against atazanavir resistant HIV1 harboring protease L23I/E34Q/K43I/M46I/I50L/G51A/L63P/A71V/V82A/T91A mutant infected in human MT4 cells assessed as reduction in p24 Gag production2020Journal of medicinal chemistry, 05-14, Volume: 63, Issue:9
Structure-Based Design of Highly Potent HIV-1 Protease Inhibitors Containing New Tricyclic Ring P2-Ligands: Design, Synthesis, Biological, and X-ray Structural Studies.
AID668810Antiviral activity against Human immunodeficiency virus 1 isolate M2 expressing protease L10I, I13V, M46I, I50V, L63P, L76V mutant infected in human MT4 cells2011ACS medicinal chemistry letters, Jun-09, Volume: 2, Issue:6
Disubstituted Bis-THF Moieties as New P2 Ligands in Nonpeptidal HIV-1 Protease Inhibitors.
AID1725418Antiviral activity against HIV1 NL4-3 infected in human MT4 cells incubated for 7 days by fully automated chemiluminescent enzyme immunoassay2020ACS medicinal chemistry letters, Oct-08, Volume: 11, Issue:10
Design, Synthesis, and X-ray Studies of Potent HIV-1 Protease Inhibitors with P2-Carboxamide Functionalities.
AID249507Effective concentration against PI-Resistant HIV strain (M5)2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Discovery and selection of TMC114, a next generation HIV-1 protease inhibitor.
AID537772Antiviral activity against multidrug resistant Human immunodeficiency virus 1 MDRC42010Journal of medicinal chemistry, Nov-11, Volume: 53, Issue:21
Structure-based design, synthesis, and structure-activity relationship studies of HIV-1 protease inhibitors incorporating phenyloxazolidinones.
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.
AID1465008Inhibition of wild-type HIV-1 protease2017Bioorganic & medicinal chemistry letters, 11-01, Volume: 27, Issue:21
Design, synthesis, X-ray studies, and biological evaluation of novel macrocyclic HIV-1 protease inhibitors involving the P1'-P2' ligands.
AID459077Antiviral activity against multi drug-resistant HIV1 isolate of subtype MM in PHA-stimulated human PBMC assessed as inhibition of p24 antigen production2010Bioorganic & medicinal chemistry letters, Feb-01, Volume: 20, Issue:3
Synthesis and biological evaluation of novel allophenylnorstatine-based HIV-1 protease inhibitors incorporating high affinity P2-ligands.
AID613959Antiviral activity against multidrug-resistant Human immunodeficiency virus 1 MDR/TM harboring protease L10I, K14R, R41K, M46L, I54V, L63P, A71V, V82A, L90M, I93L mutation infected in PHA-stimulated human PBMC assessed as inhibition of p24 Gag protein pro2011Journal of medicinal chemistry, Aug-25, Volume: 54, Issue:16
Design of HIV-1 protease inhibitors with C3-substituted hexahydrocyclopentafuranyl urethanes as P2-ligands: synthesis, biological evaluation, and protein-ligand X-ray crystal structure.
AID564042Antiviral activity against wild type HIV1 ERS104 containing protease L36P mutant infected in human PHA-PBMC cells by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
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.
AID459079Antiviral activity against wild type HIV1 isolate ERS104pre in PHA-stimulated human PBMC assessed as inhibition of p24 antigen production2010Bioorganic & medicinal chemistry letters, Feb-01, Volume: 20, Issue:3
Synthesis and biological evaluation of novel allophenylnorstatine-based HIV-1 protease inhibitors incorporating high affinity P2-ligands.
AID673367Inhibition of wild type Human immunodeficiency virus protease expressed in Escherichia coli using ArgGlu(EDANS)SerGlnAsnTyrProIleValGlnLys(DABCYL)Arg as substrate preincubated with compound for 0.5 to 1 min measured by fluorometric analysis at pH 6.52012Bioorganic & medicinal chemistry letters, Aug-01, Volume: 22, Issue:15
Enamino-oxindole HIV protease inhibitors.
AID1725420Antiviral activity against ATV-resistant HIV1 infected in human MT4 cells incubated for 7 days by fully automated chemiluminescent enzyme immunoassay2020ACS medicinal chemistry letters, Oct-08, Volume: 11, Issue:10
Design, Synthesis, and X-ray Studies of Potent HIV-1 Protease Inhibitors with P2-Carboxamide Functionalities.
AID668815Cytotoxicity against human MT4 cells2011ACS medicinal chemistry letters, Jun-09, Volume: 2, Issue:6
Disubstituted Bis-THF Moieties as New P2 Ligands in Nonpeptidal HIV-1 Protease Inhibitors.
AID1891877Antiviral activity against wild-type HIV1 NL4-3 VSV-G pseudotyped virus infected in human SUP-T1 cells assessed as inhibition of late stage viral production pretreated with viral infected SUP-T1 cells for 48 hrs followed by reinfection in TZM-bl cells by 2022Bioorganic & medicinal chemistry, 06-15, Volume: 64A kind of HIV-1 protease inhibitors containing phenols with antiviral activity against DRV-resistant variants.
AID519539Antiviral activity against wild-type HIV1 RF infected in human MT4 cells assessed as reduction in viral cytopathogenicity treated 1 hr post infection measured 5 days post infection in absence of human serum by MTT assay2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
Characterization of a novel human immunodeficiency virus type 1 protease inhibitor, A-790742.
AID1516796Selectivity ratio of EC50 for antiviral activity against multidrug-resistant HIV1 NL4-3 KY24 infected in human TZM-bl cells to EC50 for antiviral activity against wild-type HIV1 NL4-3 infected in human TZM-bl cells2019Journal of medicinal chemistry, 09-12, Volume: 62, Issue:17
HIV-1 Protease Inhibitors Incorporating Stereochemically Defined P2' Ligands To Optimize Hydrogen Bonding in the Substrate Envelope.
AID770185Ratio of EC50 for wild type HIV1 ERS104pre to EC50 for multidrug-resistant HIV1 TM harboring L10I/K14R/R41K/M46L/I54V/L63P/A71V/V82A/L90M/I93L protease-encoding region mutant2013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Highly potent HIV-1 protease inhibitors with novel tricyclic P2 ligands: design, synthesis, and protein-ligand X-ray studies.
AID770194Antiviral activity against HIV1 LAI infected in human MT2 cells by MTT assay2013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Highly potent HIV-1 protease inhibitors with novel tricyclic P2 ligands: design, synthesis, and protein-ligand X-ray studies.
AID1482906Antiviral activity against tipranavir-resistant HIV1 NL4-3 harboring protease L10I/L33I/M36I/M46I/I54V/K55R/I62V/L63P/A71V/G73S/V82T/L90M/I93L mutant infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by chemilumi2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID658565Antiviral activity against multidrug-resistant HIV1 106-PR infected in HEK293T cells assessed as inhibition of viral replication after 4 days by beta-galactosidase reporter gene assay2012Journal of natural products, Mar-23, Volume: 75, Issue:3
Library-based discovery and characterization of daphnane diterpenes as potent and selective HIV inhibitors in Daphne gnidium.
AID459083Ratio of IC50 for multi drug-resistant HIV1 isolate of subtype TM to IC50 for wild type HIV1 isolate ERS104pre2010Bioorganic & medicinal chemistry letters, Feb-01, Volume: 20, Issue:3
Synthesis and biological evaluation of novel allophenylnorstatine-based HIV-1 protease inhibitors incorporating high affinity P2-ligands.
AID415236Inhibition of HIV1 protease2009Journal 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.
AID1482905Antiviral activity against amprenavir-resistant HIV1 NL4-3 harboring protease L10F/V32I/L33F/M46L/I54M/A71V mutant infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by chemiluminescent enzyme immunoassay2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID1891879Selectivity ratio of EC50 for antiviral activity against DRV-resistant HIV1 NL4-3 VSV-G pseudotyped virus infected in human SUP-T1 cells to EC50 for antiviral activity against wild-type HIV1 NL4-3 VSV-G pseudotyped virus infected in human SUP-T1 cells2022Bioorganic & medicinal chemistry, 06-15, Volume: 64A kind of HIV-1 protease inhibitors containing phenols with antiviral activity against DRV-resistant variants.
AID249504Effective concentration against PI-Resistant HIV strain (M2)2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Discovery and selection of TMC114, a next generation HIV-1 protease inhibitor.
AID1295714Binding affinity to HIV1 protease A71V mutant2016Journal of medicinal chemistry, Apr-14, Volume: 59, Issue:7
Anti-HIV Drug Discovery and Development: Current Innovations and Future Trends.
AID1230025Antiviral activity against multidrug resistant HIV1 MDR/C containing protease mutant infected in human MT4 cells assessed as inhibition of p24 Gag protein production2015Journal of medicinal chemistry, Jul-09, Volume: 58, Issue:13
Structure-based design of potent HIV-1 protease inhibitors with modified P1-biphenyl ligands: synthesis, biological evaluation, and enzyme-inhibitor X-ray structural studies.
AID328062Inhibition of HIV1 protease dimerization in MT2 cells2007The Journal of biological chemistry, Sep-28, Volume: 282, Issue:39
Potent inhibition of HIV-1 replication by novel non-peptidyl small molecule inhibitors of protease dimerization.
AID343018Inhibition of HIV1 recombinant protease V32I/I47A mutant expressed in Escherichia coli by spectrophotometric assay2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
Inorganic polyhedral metallacarborane inhibitors of HIV protease: a new approach to overcoming antiviral resistance.
AID1520038Antiviral activity against wild-type HIV1 NL4-3 infected in human SupT1 cells infected with supernatants from virus-infected human TZM-bl cells after 48 hrs by luciferase reporter assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Preliminary SAR and biological evaluation of potent HIV-1 protease inhibitors with pyrimidine bases as novel P2 ligands to enhance activity against DRV-resistant HIV-1 variants.
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.
AID249509Effective concentration as average activity on mutant HIV panel (AVMUT)2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Discovery and selection of TMC114, a next generation HIV-1 protease inhibitor.
AID249505Effective concentration against PI-Resistant HIV strain (M3)2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Discovery and selection of TMC114, a next generation HIV-1 protease inhibitor.
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.
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).
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.
AID459074Antiviral activity against multi drug-resistant HIV1 isolate of subtype C in PHA-stimulated human PBMC assessed as inhibition of p24 antigen production2010Bioorganic & medicinal chemistry letters, Feb-01, Volume: 20, Issue:3
Synthesis and biological evaluation of novel allophenylnorstatine-based HIV-1 protease inhibitors incorporating high affinity P2-ligands.
AID1207394Inhibition of slow delayed inward rectifying potassium current (Iks) in Chinese Hamster Ovary (CHO) cells transfected with KCNQ1 / Kv1.7 / KvLQT1 and KCNE1/minK measured using IonWorks automated patch clamp platform
AID564030Antiviral activity against HIV2 EHO infected in human MT2 cells by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
AID1295717Binding affinity to HIV1 protease L90M mutant2016Journal of medicinal chemistry, Apr-14, Volume: 59, Issue:7
Anti-HIV Drug Discovery and Development: Current Innovations and Future Trends.
AID415282Antiviral activity against HIV1 bearing protease gene with B26 mutation infected in human MT4 cells after 5 days by MTT assay2009Journal 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.
AID311575Antiviral activity against HIV1 in CEM cells2007Bioorganic & medicinal chemistry, Dec-15, Volume: 15, Issue:24
Darunavir, a conceptually new HIV-1 protease inhibitor for the treatment of drug-resistant HIV.
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.
AID586624Antiviral activity against HIV1 harboring wild type 8.9NSX with mutant amino acid 116 insertion infected in HEK293T cells assessed as inhibition of viral replication after 48 hrs by luciferase assay2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Three residues in HIV-1 matrix contribute to protease inhibitor susceptibility and replication capacity.
AID726419Antiviral activity against wild type Human immunodeficiency virus 1 3B infected in human MT4 cells assessed as inhibition of viral replication2013Bioorganic & medicinal chemistry letters, Jan-01, Volume: 23, Issue:1
Design and synthesis of HIV-1 protease inhibitors for a long-acting injectable drug application.
AID1465003Inhibition of HIV-1 protease using fluorogenic substrate by fluorescence assay2017Bioorganic & medicinal chemistry letters, 11-01, Volume: 27, Issue:21
Design, synthesis, X-ray studies, and biological evaluation of novel macrocyclic HIV-1 protease inhibitors involving the P1'-P2' ligands.
AID1230024Antiviral activity against multidrug resistant HIV1 MDR/B containing protease mutant infected in human MT4 cells assessed as inhibition of p24 Gag protein production2015Journal of medicinal chemistry, Jul-09, Volume: 58, Issue:13
Structure-based design of potent HIV-1 protease inhibitors with modified P1-biphenyl ligands: synthesis, biological evaluation, and enzyme-inhibitor X-ray structural studies.
AID1295718Binding affinity to HIV1 protease I50V mutant2016Journal of medicinal chemistry, Apr-14, Volume: 59, Issue:7
Anti-HIV Drug Discovery and Development: Current Innovations and Future Trends.
AID459076Antiviral activity against multi drug-resistant HIV1 isolate of subtype TM in PHA-stimulated human PBMC assessed as inhibition of p24 antigen production2010Bioorganic & medicinal chemistry letters, Feb-01, Volume: 20, Issue:3
Synthesis and biological evaluation of novel allophenylnorstatine-based HIV-1 protease inhibitors incorporating high affinity P2-ligands.
AID1207334Inhibition of fast sodium current (INa) in HEK293 cells transfected with human Nav1.5 measured using IonWorks Quattro automated patch clamp platform
AID396257Ratio of EC50 for HIV1 HXB2 in presence of 1 mg/ml alpha1-acid glycoprotein to EC50 for HIV1 HXB22007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
AID1585151Antiviral activity against HIV1 NL4-3 infected in human MT4 cells after 7 days by MTT assay2018European journal of medicinal chemistry, Dec-05, Volume: 160Design, synthesis, and X-ray studies of potent HIV-1 protease inhibitors incorporating aminothiochromane and aminotetrahydronaphthalene carboxamide derivatives as the P2 ligands.
AID564044Antiviral activity against multidrug-resistant HIV1 isolate C containing protease L10I, I15V, K20R, L24I, M36I, M46L, I54V, I62V, L63P, K70Q, V82A, and L89M mutant infected in human PHA-PBMC cells by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
AID1575211Inhibition of HIV1 protease expressed in Escherichia coli using Arg-Glu (EDANS)-Ser-Gln-Asn-Tyr-Pro-Ile-Val-Gln-Lys(DABCYL)-Arg as substrate preincubated for 20 to 30 mins followed by substrate addition and measured for 10 mins by FRET assay2019Bioorganic & medicinal chemistry letters, 02-01, Volume: 29, Issue:3
Design, synthesis and biological evaluation of novel HIV-1 protease inhibitors with pentacyclic triterpenoids as P2-ligands.
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.
AID1230026Antiviral activity against multidrug resistant HIV1 MDR/G containing protease mutant infected in human MT4 cells assessed as inhibition of p24 Gag protein production2015Journal of medicinal chemistry, Jul-09, Volume: 58, Issue:13
Structure-based design of potent HIV-1 protease inhibitors with modified P1-biphenyl ligands: synthesis, biological evaluation, and enzyme-inhibitor X-ray structural studies.
AID697741Antiviral activity against wild type HIV1 clade B isolated form HIV1 patient infected in HEK293 cells assessed as reduction in viral replication incubated for 48 hrs by luciferase reporter based assay2012Journal of medicinal chemistry, Jul-26, Volume: 55, Issue:14
Design, synthesis, and biological and structural evaluations of novel HIV-1 protease inhibitors to combat drug resistance.
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).
AID1482916Ratio of IC50 for darunavir-resistant HIV1 derived from 20 passages harboring protease L10I/I15V/K20R/L24I/V32I/M36I/M46L/L63P/V82A/L89M mutant infected in human MT4 cells to IC50 for wild type HIV1 NL4.3 infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID1460921Antiviral activity against HIV1 LAI infected in human MT2 cells assessed as reduction in p24 Gag protein production incubated for 6 days by automated chemiluminescent enzyme immunoassay2017Bioorganic & medicinal chemistry, 10-01, Volume: 25, Issue:19
Design of novel HIV-1 protease inhibitors incorporating isophthalamide-derived P2-P3 ligands: Synthesis, biological evaluation and X-ray structural studies of inhibitor-HIV-1 protease complex.
AID519784Antiviral activity against HIV 2 subtype A clinical isolate expressing 54M-65E-71I-74N-90M protease gene sequence from HIV2 infected patient plasma and PBMC obtained at T1 during compound treatment measured after 13 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
AID564041Antiviral activity against HIV1 expressing protease L23I/E34Q/K43I/M46I/I50L/G51A/L63P/A71V/V82A/T91A mutant infected in human MT4 cells selected at 5 uM of atazanavir by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
AID673375Antiviral activity against Human immunodeficiency virus 1 HXB2 expressing protease M3 mutant infected in human MT4 cells after 5 days by MTT assay2012Bioorganic & medicinal chemistry letters, Aug-01, Volume: 22, Issue:15
Enamino-oxindole HIV protease inhibitors.
AID537769Antiviral activity against Human immunodeficiency virus 1 clade B isolated from HIV-AIDS patient2010Journal of medicinal chemistry, Nov-11, Volume: 53, Issue:21
Structure-based design, synthesis, and structure-activity relationship studies of HIV-1 protease inhibitors incorporating phenyloxazolidinones.
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.
AID1194842Inhibition of HIV1 protease expressed in Escherichia coli incubated for 20 to 30 mins at room temperature using (Arg-Glu(EDANS)-Ser-Gln-Asn-Tyr-Pro-Ile-Val-Gln-Lys(DABCYL)-Arg) substrate by FRET method2015Bioorganic & medicinal chemistry letters, May-01, Volume: 25, Issue:9
Synthesis and biological evaluation of novel HIV-1 protease inhibitors using tertiary amine as P2-ligands.
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).
AID236641Maximum plasma concentration in dog after 80 mg/kg oral administration2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Discovery and selection of TMC114, a next generation HIV-1 protease inhibitor.
AID375200Antiviral activity against multidrug-resistant HIV1 isolate TM infected in PHA-stimulated PBMC assessed as inhibition of p24 gap protein2009Journal of medicinal chemistry, Jul-09, Volume: 52, Issue:13
Design of HIV-1 protease inhibitors with pyrrolidinones and oxazolidinones as novel P1'-ligands to enhance backbone-binding interactions with protease: synthesis, biological evaluation, and protein-ligand X-ray studies.
AID1409313Antiviral activity against darunavir-resistant HIV1 at passage 51 infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by automated chemiluminescent enzyme immunoassay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Design and Synthesis of Potent HIV-1 Protease Inhibitors Containing Bicyclic Oxazolidinone Scaffold as the P2 Ligands: Structure-Activity Studies and Biological and X-ray Structural Studies.
AID1482909Antiviral activity against lopinavir-resistant HIV1 NL4-3 harboring protease L10F/V32I/M46I/I47A/A71V/I84V mutant infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by chemiluminescent enzyme immunoassay2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID459081Ratio of IC50 for multi drug-resistant HIV1 isolate of subtype C to IC50 for wild type HIV1 isolate ERS104pre2010Bioorganic & medicinal chemistry letters, Feb-01, Volume: 20, Issue:3
Synthesis and biological evaluation of novel allophenylnorstatine-based HIV-1 protease inhibitors incorporating high affinity P2-ligands.
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).
AID1516790Antiviral activity against wild-type HIV1 NL4-3 infected in human TZM-bl cells preincubated for 18 hrs followed by viral infection and measured after 48 hrs by luciferase reporter gene assay2019Journal of medicinal chemistry, 09-12, Volume: 62, Issue:17
HIV-1 Protease Inhibitors Incorporating Stereochemically Defined P2' Ligands To Optimize Hydrogen Bonding in the Substrate Envelope.
AID521549Antiviral activity against HIV 2 subtype H expressing 10I-40P-41Y-60H-63N-70T-73G-89L-92E protease gene sequence from HIV2 infected patient plasma and PBMC obtained before compound treatment measured after 13 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
AID260518Binding affinity to HIV1 protease L90M2006Journal of medicinal chemistry, Feb-23, Volume: 49, Issue:4
Effectiveness of nonpeptide clinical inhibitor TMC-114 on HIV-1 protease with highly drug resistant mutations D30N, I50V, and L90M.
AID1778342Antiviral activity against HIV1 subtype C isolate 119282021European journal of medicinal chemistry, Aug-05, Volume: 220Design and evaluation of novel piperidine HIV-1 protease inhibitors with potency against DRV-resistant variants.
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.
AID1409307Antiviral activity against HIV1 NL4-3 infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by automated chemiluminescent enzyme immunoassay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Design and Synthesis of Potent HIV-1 Protease Inhibitors Containing Bicyclic Oxazolidinone Scaffold as the P2 Ligands: Structure-Activity Studies and Biological and X-ray Structural Studies.
AID459080Antiviral activity against HIV12010Bioorganic & medicinal chemistry letters, Feb-01, Volume: 20, Issue:3
Synthesis and biological evaluation of novel allophenylnorstatine-based HIV-1 protease inhibitors incorporating high affinity P2-ligands.
AID697739Antiviral activity against multidrug resistant HIV1 MDRC4 infected in HEK293 cells assessed as reduction in viral replication incubated for 48 hrs by luciferase reporter based assay2012Journal of medicinal chemistry, Jul-26, Volume: 55, Issue:14
Design, synthesis, and biological and structural evaluations of novel HIV-1 protease inhibitors to combat drug resistance.
AID343028Ratio of Ki for HIV1 recombinant protease L10F/L19I/K20R/L33F/E35D/M36I/R41K/F53L/I54V/L63P/H69K/A71V/T74P/I84V/L89M/L90M/I93L mutant to Ki for wild-type HIV1 BH10 protease2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
Inorganic polyhedral metallacarborane inhibitors of HIV protease: a new approach to overcoming antiviral resistance.
AID738152Binding affinity to HIV1 protease Q7K, L10F, I13V, I15V, D30V, V32I, L33F, E35D, M36I, S37N, I47V, I54L, Q58E, I62V, L63P, A71V, I84V, N88D, L89T, L90M mutant expressed in Escherichia coli BL21 (DE3) assessed as dissociation constant by isothermal titrati2013Journal of medicinal chemistry, May-23, Volume: 56, Issue:10
Extreme multidrug resistant HIV-1 protease with 20 mutations is resistant to novel protease inhibitors with P1'-pyrrolidinone or P2-tris-tetrahydrofuran.
AID1891880Antiviral activity against HIV1 NL4-3 VSV-G pseudotyped virus infected in human SUP-T1 cells assessed as inhibition of late stage viral production at 100 nM pretreated for 48 hrs followed by viral infection and measured after 48 hrs by luciferase assay re2022Bioorganic & medicinal chemistry, 06-15, Volume: 64A kind of HIV-1 protease inhibitors containing phenols with antiviral activity against DRV-resistant variants.
AID697743Antiviral activity against wild type HIV1 infected in HEK293 cells assessed as reduction in viral replication incubated for 48 hrs by luciferase reporter based assay2012Journal of medicinal chemistry, Jul-26, Volume: 55, Issue:14
Design, synthesis, and biological and structural evaluations of novel HIV-1 protease inhibitors to combat drug resistance.
AID1409312Antiviral activity against darunavir-resistant HIV1 at passage 30 infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by automated chemiluminescent enzyme immunoassay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Design and Synthesis of Potent HIV-1 Protease Inhibitors Containing Bicyclic Oxazolidinone Scaffold as the P2 Ligands: Structure-Activity Studies and Biological and X-ray Structural Studies.
AID586622Ratio of EC50 for HIV1 harboring HIV1 harboring wild type 8.9NSX with gag RF79F and T81A mutant to EC50 for wild type HIV12011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Three residues in HIV-1 matrix contribute to protease inhibitor susceptibility and replication capacity.
AID1380927Antiviral activity against APV resistant HIV1 harboring protease L10F/M46I/I50V/I85V mutant infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by chemiluminescent enzyme immunoassay2018Journal of medicinal chemistry, 05-24, Volume: 61, Issue:10
Design and Synthesis of Highly Potent HIV-1 Protease Inhibitors Containing Tricyclic Fused Ring Systems as Novel P2 Ligands: Structure-Activity Studies, Biological and X-ray Structural Analysis.
AID738147Binding affinity to HIV1 protease expressed in Escherichia coli BL21 (DE3) assessed as association constant by isothermal titration calorimetric analysis2013Journal of medicinal chemistry, May-23, Volume: 56, Issue:10
Extreme multidrug resistant HIV-1 protease with 20 mutations is resistant to novel protease inhibitors with P1'-pyrrolidinone or P2-tris-tetrahydrofuran.
AID297696Inhibition of HIV1 protease I50V mutant2007Journal of medicinal chemistry, Sep-06, Volume: 50, Issue:18
Potent new antiviral compound shows similar inhibition and structural interactions with drug resistant mutants and wild type HIV-1 protease.
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.
AID726414Antiviral activity against Human immunodeficiency virus 1 3B clinical isolate harboring L10I/K20R/M36I/G48V/ I62V/A71V/V82A/I93L protease mutant infected in human MT4 cells assessed as inhibition of viral replication2013Bioorganic & medicinal chemistry letters, Jan-01, Volume: 23, Issue:1
Design and synthesis of HIV-1 protease inhibitors for a long-acting injectable drug application.
AID668813Selectivity index, ratio of EC50 for Human immunodeficiency virus 1 isolate M2 expressing protease L10I, I13V, M46I, I50V, L63P, L76V mutant to EC50 for Human immunodeficiency virus 1 3B expressing wild-type protease2011ACS medicinal chemistry letters, Jun-09, Volume: 2, Issue:6
Disubstituted Bis-THF Moieties as New P2 Ligands in Nonpeptidal HIV-1 Protease Inhibitors.
AID564060Antiviral activity against HIV1 expressing protease L10I/L24I/M46I/V82I/I84V mutant infected in human MT4 cells selected after 50 passages of GRL-216 by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants 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.
AID564043Antiviral activity against multidrug-resistant HIV1 isolate B containing protease L10I, K14R, L33I, M36I,M46I, F53I, K55R, I62V, L63P, A71V, G73S, V82A, L90M, and I93L mutant infected in human PHA-PBMC cells by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
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.
AID446193Antiviral activity against wild type HIV1 isolate ERS104pre infected in PHA-stimulated human PBMC assessed as inhibition of p24 gap protein production2009Journal of medicinal chemistry, Dec-10, Volume: 52, Issue:23
Design, synthesis, protein-ligand X-ray structure, and biological evaluation of a series of novel macrocyclic human immunodeficiency virus-1 protease inhibitors to combat drug resistance.
AID537773Resistance index, ratio of EC50 for multidrug resistant Human immunodeficiency virus 1 harboring protease M46I, I54V, V82A and L90M mutant to EC50 for wild type Human immunodeficiency virus 12010Journal of medicinal chemistry, Nov-11, Volume: 53, Issue:21
Structure-based design, synthesis, and structure-activity relationship studies of HIV-1 protease inhibitors incorporating phenyloxazolidinones.
AID1778335Cytotoxicity against HEK293T cells assessed as reduction in cell viability incubated for 24 hrs by CCK8 assay2021European journal of medicinal chemistry, Aug-05, Volume: 220Design and evaluation of novel piperidine HIV-1 protease inhibitors with potency against DRV-resistant variants.
AID726418Antiviral activity against wild type Human immunodeficiency virus 1 3B infected in human MT4 cells assessed as inhibition of viral replication in presence of 50% human plasma2013Bioorganic & medicinal chemistry letters, Jan-01, Volume: 23, Issue:1
Design and synthesis of HIV-1 protease inhibitors for a long-acting injectable drug application.
AID105016Compound was tested for the inhibition of HIV-1 in MT-4 human T-lymphoid cells infected with IIIB isolate1998Bioorganic & medicinal chemistry letters, Mar-17, Volume: 8, Issue:6
Potent HIV protease inhibitors incorporating high-affinity P2-ligands and (R)-(hydroxyethylamino)sulfonamide isostere.
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.
AID446194Antiviral activity against multidrug-resistant HIV1 isolate B infected in PHA-stimulated human PBMC assessed as inhibition of p24 gap protein production2009Journal of medicinal chemistry, Dec-10, Volume: 52, Issue:23
Design, synthesis, protein-ligand X-ray structure, and biological evaluation of a series of novel macrocyclic human immunodeficiency virus-1 protease inhibitors to combat drug resistance.
AID1380923Antiviral activity against HIV-1 LAI infected in human MT2 cells2018Journal of medicinal chemistry, 05-24, Volume: 61, Issue:10
Design and Synthesis of Highly Potent HIV-1 Protease Inhibitors Containing Tricyclic Fused Ring Systems as Novel P2 Ligands: Structure-Activity Studies, Biological and X-ray Structural Analysis.
AID566849Antiviral activity against HIV-1 MDR/G infected in human PHA-PBM cells assessed as inhibition of p24 Gag protein production2011Journal of medicinal chemistry, Jan-27, Volume: 54, Issue:2
Design and synthesis of potent HIV-1 protease inhibitors incorporating hexahydrofuropyranol-derived high affinity P(2) ligands: structure-activity studies and biological evaluation.
AID519793Antiviral activity against HIV 2 subtype B clinical isolate expressing 12T-14Y-19P-40N-41D-61N-62I-96S-99L protease gene sequence from HIV2 infected patient plasma and PBMC obtained before compound treatment measured after 13 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
AID396253Ratio of EC50 for HIV1 HXB2 in presence of 30% human serum to EC50 for HIV1 HXB2 in presence of 10% fetal bovine serum2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
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.
AID446198Antiviral activity against multidrug-resistant HIV1 isolate MM infected in PHA-stimulated human PBMC assessed as inhibition of p24 gap protein production2009Journal of medicinal chemistry, Dec-10, Volume: 52, Issue:23
Design, synthesis, protein-ligand X-ray structure, and biological evaluation of a series of novel macrocyclic human immunodeficiency virus-1 protease inhibitors to combat drug resistance.
AID396260Antiviral activity against HIV1 NL4-32007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
AID697742Antiviral activity against wild type HIV1 clade A isolated form HIV1 patient infected in HEK293 cells assessed as reduction in viral replication incubated for 48 hrs by luciferase reporter based assay2012Journal of medicinal chemistry, Jul-26, Volume: 55, Issue:14
Design, synthesis, and biological and structural evaluations of novel HIV-1 protease inhibitors to combat drug resistance.
AID1607527Antiviral activity against HIV1 NL4-3 infected in human MT4 cells assessed as reduction in p24 Gag protein production2020Journal of medicinal chemistry, 05-14, Volume: 63, Issue:9
Structure-Based Design of Highly Potent HIV-1 Protease Inhibitors Containing New Tricyclic Ring P2-Ligands: Design, Synthesis, Biological, and X-ray Structural Studies.
AID1295721Binding affinity to HIV1 protease I54V mutant2016Journal of medicinal chemistry, Apr-14, Volume: 59, Issue:7
Anti-HIV Drug Discovery and Development: Current Innovations and Future Trends.
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.
AID564029Antiviral activity against HIV1 LAI infected in human MT2 cells by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
AID375205Ratio of EC50 for multidrug-resistant HIV1 isolate MM to EC50 for wild type HIV1 isolate ERS104pre2009Journal of medicinal chemistry, Jul-09, Volume: 52, Issue:13
Design of HIV-1 protease inhibitors with pyrrolidinones and oxazolidinones as novel P1'-ligands to enhance backbone-binding interactions with protease: synthesis, biological evaluation, and protein-ligand X-ray studies.
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).
AID519795Antiviral activity against HIV 2 subtype B clinical isolate expressing 41D protease gene sequence from HIV2 infected patient plasma and PBMC obtained before compound treatment measured after 13 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
AID459075Antiviral activity against multi drug-resistant HIV1 isolate of subtype G in PHA-stimulated human PBMC assessed as inhibition of p24 antigen production2010Bioorganic & medicinal chemistry letters, Feb-01, Volume: 20, Issue:3
Synthesis and biological evaluation of novel allophenylnorstatine-based HIV-1 protease inhibitors incorporating high affinity P2-ligands.
AID1778334Antiviral activity against HIV12021European journal of medicinal chemistry, Aug-05, Volume: 220Design and evaluation of novel piperidine HIV-1 protease inhibitors with potency against DRV-resistant variants.
AID1904904Antiviral activity against wild type HIV1 NL4-32022European journal of medicinal chemistry, Apr-05, Volume: 233Design, synthesis and biological evaluation of protease inhibitors containing morpholine cores with remarkable potency against both HIV-1 subtypes B and C.
AID1380930Resistance index, ratio of EC50 for antiviral activity against APV resistant HIV1 harboring protease L10F/M46I/I50V/I85V mutant infected in human MT4 cells to EC50 for antiviral activity against wild type HIV1 NL4-3 infected in human MT4 cells2018Journal of medicinal chemistry, 05-24, Volume: 61, Issue:10
Design and Synthesis of Highly Potent HIV-1 Protease Inhibitors Containing Tricyclic Fused Ring Systems as Novel P2 Ligands: Structure-Activity Studies, Biological and X-ray Structural Analysis.
AID1465006Antiviral activity against darunavir-resistant HIV1 harboring protease L10I/I15/K20R/L24I/V32I/M36I/M46L/L63P/V82A/L89M mutant infected in human MT4 cells assessed as reduction in p24 Gag protein production2017Bioorganic & medicinal chemistry letters, 11-01, Volume: 27, Issue:21
Design, synthesis, X-ray studies, and biological evaluation of novel macrocyclic HIV-1 protease inhibitors involving the P1'-P2' ligands.
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).
AID375204Ratio of EC50 for multidrug-resistant HIV1 isolate TM to EC50 for wild type HIV1 isolate ERS104pre2009Journal of medicinal chemistry, Jul-09, Volume: 52, Issue:13
Design of HIV-1 protease inhibitors with pyrrolidinones and oxazolidinones as novel P1'-ligands to enhance backbone-binding interactions with protease: synthesis, biological evaluation, and protein-ligand X-ray studies.
AID519541Antiviral activity against HIV1 A17 infected in human MT4 cells harboring protease L10F, V32I, M46I, I47V, Q58E, and I84V mutation derived from viral passages with Lopinavir assessed as reduction in viral cytopathogenicity treated 1 hr post infection meas2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
Characterization of a novel human immunodeficiency virus type 1 protease inhibitor, A-790742.
AID564047Antiviral activity against multidrug-resistant HIV1 isolate MM containing L10I, K43T, M46L, I54V, L63P, A71V, V82A, L90M, and Q92K mutant infected in human PHA-PBMC cells by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
AID738149Inhibition of transferrin receptor-fused wild type HIV1 protease autoprocessing expressed in Escherichia coli at 0.5 to 8 uM by SDS-PAGE analysis2013Journal of medicinal chemistry, May-23, Volume: 56, Issue:10
Extreme multidrug resistant HIV-1 protease with 20 mutations is resistant to novel protease inhibitors with P1'-pyrrolidinone or P2-tris-tetrahydrofuran.
AID1241691Antiviral activity against darunavir-resistant HIV1 P20 infected in human MT4 cells by XTT assay2015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
Design of HIV-1 Protease Inhibitors with Amino-bis-tetrahydrofuran Derivatives as P2-Ligands to Enhance Backbone-Binding Interactions: Synthesis, Biological Evaluation, and Protein-Ligand X-ray Studies.
AID1207422Inhibition of transient outward potassium current (Ito) current in Chinese Hamster Ovary (CHO) K1 cells expressing human Kv4.3 measured using IonWorks Quattro automated patch clamp platform
AID238198Inhibitory activity against human immunodeficiency virus 1 protease2005Journal of medicinal chemistry, May-19, Volume: 48, Issue:10
Structure-based design: synthesis and biological evaluation of a series of novel cycloamide-derived HIV-1 protease inhibitors.
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.
AID668808Antiviral activity against Human immunodeficiency virus 1 3B expressing wild-type protease infected in human MT4 cells2011ACS medicinal chemistry letters, Jun-09, Volume: 2, Issue:6
Disubstituted Bis-THF Moieties as New P2 Ligands in Nonpeptidal HIV-1 Protease Inhibitors.
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.
AID1627021Antiviral activity against wild type HIV1 NL4-3 infected in human MT4 cells by p24 assay2016Journal of medicinal chemistry, 07-28, Volume: 59, Issue:14
Probing Lipophilic Adamantyl Group as the P1-Ligand for HIV-1 Protease Inhibitors: Design, Synthesis, Protein X-ray Structural Studies, and Biological Evaluation.
AID1594350Inhibition of GFP-tagged HIV1 protease transfected in HEK293 cells at 30 uM incubated for 24 hrs by fluorescence microscopy2019Bioorganic & medicinal chemistry, 05-01, Volume: 27, Issue:9
New heteroaryl carbamates: Synthesis and biological screening in vitro and in mammalian cells of wild-type and mutant HIV-protease inhibitors.
AID649702Ratio of EC50 for HIV1 MDR/C harboring protease L10I, I15V, K20R, L24I, M36I, M46L, I54V, I62V, L63P, K70Q, V82A, and L89 mutant to EC50 for HIV1 ERS104 harboring wild type protease2012Bioorganic & medicinal chemistry letters, Mar-15, Volume: 22, Issue:6
Substituent effects on P2-cyclopentyltetrahydrofuranyl urethanes: design, synthesis, and X-ray studies of potent HIV-1 protease inhibitors.
AID537768Antiviral activity against Human immunodeficiency virus 1 clade A isolated from HIV-AIDS patient2010Journal of medicinal chemistry, Nov-11, Volume: 53, Issue:21
Structure-based design, synthesis, and structure-activity relationship studies of HIV-1 protease inhibitors incorporating phenyloxazolidinones.
AID1482922Ratio of IC50 for amprenavir-resistant HIV1 NL4-3 harboring protease L10F/V32I/L33F/M46L/I54M/A71V mutant infected in human MT4 cells to IC50 for wild type HIV1 NL4.3 infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID1295712Binding affinity to HIV1 protease V82A mutant2016Journal of medicinal chemistry, Apr-14, Volume: 59, Issue:7
Anti-HIV Drug Discovery and Development: Current Innovations and Future Trends.
AID1604130Antiviral activity against HIV1 LAI infected in human 12D7 cells overexpressing p-gp assessed as reduction in P24 level at 0.08 to 1.25 uM incubated for 3 days followed by replacement of fresh medium containing compound and measured after 3 days by ELISA2020Journal of medicinal chemistry, 03-12, Volume: 63, Issue:5
Potential Tools for Eradicating HIV Reservoirs in the Brain: Development of Trojan Horse Prodrugs for the Inhibition of P-Glycoprotein with Anti-HIV-1 Activity.
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.
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.
AID269303Inhibition of HIV1 protease2006Journal of medicinal chemistry, Aug-24, Volume: 49, Issue:17
Structure-based design of novel HIV-1 protease inhibitors to combat drug resistance.
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.
AID1725422Antiviral activity against HIV1 DRVR P30 infected in human MT4 cells incubated for 7 days by fully automated chemiluminescent enzyme immunoassay2020ACS medicinal chemistry letters, Oct-08, Volume: 11, Issue:10
Design, Synthesis, and X-ray Studies of Potent HIV-1 Protease Inhibitors with P2-Carboxamide Functionalities.
AID738151Inhibition of transferrin receptor-fused HIV1 protease Q7K, L10F, I13V, I15V, D30V, V32I, L33F, E35D, M36I, S37N, I47V, I54L, Q58E, I62V, L63P, A71V, I84V, N88D, L89T, L90M mutant autoprocessing expressed in Escherichia coli at 100 to 150 uM by SDS-PAGE a2013Journal of medicinal chemistry, May-23, Volume: 56, Issue:10
Extreme multidrug resistant HIV-1 protease with 20 mutations is resistant to novel protease inhibitors with P1'-pyrrolidinone or P2-tris-tetrahydrofuran.
AID1627022Antiviral activity against multi-drug resistant HIV1B harboring protease L10I/L33I/M36I/M46I/F53L/K55R/I62V/L63P/A71V/G73S/V82A/L90M/I93L mutant infected in human MT4 cells by p24 assay2016Journal of medicinal chemistry, 07-28, Volume: 59, Issue:14
Probing Lipophilic Adamantyl Group as the P1-Ligand for HIV-1 Protease Inhibitors: Design, Synthesis, Protein X-ray Structural Studies, and Biological Evaluation.
AID566851Antiviral activity against HIV-1 MDR/MM infected in human PHA-PBM cells assessed as inhibition of p24 Gag protein production2011Journal of medicinal chemistry, Jan-27, Volume: 54, Issue:2
Design and synthesis of potent HIV-1 protease inhibitors incorporating hexahydrofuropyranol-derived high affinity P(2) ligands: structure-activity studies and biological evaluation.
AID613954Antiviral activity against Human immunodeficiency virus 1 LAI infected in human MT2 cells assessed as inhibition of p24 Gag protein production2011Journal of medicinal chemistry, Aug-25, Volume: 54, Issue:16
Design of HIV-1 protease inhibitors with C3-substituted hexahydrocyclopentafuranyl urethanes as P2-ligands: synthesis, biological evaluation, and protein-ligand X-ray crystal structure.
AID1227241Binding affinity to HIV1 protease PR20 mutant expressed in Escherichia coli BL21 (DE3) assessed as ligand dissociation constant by ITC method2015Journal of medicinal chemistry, Jun-25, Volume: 58, Issue:12
Substituted Bis-THF Protease Inhibitors with Improved Potency against Highly Resistant Mature HIV-1 Protease PR20.
AID1687689Inhibition of recombinant HIV1 protease measured assessed as hydrolysis of fluorogenic substrate by FRET assay2020European journal of medicinal chemistry, Jan-15, Volume: 186Rational design and Structure-Activity relationship of coumarin derivatives effective on HIV-1 protease and partially on HIV-1 reverse transcriptase.
AID519792Antiviral activity against HIV 2 subtype B clinical isolate expressing 14Y-19P-61N-64V-71I-90M-95I protease gene sequence from HIV2 infected patient plasma and PBMC obtained at T2 during compound treatment measured after 3 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
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.
AID1482924Ratio of IC50 for indinavir-resistant HIV1 NL4-3 harboring protease L10F/L24I/M46I/I54V/L63P/A71V/G73S/V82T mutant infected in human MT4 cells to IC50 for wild type HIV1 NL4.3 infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID1295713Binding affinity to HIV1 protease N88D mutant2016Journal of medicinal chemistry, Apr-14, Volume: 59, Issue:7
Anti-HIV Drug Discovery and Development: Current Innovations and Future Trends.
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.
AID1589133Antiviral activity against HIV1 NL4-3 infected in human MT4 cells assessed as reduction in p24 Gag protein production incubated for 6 days and measured on day 7 by chemiluminescent enzyme immunoassay2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Potent HIV-1 protease inhibitors incorporating squaramide-derived P2 ligands: Design, synthesis, and biological evaluation.
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.
AID613963Ratio of IC50 for Human immunodeficiency virus 1 MDR/G to IC50 for wild-type Human immunodeficiency virus 1 ERS104pre2011Journal of medicinal chemistry, Aug-25, Volume: 54, Issue:16
Design of HIV-1 protease inhibitors with C3-substituted hexahydrocyclopentafuranyl urethanes as P2-ligands: synthesis, biological evaluation, and protein-ligand X-ray crystal structure.
AID375206Ratio of EC50 for multidrug-resistant HIV1 isolate C to EC50 for wild type HIV1 isolate ERS104pre2009Journal of medicinal chemistry, Jul-09, Volume: 52, Issue:13
Design of HIV-1 protease inhibitors with pyrrolidinones and oxazolidinones as novel P1'-ligands to enhance backbone-binding interactions with protease: synthesis, biological evaluation, and protein-ligand X-ray studies.
AID1252236Antiviral activity against HIV-1 LAI infected in MT-2 cells measured after 7 days by MTT assay2015Bioorganic & medicinal chemistry letters, Nov-01, Volume: 25, Issue:21
Structure-based design, synthesis, X-ray studies, and biological evaluation of novel HIV-1 protease inhibitors containing isophthalamide-derived P2-ligands.
AID537771Antiviral activity against multidrug resistant Human immunodeficiency virus 1 harboring protease M46I, I54V, V82A and L90M mutant2010Journal of medicinal chemistry, Nov-11, Volume: 53, Issue:21
Structure-based design, synthesis, and structure-activity relationship studies of HIV-1 protease inhibitors incorporating phenyloxazolidinones.
AID415247Antiviral activity against HIV1 bearing protease gene with A17 mutation infected in human MT4 cells after 5 days by MTT assay2009Journal 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.
AID525278Antimicrobial activity against chloroquine-resistant Plasmodium falciparum Dd2 after 48 hrs2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
Antimalarial asexual stage-specific and gametocytocidal activities of HIV protease inhibitors.
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.
AID564045Antiviral activity against multidrug-resistant HIV1 isolate G containing L10I, V11I, T12E, I15V, L19I,R41K, M46L, L63P, A71T, V82A, and L90M mutant infected in human PHA-PBMC cells by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
AID1669456Inhibition of HIV1 NL4-3 protease I50V/A71V mutant expressed in Escherichia coli TAP-106 cells using EDANS/DABCYL-labelled 10-amino acid containing protease cleavage site as substrate preincubated for 1 hr followed by substrate addition and measured for 62020Journal of medicinal chemistry, 08-13, Volume: 63, Issue:15
Structural Analysis of Potent Hybrid HIV-1 Protease Inhibitors Containing Bis-tetrahydrofuran in a Pseudosymmetric Dipeptide Isostere.
AID525494Cmax in HIV-infected patient2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
Antimalarial asexual stage-specific and gametocytocidal activities of HIV protease inhibitors.
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.
AID1725421Antiviral activity against DRV resistant HIV1 infected in human MT4 cells incubated for 7 days by fully automated chemiluminescent enzyme immunoassay2020ACS medicinal chemistry letters, Oct-08, Volume: 11, Issue:10
Design, Synthesis, and X-ray Studies of Potent HIV-1 Protease Inhibitors with P2-Carboxamide Functionalities.
AID415237Antiviral activity against HIV1 assessed as inhibition of viral replication2009Journal 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.
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.
AID343017Inhibition of HIV1 recombinant protease A71V/V82T/I84V mutant expressed in Escherichia coli by spectrophotometric assay2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
Inorganic polyhedral metallacarborane inhibitors of HIV protease: a new approach to overcoming antiviral resistance.
AID673378Antiviral activity against Human immunodeficiency virus 1 HXB2 expressing protease M6 mutant infected in human MT4 cells after 5 days by MTT assay2012Bioorganic & medicinal chemistry letters, Aug-01, Volume: 22, Issue:15
Enamino-oxindole HIV protease inhibitors.
AID649699Antiviral activity against HIV1 MDR/G harboring protease L10I, V11I, T12E, I15V, L19I, R41K, M46L, L63P, A71T, V82A, and L90M mutant infected in PHA-PBM cells assessed as inhibition of p24 Gag protein production2012Bioorganic & medicinal chemistry letters, Mar-15, Volume: 22, Issue:6
Substituent effects on P2-cyclopentyltetrahydrofuranyl urethanes: design, synthesis, and X-ray studies of potent HIV-1 protease inhibitors.
AID1295710Binding affinity to HIV1 protease L63P mutant2016Journal of medicinal chemistry, Apr-14, Volume: 59, Issue:7
Anti-HIV Drug Discovery and Development: Current Innovations and Future Trends.
AID1482917Ratio of IC50 for darunavir-resistant HIV1 derived from 30 passages harboring protease L10I/I15V/K20R/L24I/V32I/M36I/M46L/L63P/K70R/V82A/I84V/L89M mutant infected in human MT4 cells to IC50 for wild type HIV1 NL4.3 infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID1207456Inhibition of rapid delayed inward rectifying potassium current (IKr) in Chinese hamster ovary (CHO) K1 cells stably expressing hERG measured using IonWorks Quattro automated patch clamp platform
AID1380925Antiviral activity against ATV resistant HIV1 harboring protease L23I/E34Q/K43I/M46I/I50L/G51A/L63P/A71V/V82A/T91A mutant infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by chemiluminescent enzyme immunoassay2018Journal of medicinal chemistry, 05-24, Volume: 61, Issue:10
Design and Synthesis of Highly Potent HIV-1 Protease Inhibitors Containing Tricyclic Fused Ring Systems as Novel P2 Ligands: Structure-Activity Studies, Biological and X-ray Structural Analysis.
AID1604116Inhibition of P-gp in human 12D7-MDR cells using NBD-Aba as substrate after 30 mins by flow cytometric analysis2020Journal of medicinal chemistry, 03-12, Volume: 63, Issue:5
Potential Tools for Eradicating HIV Reservoirs in the Brain: Development of Trojan Horse Prodrugs for the Inhibition of P-Glycoprotein with Anti-HIV-1 Activity.
AID1252235Inhibition of HIV-1 protease by fluorometric assay2015Bioorganic & medicinal chemistry letters, Nov-01, Volume: 25, Issue:21
Structure-based design, synthesis, X-ray studies, and biological evaluation of novel HIV-1 protease inhibitors containing isophthalamide-derived P2-ligands.
AID649697Antiviral activity against HIV1 MDR/B harboring protease L10I, K14R, L33I, M36I, M46I, F53I, K55R, I62V, L63P, A71V, G73S, V82A, L90M, I93L mutant infected in PHA-PBM cells assessed as inhibition of p24 Gag protein production2012Bioorganic & medicinal chemistry letters, Mar-15, Volume: 22, Issue:6
Substituent effects on P2-cyclopentyltetrahydrofuranyl urethanes: design, synthesis, and X-ray studies of potent HIV-1 protease inhibitors.
AID1465009Antiviral activity against HIV-1 harboring protease I50V mutant2017Bioorganic & medicinal chemistry letters, 11-01, Volume: 27, Issue:21
Design, synthesis, X-ray studies, and biological evaluation of novel macrocyclic HIV-1 protease inhibitors involving the P1'-P2' ligands.
AID1230028Antiviral activity against multidrug resistant HIV1 MDR/C containing protease mutant infected in human MT4 cells assessed as inhibition of p24 Gag protein production relative to wild type2015Journal of medicinal chemistry, Jul-09, Volume: 58, Issue:13
Structure-based design of potent HIV-1 protease inhibitors with modified P1-biphenyl ligands: synthesis, biological evaluation, and enzyme-inhibitor X-ray structural studies.
AID1687688Antiviral activity against wild type HIV1 assessed as inhibitory effect on virus producing cells at 10 uM by cell based inhibition assay2020European journal of medicinal chemistry, Jan-15, Volume: 186Rational design and Structure-Activity relationship of coumarin derivatives effective on HIV-1 protease and partially on HIV-1 reverse transcriptase.
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.
AID328064Inhibition of HIV1 protease dimerization in COS7 cells at 0.1 uM assessed as decrease in luciferase response ratio2007The Journal of biological chemistry, Sep-28, Volume: 282, Issue:39
Potent inhibition of HIV-1 replication by novel non-peptidyl small molecule inhibitors of protease dimerization.
AID519543Antiviral activity against HIV1 P25 infected in human MT4 cells harboring protease L10F, G16E, V32I, M46I, I47A, H69Y, I84V, and T91S mutation derived from viral passages with Lopinavir assessed as reduction in viral cytopathogenicity treated 1 hr post in2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
Characterization of a novel human immunodeficiency virus type 1 protease inhibitor, A-790742.
AID564062Antiviral activity against HIV1 expressing protease L10F/M46L/I50V/A71Vmutant infected in human MT4 cells selected at 1 uM of GRL-286 by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
AID249503Effective concentration against PI-Resistant HIV strain (M1)2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Discovery and selection of TMC114, a next generation HIV-1 protease inhibitor.
AID1778341Antiviral activity against DRV resistant HIV1 infected in human MT4 cells assessed as reduction in viral replication by measuring reduction in p24 Gag production by chemiluminescent-EIA2021European journal of medicinal chemistry, Aug-05, Volume: 220Design and evaluation of novel piperidine HIV-1 protease inhibitors with potency against DRV-resistant variants.
AID390728Inhibition of HIV1 protease V32I mutant by uncompetitive binding2008Journal of medicinal chemistry, Oct-23, Volume: 51, Issue:20
Solution kinetics measurements suggest HIV-1 protease has two binding sites for darunavir and amprenavir.
AID1778344Antiviral activity against HIV1 subtype C isolate 119412021European journal of medicinal chemistry, Aug-05, Volume: 220Design and evaluation of novel piperidine HIV-1 protease inhibitors with potency against DRV-resistant variants.
AID343014Inhibition of wild-type HIV1 BH10 protease expressed in Escherichia coli by spectrophotometric assay2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
Inorganic polyhedral metallacarborane inhibitors of HIV protease: a new approach to overcoming antiviral resistance.
AID1482912Antiviral activity against darunavir-resistant HIV1 derived from 20 passages harboring protease L10I/I15V/K20R/L24I/V32I/M36I/M46L/L63P/V82A/L89M mutant infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by chemil2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID770187Ratio of EC50 for wild type HIV1 ERS104pre to EC50 for multidrug-resistant HIV1 C harboring L10I/I15V/K20R/L24I/M36I/M46L/I54V/I62V/L63P/K70Q/V82A/L89M protease-encoding region mutant2013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Highly potent HIV-1 protease inhibitors with novel tricyclic P2 ligands: design, synthesis, and protein-ligand X-ray studies.
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.
AID343026Ratio of Ki for HIV1 recombinant protease L10I/I15V/E35D/N37S/R41K/I62V/L63P/A71V/G73S/L90M mutant to Ki for wild-type HIV1 BH10 protease2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
Inorganic polyhedral metallacarborane inhibitors of HIV protease: a new approach to overcoming antiviral resistance.
AID1704275Half life in human liver microsomes2020European journal of medicinal chemistry, Oct-15, Volume: 204Novel HIV-1 capsid-targeting small molecules of the PF74 binding site.
AID417027Effect on tenofovir disoproxil fumarate metabolism in ritonavir booster drug treated healthy human assessed as change in plasma Cmax of tenofovir at 300 mg, po, BID co-administered with 300 mg once daily dose of tenofovir disoproxil fumarate2007Antimicrobial 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.
AID564031Antiviral activity against HIV2 ROD infected in human MT2 cells by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
AID1658404Resistance index, ratio of EC50 for antiviral activity against VSV-G pseudotyped darunavir-resistant HIV1 NL4-3 in human SupT1 cells infected with supernatants of virus particles extracted from human 293T cells infected with darunavir-resistant HIV1 NL4-32020ACS medicinal chemistry letters, Jun-11, Volume: 11, Issue:6
Novel HIV-1 Protease Inhibitors with Morpholine as the P2 Ligand to Enhance Activity against DRV-Resistant Variants.
AID328063Antiviral activity against HIV1 LAI in MT2 cells2007The Journal of biological chemistry, Sep-28, Volume: 282, Issue:39
Potent inhibition of HIV-1 replication by novel non-peptidyl small molecule inhibitors of protease dimerization.
AID446191Antiviral activity against HIV1 LAI infected in human MT2 cells by MTT assay2009Journal of medicinal chemistry, Dec-10, Volume: 52, Issue:23
Design, synthesis, protein-ligand X-ray structure, and biological evaluation of a series of novel macrocyclic human immunodeficiency virus-1 protease inhibitors to combat drug resistance.
AID1658403Antiviral activity against VSV-G pseudotyped wild type HIV1 NL4-3 in human SupT1 cells infected with supernatants of virus particles extracted from human 293T cells infected with wild type HIV1 NL4-3 incubated for 48 hrs assessed as inhibition of late sta2020ACS medicinal chemistry letters, Jun-11, Volume: 11, Issue:6
Novel HIV-1 Protease Inhibitors with Morpholine as the P2 Ligand to Enhance Activity against DRV-Resistant Variants.
AID1778345Antiviral activity against HIV1 subtype C isolate-plndie2021European journal of medicinal chemistry, Aug-05, Volume: 220Design and evaluation of novel piperidine HIV-1 protease inhibitors with potency against DRV-resistant variants.
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.
AID390729Inhibition of HIV1 protease V32I mutant by enzyme inhibition2008Journal of medicinal chemistry, Oct-23, Volume: 51, Issue:20
Solution kinetics measurements suggest HIV-1 protease has two binding sites for darunavir and amprenavir.
AID1767120Antiviral activity against HIV1 NL4-3 VSV-G pseudotyped virus infected in human SUP-T1 cells assessed as inhibition of early stage viral production at 10 uM measured after 48 hrs by luciferase assay relative to control2021European journal of medicinal chemistry, Aug-05, Volume: 220Design and biological evaluation of cinnamic and phenylpropionic amide derivatives as novel dual inhibitors of HIV-1 protease and reverse transcriptase.
AID1669457Antiviral activity against wild-type HIV1 NL4-3 infected in human TZM-bl cells infected with supernatants from virus-infected human 293T cells treated with compound for 18 hrs assessed as reduction in viral replication by luciferase assay2020Journal of medicinal chemistry, 08-13, Volume: 63, Issue:15
Structural Analysis of Potent Hybrid HIV-1 Protease Inhibitors Containing Bis-tetrahydrofuran in a Pseudosymmetric Dipeptide Isostere.
AID1665063Inhibition of HIV-1 protease expressed in Escherichia coli using Arg-Glu(EDANS)-Ser-Gln-Asn-Tyr-Pro-Ile-Val-Gln-Lys(DABCYL)-Arg as substrate preincubated for 20 to 30 mins followed by substrate addition by FRET based assay2020Bioorganic & medicinal chemistry, 08-15, Volume: 28, Issue:16
Design and biological evaluation of novel HIV-1 protease inhibitors with isopropanol as P1' ligand to enhance binding with S1' subsite.
AID1604118Inhibition of P-gp in human CMEC/D3 cells using NBD-Aba as substrate after 30 mins by flow cytometric analysis2020Journal of medicinal chemistry, 03-12, Volume: 63, Issue:5
Potential Tools for Eradicating HIV Reservoirs in the Brain: Development of Trojan Horse Prodrugs for the Inhibition of P-Glycoprotein with Anti-HIV-1 Activity.
AID343025Ratio of Ki for HIV1 recombinant protease V32I/I47A mutant to Ki for wild-type HIV1 BH10 protease2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
Inorganic polyhedral metallacarborane inhibitors of HIV protease: a new approach to overcoming antiviral resistance.
AID1482925Ratio of IC50 for nelfinavir-resistant HIV1 NL4-3 harboring protease L10F/K20T/D30N/K45I/A71V/V77I mutant infected in human MT4 cells to IC50 for wild type HIV1 NL4.3 infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID1767118Antiviral activity against HIV1 NL4-3 Env-deficient VSV-G pseudotyped virus infected in human SUP-T1 cells assessed as inhibition of late stage viral production when host cells were infected with compound pretreated viral particles at 10 uM measured after2021European journal of medicinal chemistry, Aug-05, Volume: 220Design and biological evaluation of cinnamic and phenylpropionic amide derivatives as novel dual inhibitors of HIV-1 protease and reverse transcriptase.
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.
AID415245Antiviral activity against wild-type HIV1 pNL4-3 infected in human MT4 cells after 5 days by MTT assay2009Journal 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.
AID1607530Antiviral activity against darunavir resistant HIV1 harboring protease P30 L10I/I15V/K20R/L24I/V32I/M36I/ M46L/L63P/K70Q/V82A/I84V/L89M mutant infected in human MT4 cells assessed as reduction in p24 Gag production2020Journal of medicinal chemistry, 05-14, Volume: 63, Issue:9
Structure-Based Design of Highly Potent HIV-1 Protease Inhibitors Containing New Tricyclic Ring P2-Ligands: Design, Synthesis, Biological, and X-ray Structural Studies.
AID613956Antiviral activity against multidrug-resistant Human immunodeficiency virus 1 subtype B harboring protease L10I, L33I, M36I, M46I, F53L, K55R, I62 V, L63P, A71 V, G73S, V82A, L90M, I93L mutation infected in PHA-stimulated human PBMC assessed as inhibition2011Journal of medicinal chemistry, Aug-25, Volume: 54, Issue:16
Design of HIV-1 protease inhibitors with C3-substituted hexahydrocyclopentafuranyl urethanes as P2-ligands: synthesis, biological evaluation, and protein-ligand X-ray crystal structure.
AID1904899Selectivity index, ratio EC50 for Antiviral activity against DRV resistant HIV1 over EC50 for wild type HIV1 NL4-32022European journal of medicinal chemistry, Apr-05, Volume: 233Design, synthesis and biological evaluation of protease inhibitors containing morpholine cores with remarkable potency against both HIV-1 subtypes B and C.
AID1461872Inhibition of recombinant HIV-1 GFP-tagged protease expressed in HEK293 cells assessed as increase in GFP-tagged protease accumulation at 10 uM after 24 hrs by Western blot analysis2017Bioorganic & medicinal chemistry, 09-01, Volume: 25, Issue:17
Synthesis and biological evaluation in vitro and in mammalian cells of new heteroaryl carboxyamides as HIV-protease inhibitors.
AID668817Antiviral activity against Human immunodeficiency virus 1 3B expressing wild-type protease infected in human MT4 cells in presence of 50% human serum2011ACS medicinal chemistry letters, Jun-09, Volume: 2, Issue:6
Disubstituted Bis-THF Moieties as New P2 Ligands in Nonpeptidal HIV-1 Protease Inhibitors.
AID343016Inhibition of HIV1 recombinant protease M46I/A71V/V82T/I84V mutant expressed in Escherichia coli by spectrophotometric assay2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
Inorganic polyhedral metallacarborane inhibitors of HIV protease: a new approach to overcoming antiviral resistance.
AID1665067Antiviral activity against VSV-G pseudotyped darunavir-resistant HIV1 NL4-3 V32I, L33F,I54M and I84V mutant infected in human SUPT1 cells at 100 nM infected with supernatants from virus-infected human 293T cells after 48 hrs by luciferase assay relative t2020Bioorganic & medicinal chemistry, 08-15, Volume: 28, Issue:16
Design and biological evaluation of novel HIV-1 protease inhibitors with isopropanol as P1' ligand to enhance binding with S1' subsite.
AID446204Selectivity ratio of IC50 for multidrug-resistant HIV1 isolate MM to IC50 for wild type HIV1 isolate ERS104pre2009Journal of medicinal chemistry, Dec-10, Volume: 52, Issue:23
Design, synthesis, protein-ligand X-ray structure, and biological evaluation of a series of novel macrocyclic human immunodeficiency virus-1 protease inhibitors to combat drug resistance.
AID519785Antiviral activity against HIV 2 subtype A clinical isolate expressing 10I-17D-40D-43I-46V-66V/A-70R/K protease gene sequence from HIV2 infected patient plasma and PBMC obtained before compound treatment measured after 13 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
AID237955Percentage of remaining compound after 30 min incubation in rat liver microsomes was determined as metabolic stability2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Discovery and selection of TMC114, a next generation HIV-1 protease inhibitor.
AID564033Selectivity ratio of CC50 for human MT2 cells to EC50 for HIV1 LAI infected human MT2 cells2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
AID459078Antiviral activity against multi drug-resistant HIV1 isolate of subtype JSL in PHA-stimulated human PBMC assessed as inhibition of p24 antigen production2010Bioorganic & medicinal chemistry letters, Feb-01, Volume: 20, Issue:3
Synthesis and biological evaluation of novel allophenylnorstatine-based HIV-1 protease inhibitors incorporating high affinity P2-ligands.
AID1350508Ratio of EC50 for DRV-resistant protease V32I/L33F/I54M/V82I mutant in HIV1 transfected in 293T cells to EC50 for wild-type HIV-1 pNL4-3 infected in CMVdeltaR8.91 transfected 293T cells2018Journal of medicinal chemistry, 06-28, Volume: 61, Issue:12
Identification of Highly Potent Human Immunodeficiency Virus Type-1 Protease Inhibitors against Lopinavir and Darunavir Resistant Viruses from Allophenylnorstatine-Based Peptidomimetics with P2 Tetrahydrofuranylglycine.
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.
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.
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.
AID1295715Binding affinity to HIV1 protease G73S mutant2016Journal of medicinal chemistry, Apr-14, Volume: 59, Issue:7
Anti-HIV Drug Discovery and Development: Current Innovations and Future Trends.
AID396261Ratio of EC50 for HIV1 drug resistant mutant isolates from protease inhibitor treated HIV patient to EC50 for drug sensitive HIV1 NL4-32007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
AID1380926Antiviral activity against LPV resistant HIV1 harboring protease L10F/M46/I54V/V82A mutant infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by chemiluminescent enzyme immunoassay2018Journal of medicinal chemistry, 05-24, Volume: 61, Issue:10
Design and Synthesis of Highly Potent HIV-1 Protease Inhibitors Containing Tricyclic Fused Ring Systems as Novel P2 Ligands: Structure-Activity Studies, Biological and X-ray Structural Analysis.
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.
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).
AID1627023Antiviral activity against multi-drug resistant HIV1C harboring protease L10I/I15V/K20R/L24I/M36I/M46L/I54V/I62V/L63P/K70Q/V82A/L89M mutant infected in human MT4 cells by p24 assay2016Journal of medicinal chemistry, 07-28, Volume: 59, Issue:14
Probing Lipophilic Adamantyl Group as the P1-Ligand for HIV-1 Protease Inhibitors: Design, Synthesis, Protein X-ray Structural Studies, and Biological Evaluation.
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.
AID1380922Inhibition of wild type HIV-1 NL4-3 protease expressed in Escherichia coli Rosetta (DE3)pLysS using Ac-Thr-Ile-Nle-Nle-Gln-Arg-NH2 as substrate by fluorescence assay2018Journal of medicinal chemistry, 05-24, Volume: 61, Issue:10
Design and Synthesis of Highly Potent HIV-1 Protease Inhibitors Containing Tricyclic Fused Ring Systems as Novel P2 Ligands: Structure-Activity Studies, Biological and X-ray Structural Analysis.
AID566850Antiviral activity against HIV-1 MDR/TM infected in human PHA-PBM cells assessed as inhibition of p24 Gag protein production2011Journal of medicinal chemistry, Jan-27, Volume: 54, Issue:2
Design and synthesis of potent HIV-1 protease inhibitors incorporating hexahydrofuropyranol-derived high affinity P(2) ligands: structure-activity studies and biological evaluation.
AID613958Antiviral activity against multidrug-resistant Human immunodeficiency virus 1 MDR/G harboring protease L10I, V11I, T12E, I15 V, L19I, R41K, M46L, L63P, A71T, V82A, L90 M mutation infected in PHA-stimulated human PBMC assessed as inhibition of p24 Gag prot2011Journal of medicinal chemistry, Aug-25, Volume: 54, Issue:16
Design of HIV-1 protease inhibitors with C3-substituted hexahydrocyclopentafuranyl urethanes as P2-ligands: synthesis, biological evaluation, and protein-ligand X-ray crystal structure.
AID673372Antiviral activity against Human immunodeficiency virus 1 HXB2 expressing protease M8 mutant infected in human MT4 cells after 5 days by MTT assay2012Bioorganic & medicinal chemistry letters, Aug-01, Volume: 22, Issue:15
Enamino-oxindole HIV protease inhibitors.
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.
AID446195Antiviral activity against multidrug-resistant HIV1 isolate C infected in PHA-stimulated human PBMC assessed as inhibition of p24 gap protein production2009Journal of medicinal chemistry, Dec-10, Volume: 52, Issue:23
Design, synthesis, protein-ligand X-ray structure, and biological evaluation of a series of novel macrocyclic human immunodeficiency virus-1 protease inhibitors to combat drug resistance.
AID1350507Inhibition of DRV-resistant protease V32I/L33F/I54M/I84V mutant in HIV1 transfected in 293T cells after 48 hrs by lentiviral vector-based luciferase reporter gene assay2018Journal of medicinal chemistry, 06-28, Volume: 61, Issue:12
Identification of Highly Potent Human Immunodeficiency Virus Type-1 Protease Inhibitors against Lopinavir and Darunavir Resistant Viruses from Allophenylnorstatine-Based Peptidomimetics with P2 Tetrahydrofuranylglycine.
AID446190Inhibition of HIV1 protease2009Journal of medicinal chemistry, Dec-10, Volume: 52, Issue:23
Design, synthesis, protein-ligand X-ray structure, and biological evaluation of a series of novel macrocyclic human immunodeficiency virus-1 protease inhibitors to combat drug resistance.
AID564038Antiviral activity against HIV1 expressing protease L10F/D30N/K45I/A71V/T74S mutant infected in human MT4 cells selected at 5 uM of nelfinavir by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
AID1669454Inhibition of HIV1 NL4-3 protease expressed in Escherichia coli TAP-106 cells using EDANS/DABCYL-labelled 10-amino acid containing protease cleavage site as substrate preincubated for 1 hr followed by substrate addition and measured for 60 mins by FRET as2020Journal of medicinal chemistry, 08-13, Volume: 63, Issue:15
Structural Analysis of Potent Hybrid HIV-1 Protease Inhibitors Containing Bis-tetrahydrofuran in a Pseudosymmetric Dipeptide Isostere.
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).
AID311574Inhibition of HIV1 protease2007Bioorganic & medicinal chemistry, Dec-15, Volume: 15, Issue:24
Darunavir, a conceptually new HIV-1 protease inhibitor for the treatment of drug-resistant HIV.
AID1607526Antiviral activity against HIV1 LAI infected in human MT2 cells2020Journal of medicinal chemistry, 05-14, Volume: 63, Issue:9
Structure-Based Design of Highly Potent HIV-1 Protease Inhibitors Containing New Tricyclic Ring P2-Ligands: Design, Synthesis, Biological, and X-ray Structural Studies.
AID1491222Inhibition of HIV1 protease expressed in Escherichia coli using Arg-Glu (EDANS)-Ser-GlnAsn-Tyr-Pro-Ile-Val-Gln-Lys(DABCYL)-Arg as substrate preincubated for 20 to 30 mins followed by substrate addition measured for 10 mins by FRET method2017European journal of medicinal chemistry, Sep-08, Volume: 137Design and synthesis of potent HIV-1 protease inhibitors with (S)-tetrahydrofuran-tertiary amine-acetamide as P2-ligand: Structure-activity studies and biological evaluation.
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.
AID238117Binding affinity against HIV-1 protease2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Design of HIV-1 protease inhibitors active on multidrug-resistant virus.
AID649696Antiviral activity against wild type HIV1 ERS104 infected in PHA-PBM cells assessed as inhibition of p24 Gag protein production2012Bioorganic & medicinal chemistry letters, Mar-15, Volume: 22, Issue:6
Substituent effects on P2-cyclopentyltetrahydrofuranyl urethanes: design, synthesis, and X-ray studies of potent HIV-1 protease inhibitors.
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.
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.
AID673369Inhibition of Human immunodeficiency virus protease M9 mutant expressed in Escherichia coli using ArgGlu(EDANS)SerGlnAsnTyrProIleValGlnLys(DABCYL)Arg as substrate preincubated with compound for 0.5 to 1 min measured by fluorometric analysis at pH 6.52012Bioorganic & medicinal chemistry letters, Aug-01, Volume: 22, Issue:15
Enamino-oxindole HIV protease inhibitors.
AID1241694Resistance index, ratio of EC50 for darunavir-resistant HIV1 P20 infected in human MT4 cells by XTT assay to EC50 for HIV1 NL4-3 infected in human MT4 cells by XTT assay2015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
Design of HIV-1 Protease Inhibitors with Amino-bis-tetrahydrofuran Derivatives as P2-Ligands to Enhance Backbone-Binding Interactions: Synthesis, Biological Evaluation, and Protein-Ligand X-ray Studies.
AID1607533Selectivity ratio of IC50 for antiviral activity against darunavir resistant HIV1 P30 harboring protease L10I/I15V/K20R/L24I/V32I/M36I/ M46L/L63P/K70Q/V82A/I84V/L89M mutant infected in human MT4 cells to IC50 for antiviral activity against wild-type HIV1 2020Journal of medicinal chemistry, 05-14, Volume: 63, Issue:9
Structure-Based Design of Highly Potent HIV-1 Protease Inhibitors Containing New Tricyclic Ring P2-Ligands: Design, Synthesis, Biological, and X-ray Structural Studies.
AID649701Ratio of EC50 for HIV1 MDR/B harboring protease L10I, K14R, L33I, M36I, M46I, F53I, K55R, I62V, L63P, A71V, G73S, V82A, L90M, I93L mutant to EC50 for HIV1 ERS104 harboring wild type protease2012Bioorganic & medicinal chemistry letters, Mar-15, Volume: 22, Issue:6
Substituent effects on P2-cyclopentyltetrahydrofuranyl urethanes: design, synthesis, and X-ray studies of potent HIV-1 protease inhibitors.
AID1778346Resistance index, ratio of IC50 for DRV resistant HIV1 infected in human MT4 cells to IC50 for HIV1 NL4-3 infected in human MT4 cells2021European journal of medicinal chemistry, Aug-05, Volume: 220Design and evaluation of novel piperidine HIV-1 protease inhibitors with potency against DRV-resistant variants.
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.
AID1350509Ratio of EC50 for DRV-resistant protease V32I/L33F/I54M/I84V mutant in HIV1 transfected in 293T cells to EC50 for wild-type HIV-1 pNL4-3 infected in CMVdeltaR8.91 transfected 293T cells2018Journal of medicinal chemistry, 06-28, Volume: 61, Issue:12
Identification of Highly Potent Human Immunodeficiency Virus Type-1 Protease Inhibitors against Lopinavir and Darunavir Resistant Viruses from Allophenylnorstatine-Based Peptidomimetics with P2 Tetrahydrofuranylglycine.
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.
AID1230023Antiviral activity against wild type HIV1 ERS104P infected in human MT4 cells assessed as virus-induced cytopathic effect by MTT assay2015Journal of medicinal chemistry, Jul-09, Volume: 58, Issue:13
Structure-based design of potent HIV-1 protease inhibitors with modified P1-biphenyl ligands: synthesis, biological evaluation, and enzyme-inhibitor X-ray structural studies.
AID566852Antiviral activity against HIV-1 MDR/JSL infected in human PHA-PBM cells assessed as inhibition of p24 Gag protein production2011Journal of medicinal chemistry, Jan-27, Volume: 54, Issue:2
Design and synthesis of potent HIV-1 protease inhibitors incorporating hexahydrofuropyranol-derived high affinity P(2) ligands: structure-activity studies and biological evaluation.
AID1295708Binding affinity to HIV1 protease L10I mutant2016Journal of medicinal chemistry, Apr-14, Volume: 59, Issue:7
Anti-HIV Drug Discovery and Development: Current Innovations and Future Trends.
AID1657083Inhibition of HIV1 protease by FRET assay2020Bioorganic & medicinal chemistry letters, 04-01, Volume: 30, Issue:7
Design, synthesis and biological evaluation of HIV-1 protease inhibitors with morpholine derivatives as P2 ligands in combination with cyclopropyl as P1' ligand.
AID564048Antiviral activity against multidrug-resistant HIV1 isolate JSL containing L10I, L24I, I33F, E35D, M36I, N37S, M46L, I54V, R57K, I62V, L63P, A71V, G73S, and V82A mutant infected in human PHA-PBMC cells by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
AID1904908Antiviral activity against HIV1 NL4-3 Env-deficient VSV-G pseudotyped virus infected in human SUP-T1 cells assessed as inhibition of late stage viral production at 100 nM measured after 48 hrs by luciferase assay relative to control2022European journal of medicinal chemistry, Apr-05, Volume: 233Design, synthesis and biological evaluation of protease inhibitors containing morpholine cores with remarkable potency against both HIV-1 subtypes B and C.
AID1482915Antiviral activity against nelfinavir-resistant HIV1 NL4-3 harboring protease L10F/K20T/D30N/K45I/A71V/V77I mutant infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by chemiluminescent enzyme immunoassay2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID1904901Cytotoxicity against human HEK-293T cells after 24 hrs by CCK-8 assay2022European journal of medicinal chemistry, Apr-05, Volume: 233Design, synthesis and biological evaluation of protease inhibitors containing morpholine cores with remarkable potency against both HIV-1 subtypes B and C.
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.
AID690523Antiviral activity against HIV12012Journal of medicinal chemistry, Jun-28, Volume: 55, Issue:12
Dual inhibitors for aspartic proteases HIV-1 PR and renin: advancements in AIDS-hypertension-diabetes linkage via molecular dynamics, inhibition assays, and binding free energy calculations.
AID1465004Anti-viral activity against HIV-1 LAI infected in human MT2 cells assessed as reduction in p24 Gag protein production2017Bioorganic & medicinal chemistry letters, 11-01, Volume: 27, Issue:21
Design, synthesis, X-ray studies, and biological evaluation of novel macrocyclic HIV-1 protease inhibitors involving the P1'-P2' ligands.
AID396256Ratio of EC50 for HIV1 HXB2 in presence of 40 mg/ml HSA to EC50 for HIV1 HXB22007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
AID396254Ratio of EC50 for HIV1 HXB2 in presence of 40% human serum to EC50 for HIV1 HXB2 in presence of 10% fetal bovine serum2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
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.
AID1207362Inhibition of slow delayed inward rectifying potassium current (Iks) in Chinese Hamster Ovary (CHO) cells expressing hKvLQT1/hminK measured using IonWorks Quattro automated patch clamp platform
AID1767121Ratio of antiviral activity against HIV1 NL4-3 Env-deficient VSV-G pseudotyped virus infected in human SUP-T1 cells assessed as inhibition of late stage viral production when host cells were infected with compound pretreated viral particles to antiviral a2021European journal of medicinal chemistry, Aug-05, Volume: 220Design and biological evaluation of cinnamic and phenylpropionic amide derivatives as novel dual inhibitors of HIV-1 protease and reverse transcriptase.
AID1482921Antiviral activity against atazanavir-resistant HIV1 NL4-3 harboring protease L23I/E34Q/K43I/M46I/I50L/G51A/L63P/A71V/V82A/T91A mutant infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by chemiluminescent enzyme 2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID668811Antiviral activity against Human immunodeficiency virus 1 isolate M3 expressing protease L10I, K20R, M36I, G48V, I62V, A71V, V82A, I93L mutant infected in human MT4 cells2011ACS medicinal chemistry letters, Jun-09, Volume: 2, Issue:6
Disubstituted Bis-THF Moieties as New P2 Ligands in Nonpeptidal HIV-1 Protease Inhibitors.
AID459082Ratio of IC50 for multi drug-resistant HIV1 isolate of subtype G to IC50 for wild type HIV1 isolate ERS104pre2010Bioorganic & medicinal chemistry letters, Feb-01, Volume: 20, Issue:3
Synthesis and biological evaluation of novel allophenylnorstatine-based HIV-1 protease inhibitors incorporating high affinity P2-ligands.
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.
AID1778339Antiviral activity against HIV1 Env-deficient VSV-G pseudotyped virus infected in human SUP-T1 cells infected with supernatant of compound treated virus-producing 293T cells assessed as inhibition of late stage viral production at 100 nM treated for 48 hr2021European journal of medicinal chemistry, Aug-05, Volume: 220Design and evaluation of novel piperidine HIV-1 protease inhibitors with potency against DRV-resistant variants.
AID1380924Antiviral activity against wild type HIV1 NL4-3 infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by chemiluminescent enzyme immunoassay2018Journal of medicinal chemistry, 05-24, Volume: 61, Issue:10
Design and Synthesis of Highly Potent HIV-1 Protease Inhibitors Containing Tricyclic Fused Ring Systems as Novel P2 Ligands: Structure-Activity Studies, Biological and X-ray Structural Analysis.
AID673376Antiviral activity against Human immunodeficiency virus 1 HXB2 expressing protease M4 mutant infected in human MT4 cells after 5 days by MTT assay2012Bioorganic & medicinal chemistry letters, Aug-01, Volume: 22, Issue:15
Enamino-oxindole HIV protease inhibitors.
AID738143Selectivity ratio of dissociation constant K for HIV1 protease Q7K, L10F, I13V, I15V, D30V, V32I, L33F, E35D, M36I, S37N, I47V, I54L, Q58E, I62V, L63P, A71V, I84V, N88D, L89T, L90M mutant to dissociation constant K for HIV1 wild type protease2013Journal of medicinal chemistry, May-23, Volume: 56, Issue:10
Extreme multidrug resistant HIV-1 protease with 20 mutations is resistant to novel protease inhibitors with P1'-pyrrolidinone or P2-tris-tetrahydrofuran.
AID613962Ratio of IC50 for Human immunodeficiency virus 1 MDR/C to IC50 for wild-type Human immunodeficiency virus 1 ERS104pre2011Journal of medicinal chemistry, Aug-25, Volume: 54, Issue:16
Design of HIV-1 protease inhibitors with C3-substituted hexahydrocyclopentafuranyl urethanes as P2-ligands: synthesis, biological evaluation, and protein-ligand X-ray crystal structure.
AID1627024Antiviral activity against darunavir resistant HIV1 harboring protease L10I/I15V/K20R/L24I/V32I/M36I/M46L/L63P/A71T/V82A/L89M mutant at 20 passages infected in human MT4 cells by p24 assay2016Journal of medicinal chemistry, 07-28, Volume: 59, Issue:14
Probing Lipophilic Adamantyl Group as the P1-Ligand for HIV-1 Protease Inhibitors: Design, Synthesis, Protein X-ray Structural Studies, and Biological Evaluation.
AID1241688Antiviral activity against HIV1 LAI infected in human MT2 cells2015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
Design of HIV-1 Protease Inhibitors with Amino-bis-tetrahydrofuran Derivatives as P2-Ligands to Enhance Backbone-Binding Interactions: Synthesis, Biological Evaluation, and Protein-Ligand X-ray Studies.
AID1461874Inhibition of recombinant HIV-1 GFP-tagged protease expressed in HEK293 cells assessed as increase in GFP-tagged protease accumulation at 10 uM after 24 hrs by flow cytometric analysis2017Bioorganic & medicinal chemistry, 09-01, Volume: 25, Issue:17
Synthesis and biological evaluation in vitro and in mammalian cells of new heteroaryl carboxyamides as HIV-protease inhibitors.
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.
AID1482913Antiviral activity against darunavir-resistant HIV1 derived from 30 passages harboring protease L10I/I15V/K20R/L24I/V32I/M36I/M46L/L63P/K70R/V82A/I84V/L89M mutant infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID1241695Resistance index, ratio of EC50 for darunavir-resistant HIV1 P51 infected in human MT4 cells by XTT assay to EC50 for HIV1 NL4-3 infected in human MT4 cells by XTT assay2015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
Design of HIV-1 Protease Inhibitors with Amino-bis-tetrahydrofuran Derivatives as P2-Ligands to Enhance Backbone-Binding Interactions: Synthesis, Biological Evaluation, and Protein-Ligand X-ray Studies.
AID1307690Binding affinity to HIV1 protease2016Journal of medicinal chemistry, 05-12, Volume: 59, Issue:9
OpenGrowth: An Automated and Rational Algorithm for Finding New Protein Ligands.
AID519537Antiviral activity against wild-type HIV1 pNL4-3 infected in human MT4 cells assessed as reduction in viral cytopathogenicity treated 1 hr post infection measured 5 days post infection in absence of human serum by MTT assay2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
Characterization of a novel human immunodeficiency virus type 1 protease inhibitor, A-790742.
AID1409309Antiviral activity against lopinavir-resistant HIV1 infected in human MT4 cells assessed as reduction in p24 Gag protein production after 7 days by automated chemiluminescent enzyme immunoassay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Design and Synthesis of Potent HIV-1 Protease Inhibitors Containing Bicyclic Oxazolidinone Scaffold as the P2 Ligands: Structure-Activity Studies and Biological and X-ray Structural Studies.
AID726411Selectivity ratio of EC50 for Human immunodeficiency virus 1 3B clinical isolate harboring L10I/K20R/M36I/G48V/ I62V/A71V/V82A/I93L protease mutant to EC50 for wild type Human immunodeficiency virus 1 3B2013Bioorganic & medicinal chemistry letters, Jan-01, Volume: 23, Issue:1
Design and synthesis of HIV-1 protease inhibitors for a long-acting injectable drug application.
AID1891873Cytotoxicity against HEK293T cells assessed as reduction in cell viability measured after 24 hrs by CCK8 assay2022Bioorganic & medicinal chemistry, 06-15, Volume: 64A kind of HIV-1 protease inhibitors containing phenols with antiviral activity against DRV-resistant variants.
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.
AID328067Inhibition of HIV1 protease dimerization in MT2 cells at 1 uM2007The Journal of biological chemistry, Sep-28, Volume: 282, Issue:39
Potent inhibition of HIV-1 replication by novel non-peptidyl small molecule inhibitors of protease dimerization.
AID1669455Inhibition of HIV1 NL4-3 protease I84V mutant expressed in Escherichia coli TAP-106 cells using EDANS/DABCYL-labelled 10-amino acid containing protease cleavage site as substrate preincubated for 1 hr followed by substrate addition and measured for 60 min2020Journal of medicinal chemistry, 08-13, Volume: 63, Issue:15
Structural Analysis of Potent Hybrid HIV-1 Protease Inhibitors Containing Bis-tetrahydrofuran in a Pseudosymmetric Dipeptide Isostere.
AID1207550Inhibition of long-lasting type calcium current (hICa) in Chinese Hamster Ovary (CHO) cells expressing hCav1.2 measured using IonWorks Quattro automated patch clamp platform
AID1207516Inhibition of rapid delayed inward rectifying potassium current (IKr) measured using manual patch clamp assay
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.
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.
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).
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).
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.
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.
AID519788Antiviral activity against HIV 2 subtype A clinical isolate expressing 10I-40D-43I-70K-82F-84V-85L-89V-90M-91T/L-98N/K protease gene sequence from HIV2 infected patient plasma and PBMC obtained at T1 during compound treatment measured after 13 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
AID1778333Inhibition of wild type HIV1 protease using Arg-Glu (EDANS)-Ser-Gln-Asn-Tyr-Pro-Ile-Val-Gln-Lys(DABCYL)-Arg as substrate preincubated for 20 to 30 mins followed by substrate addition and measured for 10 mins by FRET assay2021European journal of medicinal chemistry, Aug-05, Volume: 220Design and evaluation of novel piperidine HIV-1 protease inhibitors with potency against DRV-resistant variants.
AID396252Ratio of EC50 for HIV1 HXB2 in presence of 20% human serum to EC50 for HIV1 HXB2 in presence of 10% fetal bovine serum2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
AID564061Antiviral activity against HIV1 expressing protease L10F/M46I/T91S mutant infected in human MT4 cells selected at 1 uM of GRL-246 by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
AID564036Antiviral activity against wild type HIV1 NL4-3 infected in human MT4 cells by MTT assay2010Antimicrobial agents and chemotherapy, Aug, Volume: 54, Issue:8
Novel protease inhibitors (PIs) containing macrocyclic components and 3(R),3a(S),6a(R)-bis-tetrahydrofuranylurethane that are potent against multi-PI-resistant HIV-1 variants in vitro.
AID726417Cytotoxicity against human MT4 cells2013Bioorganic & medicinal chemistry letters, Jan-01, Volume: 23, Issue:1
Design and synthesis of HIV-1 protease inhibitors for a long-acting injectable drug application.
AID343021Inhibition of HIV1 recombinant protease L10F/L19I/K20R/L33F/E35D/M36I/R41K/F53L/I54V/L63P/H69K/A71V/T74P/I84V/L89M/L90M/I93L mutant expressed in Escherichia coli by spectrophotometric assay2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
Inorganic polyhedral metallacarborane inhibitors of HIV protease: a new approach to overcoming antiviral resistance.
AID396238Antiviral activity against HIV1 BaL in human PBMC assessed as blockade of reverse transcriptase activity2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
AID726416Antiviral activity against Human immunodeficiency virus 1 3B clinical isolate harboring L10I/M46I/I64V/I84V/L90M/I93L protease mutant infected in human MT4 cells assessed as inhibition of viral replication2013Bioorganic & medicinal chemistry letters, Jan-01, Volume: 23, Issue:1
Design and synthesis of HIV-1 protease inhibitors for a long-acting injectable drug application.
AID343023Ratio of Ki for HIV1 recombinant protease M46I/A71V/V82T/I84V mutant to Ki for wild-type HIV1 BH10 protease2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
Inorganic polyhedral metallacarborane inhibitors of HIV protease: a new approach to overcoming antiviral resistance.
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).
AID1461873Inhibition of recombinant HIV-1 GFP-tagged protease expressed in HEK293 cells assessed as increase in GFP-tagged protease accumulation after 24 hrs by fluorescence microscopic analysis2017Bioorganic & medicinal chemistry, 09-01, Volume: 25, Issue:17
Synthesis and biological evaluation in vitro and in mammalian cells of new heteroaryl carboxyamides as HIV-protease inhibitors.
AID396249Antiviral activity against HIV1 HXB2 in human MT4 cells assessed as inhibition of viral-induced viral cytopathic effect in presence of 10% fetal bovine serum and 30% human serum by MTS assay2007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
AID260514Binding affinity to HIV1 protease D30N2006Journal of medicinal chemistry, Feb-23, Volume: 49, Issue:4
Effectiveness of nonpeptide clinical inhibitor TMC-114 on HIV-1 protease with highly drug resistant mutations D30N, I50V, and L90M.
AID297695Inhibition of HIV1 protease D30N mutant2007Journal of medicinal chemistry, Sep-06, Volume: 50, Issue:18
Potent new antiviral compound shows similar inhibition and structural interactions with drug resistant mutants and wild type HIV-1 protease.
AID770189Antiviral activity against multidrug-resistant HIV1 G harboring L10I/V11I/T12E/I15V/L19I/R41K/M46L/L63P/A71T/V82A/L90M protease-encoding region mutant infected in human PHA-PBMC cells assessed as inhibition of p24 Gag protein production.2013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Highly potent HIV-1 protease inhibitors with novel tricyclic P2 ligands: design, synthesis, and protein-ligand X-ray studies.
AID537770Antiviral activity against Human immunodeficiency virus 1 clade C isolated from HIV-AIDS patient2010Journal of medicinal chemistry, Nov-11, Volume: 53, Issue:21
Structure-based design, synthesis, and structure-activity relationship studies of HIV-1 protease inhibitors incorporating phenyloxazolidinones.
AID1207488Inhibition of rapid delayed inward rectifying potassium current (IKr) in Chinese hamster ovary (CHO) cells stable expressing hERG measured using IonWorks Barracuda automated patch clamp platform
AID711511Inhibition of HIV protease2011Journal of medicinal chemistry, Apr-14, Volume: 54, Issue:7
Macrocycles are great cycles: applications, opportunities, and challenges of synthetic macrocycles in drug discovery.
AID1725419Antiviral activity against LPV-resistant HIV1 infected in human MT4 cells incubated for 7 days by fully automated chemiluminescent enzyme immunoassay2020ACS medicinal chemistry letters, Oct-08, Volume: 11, Issue:10
Design, Synthesis, and X-ray Studies of Potent HIV-1 Protease Inhibitors with P2-Carboxamide Functionalities.
AID667088Inhibition of HIV1 protease2012Journal of medicinal chemistry, Apr-12, Volume: 55, Issue:7
Potent antiviral HIV-1 protease inhibitor GRL-02031 adapts to the structures of drug resistant mutants with its P1'-pyrrolidinone ring.
AID396258Ratio of EC50 for HIV1 HXB2 in presence of 40 mg/ml HSA and 1 mg/ml alpha1-acid glycoprotein to EC50 for HIV1 HXB22007Antimicrobial agents and chemotherapy, Sep, Volume: 51, Issue:9
In vitro antiviral activity of the novel, tyrosyl-based human immunodeficiency virus (HIV) type 1 protease inhibitor brecanavir (GW640385) in combination with other antiretrovirals and against a panel of protease inhibitor-resistant HIV.
AID1516795Selectivity ratio of EC50 for antiviral activity against multidrug-resistant HIV1 NL4-3 VSL20 infected in human TZM-bl cells to EC50 for antiviral activity against wild-type HIV1 NL4-3 infected in human TZM-bl cells2019Journal of medicinal chemistry, 09-12, Volume: 62, Issue:17
HIV-1 Protease Inhibitors Incorporating Stereochemically Defined P2' Ligands To Optimize Hydrogen Bonding in the Substrate Envelope.
AID1589132Inhibition of HIV1 protease using Ac-Thr-Ile-Nle-Nle-Gln-Arg-NH2 substrate by continuous fluorometric assay2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Potent HIV-1 protease inhibitors incorporating squaramide-derived P2 ligands: Design, synthesis, and biological evaluation.
AID1904902Selectivity index, ratio IC50 for cytotoxicity against human HEK-293T cells over IC50 for inhibition of HIV-1 protease2022European journal of medicinal chemistry, Apr-05, Volume: 233Design, synthesis and biological evaluation of protease inhibitors containing morpholine cores with remarkable potency against both HIV-1 subtypes B and C.
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.
AID1504971Inhibition of HIV1 protease expressed in Escherichia coli using Val-Ser-Gln-Asn-(beta-naphtyl)Ala-Pro-Ile-Val as substrate preincubated for 30 mins followed by substrate addition measured after 1 hr by mass spectrometric method2017ACS medicinal chemistry letters, Dec-14, Volume: 8, Issue:12
Design and Synthesis of Piperazine Sulfonamide Cores Leading to Highly Potent HIV-1 Protease Inhibitors.
AID1241689Antiviral activity against HIV1 NL4-3 infected in human MT4 cells by XTT assay2015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
Design of HIV-1 Protease Inhibitors with Amino-bis-tetrahydrofuran Derivatives as P2-Ligands to Enhance Backbone-Binding Interactions: Synthesis, Biological Evaluation, and Protein-Ligand X-ray Studies.
AID673371Antiviral activity against Human immunodeficiency virus 1 HXB2 expressing wild type protease infected in human MT4 cells after 5 days by MTT assay2012Bioorganic & medicinal chemistry letters, Aug-01, Volume: 22, Issue:15
Enamino-oxindole HIV protease inhibitors.
AID519790Antiviral activity against HIV 2 subtype B clinical isolate expressing 14Y-61N-99L protease gene sequence from HIV2 infected patient plasma and PBMC obtained before compound treatment measured after 13 months2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
In vitro phenotypic susceptibility of human immunodeficiency virus type 2 clinical isolates to protease inhibitors.
AID237954Percentage of remaining compound after 30 min incubation in dog liver microsomes was determined as metabolic stability2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Discovery and selection of TMC114, a next generation HIV-1 protease inhibitor.
AID242819Inhibitory dose against human immunodeficiency virus 1 protease2005Journal of medicinal chemistry, May-19, Volume: 48, Issue:10
Structure-based design: synthesis and biological evaluation of a series of novel cycloamide-derived HIV-1 protease inhibitors.
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.
AID690521Inhibition of HIV1 recombinant protease using Lys-Ala-Arg-Val-Nle-Nph-Glu-Ala-Nle-NH2 as substrate by spectrophotometric analysis2012Journal of medicinal chemistry, Jun-28, Volume: 55, Issue:12
Dual inhibitors for aspartic proteases HIV-1 PR and renin: advancements in AIDS-hypertension-diabetes linkage via molecular dynamics, inhibition assays, and binding free energy calculations.
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.
AID1482914Antiviral activity against darunavir-resistant HIV1 derived from 51 passages harboring protease L10I/I15V/K20R/L24I/V32I/L33F/M36I/M46L/I54M/L63P/K70Q/V82I/I84V/L89M mutant infected in human MT4 cells assessed as reduction in p24 Gag protein production af2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID260515Binding affinity to HIV1 protease I50V2006Journal of medicinal chemistry, Feb-23, Volume: 49, Issue:4
Effectiveness of nonpeptide clinical inhibitor TMC-114 on HIV-1 protease with highly drug resistant mutations D30N, I50V, and L90M.
AID236972Time to reach maximum concentration in dog after 80 mg/kg oral administration2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Discovery and selection of TMC114, a next generation HIV-1 protease inhibitor.
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.
AID673379Antiviral activity against Human immunodeficiency virus 1 HXB2 expressing protease M7 mutant infected in human MT4 cells after 5 days by MTT assay2012Bioorganic & medicinal chemistry letters, Aug-01, Volume: 22, Issue:15
Enamino-oxindole HIV protease inhibitors.
AID1295716Binding affinity to HIV1 protease I84V mutant2016Journal of medicinal chemistry, Apr-14, Volume: 59, Issue:7
Anti-HIV Drug Discovery and Development: Current Innovations and Future Trends.
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.
AID1465005Antiviral activity against wild type HIV-1 NL4-3 infected in human MT4 cells assessed as reduction in p24 Gag protein production2017Bioorganic & medicinal chemistry letters, 11-01, Volume: 27, Issue:21
Design, synthesis, X-ray studies, and biological evaluation of novel macrocyclic HIV-1 protease inhibitors involving the P1'-P2' ligands.
AID1607531Selectivity ratio of IC50 for antiviral activity against atazanavir resitant HIV1 harboring protease L23I/E34Q/K43I/M46I/I50L/G51A/L63P/A71V/V82A/T91A mutant infected in human MT4 cells to IC50 for antiviral activity against wild-type HIV1 NL4-32020Journal of medicinal chemistry, 05-14, Volume: 63, Issue:9
Structure-Based Design of Highly Potent HIV-1 Protease Inhibitors Containing New Tricyclic Ring P2-Ligands: Design, Synthesis, Biological, and X-ray Structural Studies.
AID236937Half-life in dog after 80 mg/kg oral administration2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Discovery and selection of TMC114, a next generation HIV-1 protease inhibitor.
AID343019Inhibition of HIV1 recombinant protease L10I/I15V/E35D/N37S/R41K/I62V/L63P/A71V/G73S/L90M mutant expressed in Escherichia coli by spectrophotometric assay2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
Inorganic polyhedral metallacarborane inhibitors of HIV protease: a new approach to overcoming antiviral resistance.
AID726413Selectivity ratio of EC50 for Human immunodeficiency virus 1 3B clinical isolate harboring L10I/M46I/I64V/I84V/L90M/I93L protease mutant to EC50 for wild type Human immunodeficiency virus 1 3B2013Bioorganic & medicinal chemistry letters, Jan-01, Volume: 23, Issue:1
Design and synthesis of HIV-1 protease inhibitors for a long-acting injectable drug application.
AID1767117Inhibition of wild type HIV1 protease expressed in Escherichia coli using Arg-Glu (EDANS)-Ser-Gln-Asn-Tyr-Pro-Ile-Val-Gln-Lys(DABCYL)-Arg as substrate preincubated for 20 to 30 mins followed by substrate addition and measured for 10 mins by FRET assay2021European journal of medicinal chemistry, Aug-05, Volume: 220Design and biological evaluation of cinnamic and phenylpropionic amide derivatives as novel dual inhibitors of HIV-1 protease and reverse transcriptase.
AID668812Selectivity index, ratio of EC50 for Human immunodeficiency virus 1 isolate M1 expressing protease L10I, M46I, I64V, I84V, L90M, I93L mutant to EC50 for Human immunodeficiency virus 1 3B expressing wild-type protease2011ACS medicinal chemistry letters, Jun-09, Volume: 2, Issue:6
Disubstituted Bis-THF Moieties as New P2 Ligands in Nonpeptidal HIV-1 Protease Inhibitors.
AID697738Antiviral activity against multidrug resistant HIV1 MDR1 containing protease M46I, I54V, V82A and L90M mutant infected in HEK293 cells assessed as reduction in viral replication incubated for 48 hrs by luciferase reporter based assay2012Journal of medicinal chemistry, Jul-26, Volume: 55, Issue:14
Design, synthesis, and biological and structural evaluations of novel HIV-1 protease inhibitors to combat drug resistance.
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.
AID375203Antiviral activity against multidrug-resistant HIV1 isolate G infected in PHA-stimulated PBMC assessed as inhibition of p24 gap protein2009Journal of medicinal chemistry, Jul-09, Volume: 52, Issue:13
Design of HIV-1 protease inhibitors with pyrrolidinones and oxazolidinones as novel P1'-ligands to enhance backbone-binding interactions with protease: synthesis, biological evaluation, and protein-ligand X-ray studies.
AID1227235Inhibition of wild type HIV1 protease2015Journal of medicinal chemistry, Jun-25, Volume: 58, Issue:12
Substituted Bis-THF Protease Inhibitors with Improved Potency against Highly Resistant Mature HIV-1 Protease PR20.
AID519540Antiviral activity against wild-type HIV1 pNL4-3 infected in human MT4 cells assessed as reduction in viral cytopathogenicity after 5 days post dose by MTT assay2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
Characterization of a novel human immunodeficiency virus type 1 protease inhibitor, A-790742.
AID697744Inhibition of HIV1 protease I50V, A71V mutant by FRET assay2012Journal of medicinal chemistry, Jul-26, Volume: 55, Issue:14
Design, synthesis, and biological and structural evaluations of novel HIV-1 protease inhibitors to combat drug resistance.
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).
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.
AID446203Selectivity ratio of IC50 for multidrug-resistant HIV1 isolate TM to IC50 for wild type HIV1 isolate ERS104pre2009Journal of medicinal chemistry, Dec-10, Volume: 52, Issue:23
Design, synthesis, protein-ligand X-ray structure, and biological evaluation of a series of novel macrocyclic human immunodeficiency virus-1 protease inhibitors to combat drug resistance.
AID1482923Ratio of IC50 for lopinavir-resistant HIV1 NL4-3 harboring protease L10F/V32I/M46I/I47A/A71V/I84V mutant infected in human MT4 cells to IC50 for wild type HIV1 NL4.3 infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID697918Inhibition of wild type HIV1 protease by FRET assay2012Journal of medicinal chemistry, Jul-26, Volume: 55, Issue:14
Design, synthesis, and biological and structural evaluations of novel HIV-1 protease inhibitors to combat drug resistance.
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.
AID537766Inhibition of multidrug resistant HIV1 protease L101I, L36P, A71V, G73S, I84V, L90M mutant by FRET2010Journal of medicinal chemistry, Nov-11, Volume: 53, Issue:21
Structure-based design, synthesis, and structure-activity relationship studies of HIV-1 protease inhibitors incorporating phenyloxazolidinones.
AID1778340Antiviral activity against HIV1 NL4-3 infected in human MT4 cells assessed as reduction in viral replication by measuring reduction in p24 Gag production by chemiluminescent-EIA2021European journal of medicinal chemistry, Aug-05, Volume: 220Design and evaluation of novel piperidine HIV-1 protease inhibitors with potency against DRV-resistant variants.
AID519538Antiviral activity against wild-type HIV1 pNL4-3 infected in human MT4 cells assessed as reduction in viral cytopathogenicity treated 1 hr post infection measured 5 days post infection in presence of 50% human serum by MTT assay2008Antimicrobial agents and chemotherapy, Apr, Volume: 52, Issue:4
Characterization of a novel human immunodeficiency virus type 1 protease inhibitor, A-790742.
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).
AID613961Ratio of IC50 for Human immunodeficiency virus 1 MDR/B to IC50 for wild-type Human immunodeficiency virus 1 ERS104pre2011Journal of medicinal chemistry, Aug-25, Volume: 54, Issue:16
Design of HIV-1 protease inhibitors with C3-substituted hexahydrocyclopentafuranyl urethanes as P2-ligands: synthesis, biological evaluation, and protein-ligand X-ray crystal structure.
AID446200Selectivity ratio of IC50 for multidrug-resistant HIV1 isolate B to IC50 for wild type HIV1 isolate ERS104pre2009Journal of medicinal chemistry, Dec-10, Volume: 52, Issue:23
Design, synthesis, protein-ligand X-ray structure, and biological evaluation of a series of novel macrocyclic human immunodeficiency virus-1 protease inhibitors to combat drug resistance.
AID649704Ratio of EC50 for HIV1 MDR/TM harboring protease L10I, K14R, R41K, M46L, I54V, L63P, A71V, V82A, L90M, I93L mutant to EC50 for HIV1 ERS104 harboring wild type protease2012Bioorganic & medicinal chemistry letters, Mar-15, Volume: 22, Issue:6
Substituent effects on P2-cyclopentyltetrahydrofuranyl urethanes: design, synthesis, and X-ray studies of potent HIV-1 protease inhibitors.
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.
AID1904907Antiviral activity against HIV1 subtype C isolate Indie2022European journal of medicinal chemistry, Apr-05, Volume: 233Design, synthesis and biological evaluation of protease inhibitors containing morpholine cores with remarkable potency against both HIV-1 subtypes B and C.
AID1482910Antiviral activity against HIV1 Lai infected in human MT2 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Design and Development of Highly Potent HIV-1 Protease Inhibitors with a Crown-Like Oxotricyclic Core as the P2-Ligand To Combat Multidrug-Resistant HIV Variants.
AID446202Selectivity ratio of IC50 for multidrug-resistant HIV1 isolate G to IC50 for wild type HIV1 isolate ERS104pre2009Journal of medicinal chemistry, Dec-10, Volume: 52, Issue:23
Design, synthesis, protein-ligand X-ray structure, and biological evaluation of a series of novel macrocyclic human immunodeficiency virus-1 protease inhibitors to combat drug resistance.
AID504749qHTS profiling for inhibitors of Plasmodium falciparum proliferation2011Science (New York, N.Y.), Aug-05, Volume: 333, Issue:6043
Chemical genomic profiling for antimalarial therapies, response signatures, and molecular targets.
AID1347127qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
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.
AID1745845Primary qHTS for Inhibitors of ATXN expression
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.
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.
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.
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.
AID1347126qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
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.
AID1347113qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347121qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
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.
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.
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.
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.
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.
AID1347119qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
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.
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.
AID1347114qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
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.
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.
AID1347122qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347111qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347115qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347129qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
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.
AID1347116qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347112qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347125qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
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.
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.
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.
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.
AID1347110qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for A673 cells)2018Oncotarget, 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.
AID1347109qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347128qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347124qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347117qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
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.
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.
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.
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.
AID1347123qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347118qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
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.
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.
AID977610Experimentally measured binding affinity data (Ki) for protein-ligand complexes derived from PDB2012Protein science : a publication of the Protein Society, Mar, Volume: 21, Issue:3
Critical differences in HIV-1 and HIV-2 protease specificity for clinical inhibitors.
AID1794808Fluorescence-based screening to identify small molecule inhibitors of Plasmodium falciparum apicoplast DNA polymerase (Pf-apPOL).2014Journal of biomolecular screening, Jul, Volume: 19, Issue:6
A High-Throughput Assay to Identify Inhibitors of the Apicoplast DNA Polymerase from Plasmodium falciparum.
AID1794808Fluorescence-based screening to identify small molecule inhibitors of Plasmodium falciparum apicoplast DNA polymerase (Pf-apPOL).
AID1796146Protease Inhibition Assay from Article 10.1021/jm050943c: \\Effectiveness of nonpeptide clinical inhibitor TMC-114 on HIV-1 protease with highly drug resistant mutations D30N, I50V, and L90M.\\2006Journal of medicinal chemistry, Feb-23, Volume: 49, Issue:4
Effectiveness of nonpeptide clinical inhibitor TMC-114 on HIV-1 protease with highly drug resistant mutations D30N, I50V, and L90M.
AID1800111Drug Susceptibility Assay from Article 10.1021/cb400468c: \\Testing the substrate-envelope hypothesis with designed pairs of compounds.\\2013ACS chemical biology, Nov-15, Volume: 8, Issue:11
Testing the substrate-envelope hypothesis with designed pairs of compounds.
AID1797105Protease Inhibition Assay from Article 10.1021/jm060561m: \\Structure-Based Design of Novel HIV-1 Protease Inhibitors To Combat Drug Resistance.\\2006Journal of medicinal chemistry, Aug-24, Volume: 49, Issue:17
Structure-based design of novel HIV-1 protease inhibitors to combat drug resistance.
AID1805801Various Assay from Article 10.1021/acs.jmedchem.1c00409: \\Perspectives on SARS-CoV-2 Main Protease Inhibitors.\\2021Journal of medicinal chemistry, 12-09, Volume: 64, Issue:23
Perspectives on SARS-CoV-2 Main Protease Inhibitors.
AID1800303HIV-1 protease inhibition assay from Article 10.1016/j.chembiol.2013.07.014: \\Substrate envelope-designed potent HIV-1 protease inhibitors to avoid drug resistance.\\2013Chemistry & biology, Sep-19, Volume: 20, Issue:9
Substrate envelope-designed potent HIV-1 protease inhibitors to avoid drug resistance.
AID1799362Enzyme Inhibition Assay (Ki) and Antiviral Activity Assay (EC50/IC50) from Article 10.1021/jm900695w: \\Design, synthesis, protein-ligand X-ray structure, and biological evaluation of a series of novel macrocyclic human immunodeficiency virus-1 protease in2009Journal of medicinal chemistry, Dec-10, Volume: 52, Issue:23
Design, synthesis, protein-ligand X-ray structure, and biological evaluation of a series of novel macrocyclic human immunodeficiency virus-1 protease inhibitors to combat drug resistance.
AID977610Experimentally measured binding affinity data (Ki) for protein-ligand complexes derived from PDB2004Journal of molecular biology, Apr-23, Volume: 338, Issue:2
High resolution crystal structures of HIV-1 protease with a potent non-peptide inhibitor (UIC-94017) active against multi-drug-resistant clinical strains.
AID1811Experimentally measured binding affinity data derived from PDB2004Journal of molecular biology, Apr-23, Volume: 338, Issue:2
High resolution crystal structures of HIV-1 protease with a potent non-peptide inhibitor (UIC-94017) active against multi-drug-resistant clinical strains.
AID1811Experimentally measured binding affinity data derived from PDB2006Journal of molecular biology, Oct-13, Volume: 363, Issue:1
Ultra-high resolution crystal structure of HIV-1 protease mutant reveals two binding sites for clinical inhibitor TMC114.
AID977610Experimentally measured binding affinity data (Ki) for protein-ligand complexes derived from PDB2006Journal of molecular biology, Oct-13, Volume: 363, Issue:1
Ultra-high resolution crystal structure of HIV-1 protease mutant reveals two binding sites for clinical inhibitor TMC114.
AID977611Experimentally measured binding affinity data (Kd) for protein-ligand complexes derived from PDB2014The FEBS journal, Apr, Volume: 281, Issue:7
Thermodynamic and structural analysis of HIV protease resistance to darunavir - analysis of heavily mutated patient-derived HIV-1 proteases.
AID977610Experimentally measured binding affinity data (Ki) for protein-ligand complexes derived from PDB2008Journal of molecular biology, Aug-01, Volume: 381, Issue:1
Effect of flap mutations on structure of HIV-1 protease and inhibition by saquinavir and darunavir.
AID977611Experimentally measured binding affinity data (Kd) for protein-ligand complexes derived from PDB2012Biochemistry, Feb-07, Volume: 51, Issue:5
Terminal interface conformations modulate dimer stability prior to amino terminal autoprocessing of HIV-1 protease.
AID977610Experimentally measured binding affinity data (Ki) for protein-ligand complexes derived from PDB2006Journal of medicinal chemistry, Feb-23, Volume: 49, Issue:4
Effectiveness of nonpeptide clinical inhibitor TMC-114 on HIV-1 protease with highly drug resistant mutations D30N, I50V, and L90M.
AID1811Experimentally measured binding affinity data derived from PDB2006Journal of medicinal chemistry, Feb-23, Volume: 49, Issue:4
Effectiveness of nonpeptide clinical inhibitor TMC-114 on HIV-1 protease with highly drug resistant mutations D30N, I50V, and L90M.
AID977611Experimentally measured binding affinity data (Kd) for protein-ligand complexes derived from PDB2008The Journal of biological chemistry, May-09, Volume: 283, Issue:19
Effect of the active site D25N mutation on the structure, stability, and ligand binding of the mature HIV-1 protease.
AID977611Experimentally measured binding affinity data (Kd) for protein-ligand complexes derived from PDB2011Biochemistry, May-31, Volume: 50, Issue:21
The L76V drug resistance mutation decreases the dimer stability and rate of autoprocessing of HIV-1 protease by reducing internal hydrophobic contacts.
AID977611Experimentally measured binding affinity data (Kd) for protein-ligand complexes derived from PDB2010Journal of virology, Oct, Volume: 84, Issue:19
The effect of clade-specific sequence polymorphisms on HIV-1 protease activity and inhibitor resistance pathways.
AID977610Experimentally measured binding affinity data (Ki) for protein-ligand complexes derived from PDB2009Journal of virology, Sep, Volume: 83, Issue:17
Molecular characterization of clinical isolates of human immunodeficiency virus resistant to the protease inhibitor darunavir.
AID977610Experimentally measured binding affinity data (Ki) for protein-ligand complexes derived from PDB2012Antimicrobial agents and chemotherapy, Aug, Volume: 56, Issue:8
Mutations in HIV-1 gag and pol compensate for the loss of viral fitness caused by a highly mutated protease.
AID977611Experimentally measured binding affinity data (Kd) for protein-ligand complexes derived from PDB2012ACS chemical biology, Sep-21, Volume: 7, Issue:9
Extreme entropy-enthalpy compensation in a drug-resistant variant of HIV-1 protease.
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.
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.
AID977608Experimentally measured binding affinity data (IC50) for protein-ligand complexes derived from PDB2011Biochemical and biophysical research communications, Sep-09, Volume: 412, Issue:4
The higher barrier of darunavir and tipranavir resistance for HIV-1 protease.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (1,074)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's1 (0.09)18.2507
2000's216 (20.11)29.6817
2010's656 (61.08)24.3611
2020's201 (18.72)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 67.19

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 Index67.19 (24.57)
Research Supply Index7.22 (2.92)
Research Growth Index6.91 (4.65)
Search Engine Demand Index113.74 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (67.19)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials224 (19.51%)5.53%
Reviews116 (10.10%)6.00%
Case Studies86 (7.49%)4.05%
Observational34 (2.96%)0.25%
Other688 (59.93%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (185)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
A Single-dose, Open-label, Randomized, Crossover Pivotal Bioequivalence Study in Healthy Participants to Assess the Bioequivalence of Darunavir 675 mg in the Presence of 150 mg Cobicistat When Administered as a Fixed Dose Combination (Darunavir/Cobicistat [NCT04718805]Phase 122 participants (Actual)Interventional2021-01-26Completed
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
[NCT01274780]Phase 4180 participants (Actual)Interventional2011-05-31Completed
Phase III Multicenter Randomized Trial Evaluating in Patients at the Time of the Primary HIV-1 Infection, the Impact on the Viral Reservoir of a Combination Including Tenofovir/Emtricitabine and Dolutegravir or Tenofovir/Emtricitabine and Darunavir/Cobici [NCT02987530]Phase 3101 participants (Actual)Interventional2017-04-11Completed
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.)
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
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
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)
Switching From Lopinavir/Ritonavir Plus Tenofovir and Emtricitabine (or Lamivudine) to Darunavir (Prezista) and Raltegravir to Evaluate Renal Function [NCT01294761]59 participants (Actual)Interventional2011-02-28Completed
A Phase 4, Single-arm, Open-label Study to Evaluate the Efficacy and Safety of Darunavir/Cobicistat/Emtricitabine/Tenofovir Alafenamide (D/C/F/TAF) Once Daily Fixed-dose Combination (FDC) Regimen in Antiretroviral Treatment-experienced Human Immunodeficie [NCT04388904]Phase 475 participants (Actual)Interventional2021-09-01Active, not 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
Dolutegravir, Darunavir/Ritonavir and Optimized NRTI Recycling as a Third-line Antiretroviral Regimen in Cambodia [NCT03602690]Phase 254 participants (Anticipated)Interventional2018-10-04Recruiting
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
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
Body Composition Sub-study of the D2EFT Trial [NCT03675815]Phase 4155 participants (Actual)Interventional2019-12-05Active, not recruiting
A Phase IV 48 Week, Open Label, Pilot Study of Darunavir Boosted by Cobicistat in Combination With Rilpivirine to Treat HIV+ Naïve Subjects (PREZENT) [NCT02404233]Phase 430 participants (Anticipated)Interventional2015-03-31Not yet recruiting
Multicenter, Open, Pilot Clinical Trial Aimed to Compare the Efficacy of RAL1200 QD vs DRV-cb 800-150 QD Both in Combination With TAF/FTC in Patients With HIV Infection and CD4 Count Under 200 Cells/microL [NCT03842488]Phase 475 participants (Anticipated)Interventional2019-04-30Not yet recruiting
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
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
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 Phase I, Open-label, Randomized, Crossover Trial in Healthy Subjects Receiving DRV Combined With RTV Low Dose to Compare the Oral Bioavailability of DRV Suspension to That of DRV 300 mg Tablet Under Fasted and Fed Conditions, and to Assess Multiple Dose [NCT00752310]Phase 123 participants (Actual)Interventional2008-04-30Completed
Intensive Pharmacokinetic Studies of Antiretroviral Drug Combinations in Children [NCT00260078]Phase 1/Phase 275 participants (Actual)Interventional2006-02-28Completed
Optimization of Darunavir Therapy Through Population Pharmacokinetic Modeling, Simulations and Dosage Guidelines [NCT03101644]Phase 4127 participants (Actual)Interventional2017-03-23Completed
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
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 Phase II Randomized, Controlled, Partially Blinded Trial to Investigate Dose-response of TMC114/RTV in 3-class-experienced HIV-1 Infected Subjects, Followed by an Open-label Period on the Recommended Dose of TMC114/RTV. [NCT00650832]Phase 2334 participants (Actual)Interventional2003-10-31Completed
Open-label Safety Study of TMC114 in Combination With Low Dose RTV and Other ARVs in Highly Experienced HIV-1 Infected Patients With Limited or no Treatment Options. [NCT00115050]Phase 3260 participants (Actual)Interventional2005-06-30Completed
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)
Phase I, Double-blind, Randomized, Placebo-controlled Trial to Examine the Safety, Tolerability and Plasma Pharmacokinetics of Increasing Single Oral Doses of TMC558445 With and Without Food, and Increasing Repeated Oral Doses in Combination With a Single [NCT00838760]Phase 154 participants (Actual)Interventional2009-02-28Completed
Drug-drug Interactions Between Antiretroviral Drugs and Cardiovascular Drugs in Elderly Patients From the Swiss HIV Cohort Study [NCT03515772]21 participants (Actual)Observational2018-04-23Completed
Pharmacokinetic Properties of Antiretroviral and Related Drugs During Pregnancy and Postpartum [NCT00042289]1,578 participants (Actual)Observational2003-06-09Completed
Switch to Darunavir/r + Maraviroc QD in Patients With R5 Tropism by Viral DNA Genotyping With Suppressed Viremia (GUSTA): a Multicenter, Open-label, Randomized Controlled Trial [NCT01367210]Phase 4165 participants (Actual)Interventional2011-06-30Terminated(stopped due to Because a higher rate of virological failures in study versus control arm.)
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
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
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 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 Randomised, Open Label Switch Study Comparing Darunavir/Ritonavir 400mg/100mg Daily With Lopinavir/Ritonavir 800mg/200mg Daily, in HIV-positive Participants [NCT02671383]Phase 3300 participants (Actual)Interventional2016-06-30Completed
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
Efficacy and Safety of Darunavir and Cobicistat for Treatment of COVID-19 [NCT04252274]Phase 330 participants (Anticipated)Interventional2020-01-30Recruiting
Interaction of Buprenorphine With HIV Medications and Tuberculosis Medications [NCT00877591]Phase 163 participants (Actual)Interventional2008-04-30Completed
Elvitegravir/Cobicistat/Tenofovir DF/Emtricitabine With Darunavir in Treatment-experienced Patients: Quality Control Monitoring of a Treatment Simplification Strategy [NCT02199613]Phase 410 participants (Actual)Interventional2014-10-31Completed
Evaluation of the Capacity of a Weekly Strategy of 4 Consecutive Days on Treatment Followed by 3 Days Off Treatment, in HIV-1 Infected Patients With Undetectable Viral Load for at Least 12 Months, to Maintain a Virological Success With This Intermittent M [NCT02157311]Phase 3100 participants (Actual)Interventional2014-07-31Completed
Phase II Trial Assessing the Efficacy of a Reduced Dose Strategy of Darunavir to 400 mg/d in HIV-1 Infected Patients Virologically Suppressed Under a Once Daily Regimen Including Darunavir 800 mg/d and Two Nucleoside Reverse Transcriptase Inhibitors (NRTI [NCT02384967]Phase 2100 participants (Actual)Interventional2015-03-31Completed
An Open Label Trial of TMC114/Rtv in HIV-1 Infected Subjects Who Were Randomized in the Trials TMC114-C201, TMC114-C207 or in Sponsor Selected Phase I Trials [NCT02187107]Phase 252 participants (Actual)Interventional2005-01-31Completed
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
A Randomized Study to Compare the Efficacy of Vorinostat/Hydroxychloroquine/Maraviroc (VHM) in Controlling HIV After Treatment Interruption in Subjects Who Initiated ART During Acute HIV Infection [NCT02475915]Phase 1/Phase 215 participants (Actual)Interventional2015-01-31Completed
A Single-dose, Open-label, Randomized, Crossover Study to Assess the Impact of Food on the Pharmacokinetics of Darunavir, Cobicistat, Emtricitabine, and Tenofovir Alafenamide Administered as a Fixed-dose Combination Tablet, and the Relative Bioavailabilit [NCT02475135]Phase 172 participants (Actual)Interventional2015-06-01Completed
Effect of Darunavir/Cobicistat/Emtricitabine/Tenofovir Alafenamide and Bictegravir/Emtricitabine/Tenofovir Alafenamide on the Circulatory microRNA Profile in Treatment naïve HIV Patients, and Its Correlation With Change in Body Weight [NCT05463783]Phase 430 participants (Anticipated)Interventional2023-03-14Recruiting
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
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 Randomised, Controlled, Open-label Trial to Compare the Efficacy, Safety and Tolerability of a Treatment Simplification by Darunavir/Ritonavir (DRV/r) 800/100 mg O.D. vs a Triple Combination Therapy With DRV/r in HIV-1 Infected Patients With Undetectabl [NCT00458302]Phase 3256 participants (Actual)Interventional2007-06-30Completed
Phase I, Open-label, Randomized, 3-way Crossover Trial to Assess the Pharmacokinetics of Darunavir (DRV) Given Once-daily With Different Doses of Ritonavir in Healthy Subjects. [NCT00744887]Phase 121 participants (Actual)Interventional2008-08-31Completed
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
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
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
Drug Interaction Study Between Inhaled Beclomethasone and Protease Inhibitors in Healthy Volunteers [NCT00936793]Phase 153 participants (Actual)Interventional2009-07-06Completed
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
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
Impact of Once-Weekly Rifapentine and Isoniazid on the Steady State Pharmacokinetics of Dolutegravir and Darunavir Boosted With Cobicistat in Healthy Volunteers [NCT02771249]Phase 137 participants (Actual)Interventional2016-06-03Completed
A Phase IIa Open-label, Randomized Trial to Determine the Antiviral Activity in 60 HIV Positive Subjects With Multiple PI Resistant Strains, Receiving Either Control Treatment or a Daily Dose of 800, 1600, 2400 or 3600 mg TMC114 (Darunavir) for 13 Days Fo [NCT00964327]Phase 242 participants (Actual)Interventional2001-08-31Completed
Implication for Strategies of Long Term Control of Viral Replication in Patient With Primary HIV Infection (PHI) Treated With Multitarget Antiviral Therapy (MT-ART) [NCT04225325]Phase 4112 participants (Anticipated)Interventional2018-05-07Recruiting
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
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
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
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 Double-Blind (3rd Party Open), Placebo-Controlled, Crossover, Dose Escalating Study To Evaluate The Safety, Tolerability And Pharmacokinetics Of Single Oral Doses Of PF-03716539, To Assess The Potential Of PF-03716539 To Inhibit CYP3A4 (In Vivo) And To [NCT00783484]Phase 137 participants (Actual)Interventional2008-10-31Completed
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
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
Steady-state Pharmacokinetics of Atazanavir/Cobicistat and Darunavir/Cobicistat Once Daily Over 72 Hours in Healthy Volunteers [NCT02589158]Phase 116 participants (Actual)Interventional2015-11-30Completed
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
A Single-Dose, Open-Label, Randomized, Crossover Study to Assess the Bioequivalence of Darunavir 800 mg, Emtricitabine 200 mg, and Tenofovir Alafenamide 10 mg, in the Presence of Cobicistat 150 mg, Administered as Either a Fixed-Dose Combination Tablet or [NCT02578550]Phase 1126 participants (Actual)Interventional2015-11-30Completed
Non-comparative, Opened Study, Evaluating in HIV-1 Infected Patients With Undetectable Viral Load, Treated by an Antiretroviral Combination Including a Protease Inhibitor Boosted With Ritonavir and Administered by Oral Route Twice a Day, the Substitutabil [NCT00849160]Phase 3100 participants (Actual)Interventional2009-05-31Completed
Clinical Trial to Evaluate Drug-drug Interactions Between Darunavir/Cobicistat and Etravirine in Hiv- Infected Patients [NCT02818348]Phase 130 participants (Actual)Interventional2016-06-30Completed
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
"Crossover, Open-label, Randomized, Single-dose, Bioequivalence Study of GP30101 Film-coated Tablets (LLC GEROPHARM, Russia) 800 mg Versus Prezista® (Jonson&Jonson, Russia) Film-coated Tablets 800 mg in Healthy Volunteers Under Fed Conditions" [NCT04268472]44 participants (Actual)Interventional2019-06-06Completed
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
The Optimized Treatment That Includes or Omits NRTIs Trial: A Randomized Strategy Study for HIV-1-Infected Treatment-Experienced Subjects Using the cPSS to Select an Effective Regimen [NCT00537394]Phase 3517 participants (Actual)Interventional2008-01-31Completed
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
The ARDENT Study: Atazanavir, Raltegravir, or Darunavir With Emtricitabine/Tenofovir for Naive Treatment [NCT00811954]Phase 31,814 participants (Actual)Interventional2009-05-31Completed
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
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
Multicenter, International, Prospective, Phase III, Randomized, Superiority Trial Comparing Two Maintenance Strategies With Mono or Bi-therapy of Protease Inhibitors With or Without Lamivudine in Virologically Suppressed HIV Patients on Second Line Antire [NCT01905059]Phase 3265 participants (Actual)Interventional2014-02-28Completed
"Systematic Adherence Intervention Phase Before Switching to 3rd-line ART in Patients With 2nd-line ART Virologic Failure in Sub-Saharan Africa : a Phase 2b Non-randomized Study." [NCT02025868]Phase 2201 participants (Actual)Interventional2013-03-31Active, not recruiting
Dual Therapy With Raltegravir 400 mg BID and Darunavir/Ritonavir 800/100 mg QD in HIV Infected Patients Failing to Nucleoside Reverse Transcriptase Inhibitors Based Regimens [NCT01258374]15 participants (Actual)Observational2010-05-31Completed
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
Optimisation of Primary HIV1 Infection Treatment (ANRS 147 OPTIPRIM) [NCT01033760]Phase 390 participants (Actual)Interventional2010-04-30Completed
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 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
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
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
A Phase IV, Prospective, Multicenter , Randomized Open Label, 48 Weeks Study to Evaluate the Antiretroviral Efficacy and Safety of Atazanavir or Darunavir,Each in Combination With a Fixed Dose of Tenofovir Emtricitabine in HIV-1-infected Treatment-naïve S [NCT01928407]Phase 4120 participants (Actual)Interventional2011-02-23Completed
Early Access of TMC114 in Combination With Low-dose Ritonavir (TMC/r)) and Other Antiretrovirals (ARVs) in Highly Treatment Experienced HIV-1 Infected Patients With Limited to no Treatment Options. [NCT00245739]0 participants Expanded AccessApproved for marketing
A Randomized, Controlled, Open-label Trial to Compare the Efficacy, Safety and Tolerability of TMC114/RTV Versus LPV/RTV in Treatment-Experienced HIV-1 Infected Patients [NCT00110877]Phase 3604 participants (Actual)Interventional2005-04-30Completed
Open-Label, Single-Sequence, Two-Cohort Study to Evaluate the Effect of Darunavir/Cobicistat and Cobicistat on BMS-626529 in Healthy Subjects [NCT02277600]Phase 132 participants (Actual)Interventional2014-11-05Completed
A Phase 1 Pharmacokinetic Study to Assess the Steady State Pharmacokinetic Profile and Short Term Safety of Maraviroc Dosed With Darunavir/Ritonavir All Once Daily, With and Without Nucleoside Analogues, in HIV-1 Infected Subjects [NCT01348763]Phase 113 participants (Actual)Interventional2011-10-31Completed
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
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
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
Multicenter, National, Prospective, Open Label, Randomized, Pilot, Proof-of-concept Study on the Use of Rilpivirine Plus Darunavir/Cobicistat as Substitutive Agents in Virologic Suppressed Patients [NCT04064632]Phase 41,609 participants (Actual)Interventional2017-02-01Active, not recruiting
Evaluation of Safety and Efficacy of Raltegravir/Darunavir Combination in Antiretroviral-Naive Patients [NCT00677300]Phase 485 participants (Actual)Interventional2009-01-31Completed
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 Phase II Randomized, Controlled, Partially Blinded Trial to Investigate Dose Response of TMC114/RTV in 3-class-experienced HIV-1 Infected Patients, Followed by an Open-label Period on the Recommended Dose of TMC114/RTV. [NCT00071097]Phase 2330 participants (Actual)Interventional2003-10-31Completed
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
Phase I, Open-label, 3-way Crossover Trial in Healthy Male Volunteers to Evaluate the Pharmacokinetics of TMC114 and TMC41629 After a Single Oral Dose of 2 Controlled-release Coformulations as Compared to an Immediate-release Coformulation of TMC114/TMC41 [NCT00854204]Phase 112 participants (Actual)Interventional2008-11-30Completed
A Single-Dose, Open-Label, Randomized, Replicate Crossover Pivotal Bioequivalence Study in Healthy Subjects to Assess the Bioequivalence of Darunavir 675 mg, Emtricitabine 200 mg, and Tenofovir Alafenamide 10 mg in the Presence of Cobicistat 150 mg When A [NCT04236453]Phase 116 participants (Actual)Interventional2020-01-23Terminated(stopped due to COVID-19 pandemic impacted the BE assessment of the trial. The clinical team decided to terminate the trial in order to start a new pivotal BE trial)
A Single-dose, Open-label, Randomized, Crossover Pivotal Bioequivalence Study in Healthy Participants to Assess the Bioequivalence of Darunavir 600 mg in the Presence of Cobicistat 90 mg When Administered as a Fixed Dose Combination Tablet (Darunavir/Cobi [NCT05378906]Phase 132 participants (Actual)Interventional2022-06-07Completed
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.)
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
Phase IV, Open Label, Randomized, Clinical Trial to Evaluate the Reversibility of Abacavir/Lamivudine/Dolutegravir CNS-Related Neurotoxicity After Switching to Tenofovir Alafenamide/Emtricitabine/Darunavir/Cobicistat [NCT03685500]Phase 478 participants (Actual)Interventional2018-12-04Completed
Monotherapy in Africa: Evaluation of New Therapy [NCT02155101]Phase 3120 participants (Actual)Interventional2014-05-31Completed
A Study to Assess the Acceptability of the Darunavir/Cobicistat (DRV/COBI) Fixed-dose Combination (FDC) Tablet in Human Immunodeficiency Virus (HIV)-1 Infected Children Aged >=3 Years and Weighing >=15 kg to <25 kg [NCT05197075]Phase 115 participants (Actual)Interventional2022-08-03Completed
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
A Randomized, Controlled Trial to Evaluate the Efficacy of Substituting Darunavir/Ritonavir (DRV/r) for Dual-boosted Protease Inhibitors in Individuals With Virologic Suppression for at Least 12 Weeks [NCT00543101]Phase 424 participants (Actual)Interventional2007-10-31Completed
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
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
Contribution of the Integrase Inhibitor Dolutegravir to Obesity and Cardiovascular Disease in Persons Living With HIV [NCT04340388]Phase 410 participants (Actual)Interventional2020-09-17Completed
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
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
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
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
An Open Label Trial of TMC114/RTV in HIV-1 Infected, Treatment-experienced Subjects. [NCT00081588]Phase 2555 participants (Actual)Interventional2003-11-30Completed
A Phase 2, Randomized, Double-Blinded Study of the Safety and Efficacy of Darunavir/Cobicistat/Emtricitabine/GS-7340 Single Tablet Regimen Versus Cobicistat-boosted Darunavir Plus Emtricitabine/Tenofovir Disoproxil Fumarate Fixed Dose Combination in HIV-1 [NCT01565850]Phase 2153 participants (Actual)Interventional2012-04-30Completed
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
A Pharmacokinetic Evaluation of Etonogestrel Implant in HIV-infected Women on Darunavir Versus Ripilvirine-based Antiretroviral Therapy [NCT03589040]Phase 260 participants (Anticipated)Interventional2018-09-25Recruiting
A Single-dose, Open-label, Randomized, Crossover Study to Assess the Relative Bioavailability of the Fixed-dose Combination Tablet Darunavir/Cobicistat/Emtricitabine/Tenofovir Alafenamide (D/C/F/TAF) Administered Orally as a Whole Tablet, as a Split Table [NCT02984852]Phase 130 participants (Actual)Interventional2016-12-31Completed
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.)
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
Pharmacokinetic Properties of Antiretroviral and Anti-Tuberculosis Drugs During Pregnancy and Postpartum [NCT04518228]325 participants (Anticipated)Observational2021-06-08Recruiting
Phase III Randomized, Controlled, Open-label Trial to Investigate the Antiviral Activity, Tolerability and Safety of TMC114/r in Treatment- Naive HIV-1 Infected Patients. [NCT00258557]Phase 3692 participants (Actual)Interventional2005-09-30Completed
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 Single-Dose, Open-Label, 3-Panel, Randomized, Pivotal Crossover Study to Assess the Bioequivalence of Darunavir When Co-Administrated With Cobicistat as Either a Fixed Dose Combination Tablet (G006) or as Single Agents Under Fed and Fasted Conditions in [NCT01619527]Phase 1133 participants (Actual)Interventional2012-04-30Completed
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
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 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
Regulatory Post Marketing Surveillance of Prezista 400mg Tablet [NCT01741831]225 participants (Actual)Observational2012-07-31Completed
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
Strategic Study of Dual-therapy With Darunavir/Ritonavir and Rilpivirine QD Versus Triple-therapy in Patients With Suppressed Viral Load: Virological Efficacy and Evaluation of Non-HIV Related Morbidity. [NCT01792570]Phase 337 participants (Actual)Interventional2014-09-30Completed
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
Body Compartment Pharmacokinetics of Anti-retroviral Agents That May be Used for Future HIV Post-exposure Prophylaxis Regimens. [NCT03472963]Phase 135 participants (Actual)Interventional2018-04-27Completed
A Phase 1, Open-label, Parallel-group Study To Assess The Effect Of Cyp3a5 Genotype On The Pharmacokinetics Of Maraviroc And Cyp3a5-derived Metabolites With And Without Darunavir/Cobicistat In African-american And Caucasian Healthy Volunteers [NCT02625207]Phase 147 participants (Actual)Interventional2015-11-06Completed
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 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
An Open-label Study to Evaluate the Pharmacokinetics (PK) of Darunavir (DRV) and Cobicistat (COBI) After a Single-oral Administration of Darunavir/Cobicistat Fixed-Dose Combination Tablet in Healthy Japanese Adult Subjects [NCT03123848]Phase 48 participants (Actual)Interventional2017-04-14Completed
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
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
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
Efficacy and Safety of a Simplification Strategy Based on Dolutegravir and Darunavir / Cobicistat vs Optimized Treatment in Suppressed HIV-1-infected Patients Carrying Archived Multidrug Resistance Mutations. [NCT03683524]Phase 496 participants (Actual)Interventional2018-11-19Completed
Impact of Steady State Cobicistat and Darunavir/Cobicistat on the Pharmacokinetics and Pharmacodynamics of Oral Anticoagulants (Rivaroxaban, Apixaban) in Healthy Volunteers [NCT03864406]Phase 112 participants (Actual)Interventional2019-06-04Completed
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
A Phase 3, Randomized, Active-controlled, Double-blind Study to Evaluate Efficacy and Safety of Darunavir/Cobicistat/Emtricitabine/Tenofovir Alafenamide (D/C/F/TAF) Once Daily Fixed Dose Combination Regimen Versus a Regimen Consisting of Darunavir/Cobicis [NCT02431247]Phase 3725 participants (Actual)Interventional2015-07-06Completed
Management Using the Latest Technologies in Resource-limited Settings to Optimize Combination Therapy After Viral Failure (MULTI-OCTAVE) [NCT01641367]Phase 4545 participants (Actual)Interventional2013-02-22Completed
A Single-Dose, Open-Label, Randomized, Replicate Crossover Pivotal Bioequivalence Study in Healthy Adult Participants to Assess the Bioequivalence of Darunavir 675 mg, Emtricitabine 200 mg, and Tenofovir Alafenamide 10 mg in the Presence of Cobicistat 150 [NCT04661397]Phase 137 participants (Actual)Interventional2021-01-05Completed
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 Retrospective Study to Evaluate the Safety and Efficacy of a Nucleoside-Sparing Regimen of Darunavir, Ritonavir, and Dolutegravir [NCT03198884]20 participants (Actual)Observational2017-01-01Completed
HERV-K Suppression Using Antiretroviral Therapy in Volunteers With Amyotrophic Lateral Sclerosis (ALS) [NCT02437110]Phase 1122 participants (Actual)Interventional2019-04-01Active, not recruiting
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.)
MARaviroc-based Treatment Switch in HIV-positive Patients With HAND: Consequences of Reducing Antiretroviral-associated Neurotoxicity [NCT03163277]Phase 438 participants (Actual)Interventional2017-05-15Terminated(stopped due to Sloww accrual and COVID-19 related problems (impossible to perform LPs))
Population Pharmacokinetics of Antiretroviral in Children [NCT03194165]65 participants (Actual)Observational2017-06-16Completed
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 Phase 3b, Open-Label, Single Arm Study to Evaluate the Safety and Efficacy of Cobicistat-boosted Darunavir Plus Two Fully Active Nucleoside Reverse Transcriptase Inhibitors in HIV-1 Infected, Antiretroviral Treatment-Naïve and -Experienced Adults With N [NCT01440569]Phase 3314 participants (Actual)Interventional2011-09-30Completed
The Pharmacokinetics of Dolutegravir, Darunavir/Cobocistat When Co-administered in Healthy Volunteers [NCT03094507]Phase 121 participants (Actual)Interventional2017-04-19Completed
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
Darunavir/Cobicistat and Dolutegravir to Maintain Virologic Suppression and Reduce NRTI-associated Toxicity (The 'deNUC' Study; TMC114HIV2030) [NCT02499978]Phase 2/Phase 30 participants (Actual)Interventional2016-05-31Withdrawn
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
Mechanisms of Immune Reconstitution & Reduced Immune Activation Following Darunavir-based ART [NCT01869634]Phase 437 participants (Actual)Interventional2013-06-30Completed
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
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
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
HIV Reservoir Dynamics After Switching to Dolutegravir in Patients With Two NRTI and a Protease Inhibitor Based Regimen. A Phase IV Open Randomized Trial [NCT02513147]Phase 444 participants (Actual)Interventional2015-06-30Completed
A Phase 3 Open-label Safety Study of Cobicistat-containing Highly Active Antiretroviral Regimens in HIV-1 Infected Patients With Mild to Moderate Renal Impairment [NCT01363011]Phase 3106 participants (Actual)Interventional2011-05-31Completed
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
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]Pharmacokinetic (PK) Parameter: Infant Plasma Washout Concentration of 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]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 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: 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 mg/L With Median (IQR) 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: Cord/Maternal Blood Concentration Ratio 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: Cord/Maternal Blood Concentration Ratio With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]Plasma Concentration for Contraceptives
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]Area Under the Curve From 0 to 24 Hours (AUC24) of ARVs for Contraceptive Arms
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]Number of Women Who Met PK Target of Area Under the Curve (AUC) for ARVs
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: Trough Concentration (C24) 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 (C12) With Median (Range) 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: Trough Concentration (C12) With Geometric Mean (95% CI) 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: Maximum Concentration (Cmax) in ng/mL With Median (95% CI) for ARVs and TB Drugs
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 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 ETR Subgroup - Change From Baseline in CD4+ Cell Count Using the Imputation Method of LOCF
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 VL < 50 HIV-1 RNA Copies/mL (TLOVR) Subjects by Race
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
NCT00458302 (13) [back to overview]Change From Baseline in Health-Related Quality of Life - FAHI Questionnaire Functional and Global Well-Being Subscale
NCT00458302 (13) [back to overview]Change From Baseline in Health-Related Quality of Life - FAHI Questionnaire Physical Well-Being Subscale
NCT00458302 (13) [back to overview]Change From Baseline in Health-Related Quality of Life - FAHI Questionnaire Total Score
NCT00458302 (13) [back to overview]Mean Change From Baseline in CD4+ Cell Count
NCT00458302 (13) [back to overview]Resistance Determinations
NCT00458302 (13) [back to overview]Virological Response [Intent To Treat (ITT), TLOVR - All Switches Included, < 50 Copies/ml, Week 144]
NCT00458302 (13) [back to overview]Change From Baseline in Health-Related Quality of Life - FAHI Questionnaire Social Well-Being Subscale
NCT00458302 (13) [back to overview]Virological Response [Intent To Treat (ITT) - TLOVR, < 50 Copies/ml, Week 48]
NCT00458302 (13) [back to overview]Virological Response [Per Protocol (PP) - Time to Loss of Virologic Response (TLOVR), < 50 Copies/ml, Week 48]
NCT00458302 (13) [back to overview]Virological Response [Per Protocol (PP), TLOVR - Switch Equals Failure, < 50 Copies/ml, Week 144]
NCT00458302 (13) [back to overview]Virological Response [Per Protocol (PP), TLOVR - Switch Equals Failure, <200 Copies/ml, Week 144]
NCT00458302 (13) [back to overview]Change From Baseline in Health-Related Quality of Life - FAHI Questionnaire Cognitive Function Subscale
NCT00458302 (13) [back to overview]Change From Baseline in Health-Related Quality of Life - FAHI Questionnaire Emotional Well-Being Subscale
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]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 Median Changes From Baseline at 4, 8, 12, 16, 24, 36 and 48 Weeks.
NCT00460746 (10) [back to overview]Median Change From Baseline in Glucose at Week 48.
NCT00460746 (10) [back to overview]Median Change From Baseline in HDL Cholesterol.
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 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]Median Change From Baseline in Total Cholesterol at Week 48.
NCT00460746 (10) [back to overview]Median Change From Baseline in Total Cholesterol (TC) / High Denisty Lipoprotein (HDL) Ratio at Week 48.
NCT00524368 (12) [back to overview]Change in log10 Viral Load From Baseline at Week 48
NCT00524368 (12) [back to overview]Number of Participants Developing Mutations at Endpoint
NCT00524368 (12) [back to overview]Change in CD4+ Cell Count From Baseline
NCT00524368 (12) [back to overview]Change From Baseline in Total Functional Assessment of HIV Infection (FAHI) Score
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]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]Virologic Response at Week 48 (Viral Load Less Than 400 Copies/mL)
NCT00524368 (12) [back to overview]Time to Loss of Virologic Response
NCT00524368 (12) [back to overview]Time to Reach First Virologic Response
NCT00524368 (12) [back to overview]Time-averaged Difference (DAVG) of log10 Plasma Viral Load Over 48 Weeks
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]Predose Plasma Concentration (C0h) of DRV and Rtv.
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.
NCT00534352 (12) [back to overview]Number of Participants Contributing to the Pharmacokinetic (PK) Evaluations: Cmin, Cmax, AUC24 & Css,av
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]Number of Participants With Treatment-Emergent Non-Graded Laboratory Abnormalities(Worst Abnormality): Glucose- Insulin
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 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]CD4+ Cell Count (x 10^6 Cell/L): Baseline and Median Changes From Baseline (ITT-Observed Case)
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 With Treatment-Emergent Non-Graded Laboratory Abnormalities(Worst Abnormality): Lipids- High-density Lipoprotein (HDL)
NCT00537394 (15) [back to overview]Number of Participants With Plasma HIV-1 Viral Load < 50 Copies/ml
NCT00537394 (15) [back to overview]Time From Randomization to Discontinuation of Randomized NRTI Component of Study Treatment
NCT00537394 (15) [back to overview]Time From Treatment Dispensation to First Grade 3 or Higher (and at Least One Grade Higher Than Baseline) Signs/Symptom or Laboratory Abnormality
NCT00537394 (15) [back to overview]Time From Treatment Dispensation to First Study ARV Modification (Excluding NRTIs, if Applicable)
NCT00537394 (15) [back to overview]Time From Treatment Dispensation to Serious Non-AIDS-defining Events
NCT00537394 (15) [back to overview]Change in Summarized Quality of Life Score
NCT00537394 (15) [back to overview]Number of Participants Self-reporting Non-adherence to Assigned Study ARVS (Excluding NRTIs, if Applicable)
NCT00537394 (15) [back to overview]Time From Randomization to Confirmed Virological Failure
NCT00537394 (15) [back to overview]Change in Plasma HIV-1 Viral Load From Baseline to Week 1
NCT00537394 (15) [back to overview]Number of Participants With Change in Virus Co-receptor Tropism Among Those With R5-only Tropic Virus at Study Entry
NCT00537394 (15) [back to overview]Participants With Newly Acquired HIV Drug Resistance Between Study Entry and Confirmed Virologic Failure
NCT00537394 (15) [back to overview]Percent of Participants With Regimen Failure, Defined as a Confirmed Virologic Failure or Discontinuation of Randomized NRTI Component of Study Treatment
NCT00537394 (15) [back to overview]Change in Cardiovascular Risk Score From Baseline
NCT00537394 (15) [back to overview]Change in CD4 Count From Baseline
NCT00537394 (15) [back to overview]Change in Fasting Non-HDL Cholesterol From Baseline
NCT00543101 (4) [back to overview]Economic Impact of a Substitution of Dual Boosted PIs With DRV/r
NCT00543101 (4) [back to overview]The Percentage of Participants With Successful Virologic Suppression
NCT00543101 (4) [back to overview]Lipid Fraction Results, Mean of the Change From Baseline to Week 24.
NCT00543101 (4) [back to overview]Treatment Satisfaction (+3, Much More Satisfied Now to -3, Much Less Satisfied Now)
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 Maximum Plasma Concentration (Cmax)
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 Area Under the Plasma Concentration-time Curve (AUC) Over the Dosing Interval
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 Alone: 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]Pravastatin + Darunavir/Ritonavir: Pravastatin Maximum Plasma Concentration (Cmax)
NCT00630734 (10) [back to overview]Ritonavir Maximum Plasma Concentration (Cmax)
NCT00757783 (16) [back to overview]Antiviral Activity, Human Immunodeficiency Virus Type 1 (HIV-1) RNA.
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]Change From Baseline in Insulin 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 HIV-1 RNA Viral Load 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 Glucose at Week 12 and 48.
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 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 Cluster of Differentiation (CD) 4 Cell Count at Week 12 and 48, Last Observation Carried Forward (LOCF).
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 CD4 Cell Count at Week 12 and 48, Observed Values.
NCT00757783 (16) [back to overview]Change From Baseline in Apolipoprotein A1 in the LE Set at Week 12 and 48.
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 Total Cholesterol (TC) Levels in the LE Set 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.
NCT00811954 (19) [back to overview]Change in Fasting Total 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 HDL Cholesterol Level From Baseline
NCT00811954 (19) [back to overview]CD4+ T-cell Count Changes 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]CD4+ T-cell Count
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 NRTI Resistance
NCT00811954 (19) [back to overview]Presence of Mutations Associated With INI Resistance
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]Self-reported Adherence
NCT00811954 (19) [back to overview]Incidence of Death or AIDS Defining Events (CDC Category C)
NCT00811954 (19) [back to overview]Cumulative Probability of Time to Loss of Virologic Response (TLOVR) by Week 96
NCT00811954 (19) [back to overview]Cumulative Probability of First Virologic Failure by Week 96
NCT00811954 (19) [back to overview]Change in Waist:Height Ratio From Baseline
NCT00811954 (19) [back to overview]Change in Waist Circumference From Baseline
NCT00811954 (19) [back to overview]Change in Framingham 10-year Risk of MI or Coronary Death From Baseline
NCT00811954 (19) [back to overview]Change in Fasting Triglycerides Level From Baseline
NCT00811954 (19) [back to overview]Cumulative Incidence of First Adverse Event by Week 96
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]Maximum Observed Plasma Concentration (Cmax) of Maraviroc
NCT00827112 (16) [back to overview]Minimum Observed Plasma Concentration (Cmin) of Maraviroc
NCT00827112 (16) [back to overview]Number of Participants With Genotypic Resistance
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]Time to Loss of Virological Response (TLOVR)
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
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]Time-Averaged Difference (TAD) in log10 Viral Load
NCT00827112 (16) [back to overview]Number of Participants With Phenotypic Resistance
NCT00827112 (16) [back to overview]Percentage of Participants With Less Than 50 Copies/mL of HIV-1 RNA
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]Change in Fasting Low-density Lipoprotein at Week 24
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]Change in CD4 Count 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 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]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]Plasma Trough Concentration of Raltegravir
NCT00830804 (17) [back to overview]Plasma Trough Concentration of Darunavir
NCT00830804 (17) [back to overview]Number of Participants With Protease Drug Resistance at Virologic Failure
NCT00830804 (17) [back to overview]Number of Participants With Pretreatment Drug Resistance
NCT00830804 (17) [back to overview]Changes in Fasting Total Cholesterol, High-density Lipoprotein and Triglyceride at Week 48
NCT00830804 (17) [back to overview]Changes in Fasting Total Cholesterol, High-density Lipoprotein and Triglyceride at Week 24
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]Proportion of Participants With Plasma HIV-1 RNA <50 Copies/ml or <200 Copies/ml at Week 48
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]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]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
NCT00851799 (27) [back to overview]Fold Change in D-dimer From Study Entry to Weeks 48 and 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]Fold Change in Soluble CD14 From Study Entry to Weeks 48 and 96
NCT00851799 (27) [back to overview]Fold Change in Soluble CD163 From Study Entry to Weeks 48 and 96
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]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]Percent Change in Bone Mineral Density (BMD) of the Hip From Study Entry to Week 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]Percent Change in Visceral Abdominal Fat (VAT) From Study Entry to Week 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
NCT00855335 (15) [back to overview]Mean Change From Baseline in CD4+ Cell Count
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]Time to Reach the Maximum Plasma Concentration (Tmax)
NCT00855335 (15) [back to overview]Predose (Trough) Plasma Concentration (C0h)
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 Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA) Plasma Viral Load (<) 50 Copies/Milliliter (mL)
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 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]Area Under the Plasma Concentration-Time Curve From Time of Administration to 24 Hours Post-dose (AUC0-24h)
NCT00855335 (15) [back to overview]Mean Change From Baseline in CD4+ Cell Count
NCT00855335 (15) [back to overview]Maximum Plasma Concentration (Cmax)
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 Participants With Resistance at Virological Failure
NCT00855335 (15) [back to overview]Number of Infants With Human Immunodeficiency Virus (HIV) Positive Test Result
NCT00855413 (38) [back to overview]Overall Neurocognitive Impairment at Week 24
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 Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure
NCT00855413 (38) [back to overview]Correlation of HIV RNA Levels in CSF and Drug Levels With Neurocognitive Functioning
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]Number of Participants With Virologic Response
NCT00855413 (38) [back to overview]Number of Participants With Neurocognitive Impairment at Baseline
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]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]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]Median Change in CD4 Cell Count From Week 0 to Week 48.
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 Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure
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]Number of Participants With Neurocognitive Impairment at Week 24
NCT00855413 (38) [back to overview]Number of Participants With Neurocognitive Impairment at Week 48
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 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]Minimum Etravirine Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture
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]Change in Overall Neurocognitive Impairment From Baseline to Week 24 or 48
NCT00855413 (38) [back to overview]Adverse Events Possibly or Definitely Related to Study Treatment Through Week 48
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]Number of Participants With Virologic Response
NCT00855413 (38) [back to overview]HIV RNA Detection in Semen
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]Minimum Darunavir Exposure Range in Semen Among Participants Who Consented to an Optional Collection of Semen
NCT00855413 (38) [back to overview]Maximum Darunavir Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen
NCT00885495 (4) [back to overview]To Investigate the Effect of Rosuvastatin on the Steady State Pharmacokinetics of Darunavir/Ritonavir.
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]Cmax of Rosuvastatin
NCT00885495 (4) [back to overview]AUC of Rosuvastatin
NCT00915655 (2) [back to overview]Virological Response[Viral Load <50 Copies/mL, TLOVR]
NCT00915655 (2) [back to overview]Virological Response [Viral Load <50 Copies/mL, FDA-SNAPSHOT]
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 Plasma log10 Viral Load
NCT00919854 (6) [back to overview]Mean Change From Baseline to Week 24 and Week 48 in CD4+ Percentage
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 Less Than or Equal to 1 log10 Decrease in Plasma Viral Load at Week 24 and 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
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
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 WHO Stage 3 and 4 HIV Related Events
NCT00928187 (13) [back to overview]Tolerance: Neuropathies (Grade 1 to 4)
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]Adherence
NCT00928187 (13) [back to overview]Number of Patients With HIV Plasma Viral Load < 50 Copies/ml
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]Number of Patients Discontinuing Study Treatment
NCT00993148 (9) [back to overview]Proportion 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]Trough Concentrations (Ctrough) of Maraviroc
NCT00993148 (9) [back to overview]Percentage of Participants With Plasma HIV-1 RNA >50 Copies/mL
NCT00993148 (9) [back to overview]Drug Adherence, Number of Participants With Missed Doses
NCT00993148 (9) [back to overview]Drug Resistance Mutations and Co-receptor Tropism Assessed by Trofile ES
NCT00993148 (9) [back to overview]Median CD4 Count Change From Baseline
NCT00993148 (9) [back to overview]Percentage of Participants With Plasma HIV-1 RNA >50
NCT00993148 (9) [back to overview]Percentage of Participants With Virologic Failure or Off Study Treatment Regimen
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 8
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]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]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 30
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 24
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 16
NCT01199939 (14) [back to overview]Number of Participants With Virologic Failure
NCT01199939 (14) [back to overview]Time to Reach First Confirmed Virologic Response
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
NCT01258374 (4) [back to overview]Geometric Mean of t1/2, of Raltegravir (RAL) at a Dose of 400 mg Twice a Day Plus Darunavir/Ritonavir (DRV/RTV) at a Dose of 800/100 mg Once a Day in HIV-1-infected Patients Were Mesured After 15 Days of Therapy
NCT01258374 (4) [back to overview]Geometric Mean of C-max, of Raltegravir (RAL) at a Dose of 400 mg Twice a Day Plus Darunavir/Ritonavir (DRV/RTV) at a Dose of 800/100 mg Once a Day in HIV-1-infected Patients Were Mesured After 15 Days of Therapy.
NCT01258374 (4) [back to overview]Geometric Mean of AUC, of Raltegravir (RAL) at a Dose of 400 mg Twice a Day Plus Darunavir/Ritonavir (DRV/RTV) at a Dose of 800/100 mg Once a Day in HIV-1-infected Patients Were Mesured After 15 Days of Therapy.
NCT01258374 (4) [back to overview]Geometric Mean of C-Trough, of Raltegravir (RAL) at a Dose of 400 mg Twice a Day Plus Darunavir/Ritonavir (DRV/RTV) at a Dose of 800/100 mg Once a Day in HIV-1-infected Patients Were Mesured After 15 Days of Therapy.
NCT01281813 (3) [back to overview]Number of Participants With Serious Adverse Events
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 Adverse Events Leading to Study Drug Discontinuation
NCT01345630 (25) [back to overview]Tropism Change Between Screening or Baseline and PDTF
NCT01345630 (25) [back to overview]Virologic Outcomes at Week 48 Using Protocol-Defined Treatment Failure (PDTF).
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]Severity of Abnormal Laboratory Values
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]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]Changes in Peripheral Fat Distribution Using Dual Energy X-ray Absorptiometry [DEXA] Scan From Baseline and at Week 48.
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]Frequency of Adverse Events (AE).
NCT01345630 (25) [back to overview]Number of Participants Who Discontinued Due to AEs
NCT01345630 (25) [back to overview]Number of Participants With Grade 3 or 4 AEs
NCT01345630 (25) [back to overview]Number of Participants With Treatment-emergent Serious Adverse Events
NCT01345630 (25) [back to overview]Number of Treatment-related AEs
NCT01345630 (25) [back to overview]Percentage of Participants With Plasma HIV-1 RNA <50 Copies/mL.
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]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]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 in CD4+/CD8+ Ratio From Baseline to Week 48
NCT01345630 (25) [back to overview]Number of Participants With Abnormal Laboratory Values
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 Viral Resistance to Maraviroc (Maraviroc Treated Participants Only) in Participants Meeting PDTF Criteria.
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]Percent Change From Baseline in Immune Cell Function at Week 48: Lymphocyte Activation Marker CD8 (%)
NCT01348763 (2) [back to overview]The Ratio of the Maximum Plasma Concentration of Maraviroc Between 20 and 10 Day
NCT01348763 (2) [back to overview]Number of Participants With Changes in Haematology and Biochemistry Laboratory Tests
NCT01363011 (36) [back to overview]Plasma Pharmacokinetics of COBI: Ctau (Cohort 1)
NCT01363011 (36) [back to overview]Plasma Pharmacokinetics of COBI: Ctau (Cohort 2)
NCT01363011 (36) [back to overview]Plasma Pharmacokinetics of COBI: t1/2 (Cohort 1)
NCT01363011 (36) [back to overview]Plasma Pharmacokinetics of COBI: t1/2 (Cohort 2)
NCT01363011 (36) [back to overview]Plasma Pharmacokinetics of COBI: Tmax (Cohort 2)
NCT01363011 (36) [back to overview]Change From Baseline in eGFR Using the Modification of Diet in Renal (MDRD) Equation at Week 24 (Cohort 1)
NCT01363011 (36) [back to overview]Plasma Pharmacokinetics of COBI: Tmax (Cohort 1)
NCT01363011 (36) [back to overview]Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 24 (Cohort 1)
NCT01363011 (36) [back to overview]Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 24 (Cohort 2)
NCT01363011 (36) [back to overview]Change From Baseline in Actual Glomerular Filtration Rate (aGFR) at Weeks 2, 4, and 24 (Cohort 1)
NCT01363011 (36) [back to overview]Change From Baseline in aGFR at Weeks 2, 4, and 24 (Cohort 2)
NCT01363011 (36) [back to overview]Change From Baseline in eGFR Using the Chronic Kidney Disease, Epidemiology Collaboration (CKD-EPI) Formula Based on Cystatin C Equation at Week 24 (Cohort 1)
NCT01363011 (36) [back to overview]Change From Baseline in eGFR-CG at Week 24 (Cohort 2)
NCT01363011 (36) [back to overview]Change From Baseline in eGFR-CG at Weeks 48 and 96 (Cohort 1)
NCT01363011 (36) [back to overview]Change From Baseline in eGFR-CG at Weeks 48 and 96 (Cohort 2)
NCT01363011 (36) [back to overview]Change From Baseline in eGFR-CKD-EPI Based on Cystatin C Equation (Adjusted) at Weeks 48 and 96 (Cohort 1)
NCT01363011 (36) [back to overview]Change From Baseline in eGFR-CKD-EPI Based on Cystatin C Equation (Adjusted) at Weeks 48 and 96 (Cohort 2)
NCT01363011 (36) [back to overview]Change From Baseline in eGFR-CKD-EPI Based on Cystatin C Equation at Weeks 48 and 96 (Cohort 1)
NCT01363011 (36) [back to overview]Change From Baseline in eGFR-CKD-EPI Based on Cystatin C Equation at Weeks 48 and 96 (Cohort 2)
NCT01363011 (36) [back to overview]Change From Baseline in eGFR-CKD-EPI Based on Cystatin C Equation, Adjusted at Week 24 (Cohort 1)
NCT01363011 (36) [back to overview]Change From Baseline in eGFR-CKD-EPI Based on Cystatin C Equation, Adjusted at Week 24 (Cohort 2)
NCT01363011 (36) [back to overview]Change From Baseline in eGFR-CKD-EPI Formula Based on Cystatin C Equation at Week 24 (Cohort 2)
NCT01363011 (36) [back to overview]Change From Baseline in eGFR-MDRD at Week 24 (Cohort 2)
NCT01363011 (36) [back to overview]Change From Baseline in eGFR-MDRD at Weeks 48 and 96 (Cohort 1)
NCT01363011 (36) [back to overview]Change From Baseline in eGFR-MDRD at Weeks 48 and 96 (Cohort 2)
NCT01363011 (36) [back to overview]Change From Baseline in Estimated Glomerular Filtration Rate (eGFR) Using the Cockcroft-Gault (CG) Equation at Week 24 (Cohort 1)
NCT01363011 (36) [back to overview]Percentage of Participants Who Experienced Adverse Events (Cohort 1)
NCT01363011 (36) [back to overview]Percentage of Participants Who Experienced Adverse Events (Cohort 2)
NCT01363011 (36) [back to overview]Percentage of Participants Who Experienced Graded Laboratory Abnormalities (Cohort 1)
NCT01363011 (36) [back to overview]Percentage of Participants Who Experienced Graded Laboratory Abnormalities (Cohort 2)
NCT01363011 (36) [back to overview]Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Weeks 48 and 96 (Cohort 1)
NCT01363011 (36) [back to overview]Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Weeks 48 and 96 (Cohort 2)
NCT01363011 (36) [back to overview]Plasma Pharmacokinetics of COBI: AUCtau (Cohort 1)
NCT01363011 (36) [back to overview]Plasma Pharmacokinetics of COBI: AUCtau (Cohort 2)
NCT01363011 (36) [back to overview]Plasma Pharmacokinetics of COBI: Cmax (Cohort 1)
NCT01363011 (36) [back to overview]Plasma Pharmacokinetics of COBI: Cmax (Cohort 2)
NCT01374802 (4) [back to overview]AUCτ,ss of Darunavir
NCT01374802 (4) [back to overview]Cmax,ss of Darunavir
NCT01374802 (4) [back to overview]Cτ,ss of Darunavir
NCT01374802 (4) [back to overview]Tmax,ss of Darunavir
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 Lumbar Z Score: Median Change in Lumbar Z Score
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 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 Brachial Artery FMD: Median Change in FMD (%)
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 Femoral Neck T Score: Median Change in Femoral Neck T Score
NCT01391013 (17) [back to overview]Change From Baseline to Week 48 in Visceral Fat Content in Abdomen: Median Change in Visceral Abdominal Tissue (VAT)
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 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 Mean High-density Lipoprotein (HDL) Cholesterol at Week 24 and Week 48: Median Change in HDL
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 Triglycerides at Week 24 and Week 48: Median Change in Triglycerides
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 48 in Precursors of Circulating Endothelial Cells
NCT01400412 (20) [back to overview]Change in Levels of sCD14 From Baseline
NCT01400412 (20) [back to overview]Change in Levels of sCD163 From Baseline to Week 48
NCT01400412 (20) [back to overview]Cumulative Probability of Virologic Failure by 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]Number of Participants Who Experienced Bone Fractures
NCT01400412 (20) [back to overview]Percent Change From Baseline in Total Hip Bone Mineral Density (BMD)
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]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 CD57+ on CD8+ T Cells From Baseline to Week 48
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]Percent Change in Lumbar Spine Bone Mineral Density (BMD)
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]Change in Level of IP-10 From Baseline to Week 48
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]CD8+ T-cell Change From Baseline to Week 48
NCT01400412 (20) [back to overview]Change in CD4 Count From Baseline to Week 24
NCT01400412 (20) [back to overview]Change in CD4 Count From Baseline to Week 48
NCT01400412 (20) [back to overview]Change in Levels of D-dimer From Baseline
NCT01400412 (20) [back to overview]Change in Levels of IL-6 From Baseline to Week 48
NCT01422369 (2) [back to overview]Number of Participants With at Least One Adverse Event.
NCT01422369 (2) [back to overview]NK-104 AUC
NCT01423812 (7) [back to overview]Change in Total Cholesterol From Baseline to 48 Weeks
NCT01423812 (7) [back to overview]Number of Participants With Greater Than 95% Adherence at 48 Weeks
NCT01423812 (7) [back to overview]Primary Efficacy Endpoint for Virologic Suppression in HIV-infected Subjects
NCT01423812 (7) [back to overview]Secondary Efficacy Endpoints
NCT01423812 (7) [back to overview]Secondary Efficacy Endpoints
NCT01423812 (7) [back to overview]Absolute Value Change in CD4+ From Baseline to Week 48
NCT01423812 (7) [back to overview]Assessment of Virologic Failure
NCT01440569 (7) [back to overview]Percentage of Participants With Onset of Any Treatment-emergent Grade 3 or 4 Adverse Event Between Baseline and Week 24
NCT01440569 (7) [back to overview]Change From Baseline in CD4+ Cell Count at Week 24
NCT01440569 (7) [back to overview]Change From Baseline in CD4+ Cell Count at Week 48
NCT01440569 (7) [back to overview]Percentage of Participants Achieving HIV-1 RNA < 50 Copies/mL at Week 24 (Snapshot Analysis)
NCT01440569 (7) [back to overview]Percentage of Participants Achieving HIV-1 RNA < 50 Copies/mL at Week 48 (Snapshot Analysis)
NCT01440569 (7) [back to overview]Percentage of Participants Experiencing Any Treatment-emergent Adverse Event and Any Treatment-emergent Adverse Event Leading to Discontinuation of Study Drug Through Week 24
NCT01440569 (7) [back to overview]Percentage of Participants Experiencing Any Treatment-emergent Adverse Event and Any Treatment-emergent Adverse Event Leading to Discontinuation of Study Drug Through Week 48
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
NCT01448707 (7) [back to overview]Virologic Response (FDA Snapshot, Switch Included)
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]Change From Baseline in Global Neurocognitive Performance z-Score
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
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 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]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]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]Time to Virologic Suppression (<50 Copies/mL) Through 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
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]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 Treatment-Emergent Adverse Events (TEAEs)
NCT01565850 (6) [back to overview]Change From Baseline in HIV-1 RNA at Week 48
NCT01565850 (6) [back to overview]Change From Baseline in HIV-1 RNA at Week 24
NCT01565850 (6) [back to overview]Change From Baseline in CD4+ Cell Count at Week 48
NCT01565850 (6) [back to overview]Change From Baseline in CD4+ Cell Count at Week 24
NCT01565850 (6) [back to overview]Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 48
NCT01565850 (6) [back to overview]Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 24
NCT01641367 (60) [back to overview]Time to Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study
NCT01641367 (60) [back to overview]Percent of Participants That Developed Immune Reconstitution Inflammatory Syndrome (IRIS) by Week 48 [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Number of Participants With Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study
NCT01641367 (60) [back to overview]Number of Participants With Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Number of Participants With Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study, With a New Resistance-associated Mutation Detected in Population-based Sequencing
NCT01641367 (60) [back to overview]Percent of Participants With a Dose Modification Due to Grade 3 or 4 Toxicity by Week 48
NCT01641367 (60) [back to overview]Time to First Dose Modification Due to Grade 3 or 4 Toxicity [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Time to First Dose Modification Due to Grade 3 or 4 Toxicity
NCT01641367 (60) [back to overview]Time to Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study, With a New Resistance-associated Mutation Detected in Population-based Sequencing [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Time to Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study, With a New Resistance-associated Mutation Detected in Population-based Sequencing
NCT01641367 (60) [back to overview]Time to Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of Triglycerides
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to Treatment Modification or Discontinuation.
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to Treatment Modification or Discontinuation Due to Toxicity [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to Treatment Modification or Discontinuation Due to Toxicity
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to Treatment Modification or Discontinuation [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to the First of Death, an AIDS-defining Event or a Non-AIDS-defining Event [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to the First of Death, an AIDS-defining Event or a Non-AIDS-defining Event
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to the First of Death or Hospitalization.
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to the Development of Immune Reconstitution Inflammatory Syndrome (IRIS) [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to the Development of Immune Reconstitution Inflammatory Syndrome (IRIS)
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to Death [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to Death
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of Triglycerides [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of Total Cholesterol [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of Total Cholesterol
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of High-density Lipoprotein Cholesterol [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of High-density Lipoprotein Cholesterol
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of Glucose [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of Glucose
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of Calculated Low-density Lipoprotein Cholesterol [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to the First of Death or Hospitalization [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Percent of Participants With Treatment Modification or Discontinuation by Week 48 [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of Calculated Low-density Lipoprotein Cholesterol
NCT01641367 (60) [back to overview]Change From Baseline in CD4+ T-cell Count [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Change From Baseline in CD4+ T-cell Count
NCT01641367 (60) [back to overview]Proportion of Participants With Plasma HIV-1 RNA ≤200 Copies/mL at 72 Weeks [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Proportion of Participants With Plasma HIV-1 RNA ≤200 Copies/mL at 72 Weeks
NCT01641367 (60) [back to overview]Proportion of Participants With Plasma HIV-1 RNA ≤200 Copies/mL at 48 Weeks [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Proportion of Participants With Plasma HIV-1 RNA ≤200 Copies/mL at 48 Weeks
NCT01641367 (60) [back to overview]Proportion of Participants With Plasma HIV-1 RNA ≤200 Copies/mL at 24 Weeks [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Proportion of Participants With Plasma HIV-1 RNA ≤200 Copies/mL at 24 Weeks
NCT01641367 (60) [back to overview]Percent of Participants With Treatment Modification or Discontinuation Due to Toxicity by Week 48 [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Percent of Participants With Treatment Modification or Discontinuation Due to Toxicity by Week 48
NCT01641367 (60) [back to overview]Percent of Participants Experiencing Death, AIDS-defining Event or a Non-AIDS-defining Event by Week 48
NCT01641367 (60) [back to overview]Percent of Participants With Treatment Modification or Discontinuation by Week 48
NCT01641367 (60) [back to overview]Percent of Participants With Death or Hospitalization by Week 48 [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Percent of Participants With Death or Hospitalization by Week 48
NCT01641367 (60) [back to overview]Percent of Participants With Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study, With a New Resistance-associated Mutation Detected in Population-based Sequencing, by Week 48 [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Percent of Participants With Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study, With a New Resistance-associated Mutation Detected in Population-based Sequencing, by Week 48
NCT01641367 (60) [back to overview]Percent of Participants With Confirmed Virologic Failure by Week 48 [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Number of Participants With Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study, With a New Resistance-associated Mutation Detected in Population-based Sequencing [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Number of Weeks of Follow-up
NCT01641367 (60) [back to overview]Percent of Participants With a Dose Modification Due to Grade 3 or 4 Toxicity by Week 48 [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Number of Weeks of Follow-up [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Percent of Participants Experiencing Death by Week 48
NCT01641367 (60) [back to overview]Percent of Participants Experiencing Death by Week 48 [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Percent of Participants With Confirmed Virologic Failure by Week 48
NCT01641367 (60) [back to overview]Percent of Participants Experiencing Death, AIDS-defining Event or a Non-AIDS-defining Event by Week 48 [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Percent of Participants That Developed Immune Reconstitution Inflammatory Syndrome (IRIS) by Week 48
NCT01695954 (3) [back to overview]AUC
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
NCT01695954 (3) [back to overview]GMR of Cmax of Pitavastatin When Coadministered With Efavirenz or With Darunavir/Ritonavir
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
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]Time of Maximum Observed Plasma Concentration (Tmax) of Daclatasvir
NCT02159352 (11) [back to overview]Plasma Concentration Observed at 24 Hours Postdose (C24) of Daclatasvir
NCT02159352 (11) [back to overview]Number of Participants With Abnormalities in Vital Sign Measurements
NCT02159352 (11) [back to overview]Maximum Observed Plasma Concentration (Cmax) for Daclatasvir
NCT02159352 (11) [back to overview]Number of Participants With Abnormalities in Electrocardiogram (ECG) Findings
NCT02159352 (11) [back to overview]Number of Participants With Abnormalities in Urinalysis and Other Chemistry Testing Results
NCT02159352 (11) [back to overview]Number of Participants With Marked Abnormalities in Hematology Laboratory Test Results
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 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]Area Under the Concentration-Time Curve in 1 Dosing Interval (AUC[TAU]) for Daclatasvir
NCT02159352 (11) [back to overview]Dose-normalized Area Under the Concentration-Time Curve in 1 Dosing Interval (AUC[TAU]/D) of Daclatasvir
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 Non-High Density Lipoprotein Cholesterol (Non-HDL-C) at Week 48
NCT02275780 (16) [back to overview]Mean Change From Baseline in Fasting Total Cholesterol at Week 48
NCT02275780 (16) [back to overview]Mean Change From Baseline in Fasting Triglyceride at Week 48
NCT02275780 (16) [back to overview]Percentage of Participants Achieving Plasma HIV-1 RNA <50 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]Change From Baseline in Mean CD4+ T-cell Count 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]Percentage of Participants Achieving Plasma HIV-1 RNA <50 Copies/mL 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 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]Mean Change From Baseline in Fasting High Density Lipoprotein Cholesterol (HDL-C) at Week 48
NCT02431247 (64) [back to overview]Change From Baseline in Alkaline Phosphatase (ALP) Levels at Weeks 24 and 48
NCT02431247 (64) [back to overview]Change From Baseline in BMD T-score of Hip and Spine
NCT02431247 (64) [back to overview]Change From Baseline in Levels of 25-Hydroxyvitamin D (25-OH Vitamin D), at Week 24 and 48
NCT02431247 (64) [back to overview]Change From Baseline in Levels of Parathyroid Hormone (PTH) at Weeks 24 and 48
NCT02431247 (64) [back to overview]Change From Baseline in Levels of Serum Collagen Type 1 Beta Carboxy Telopeptide (CTX) at Weeks 24 and 48
NCT02431247 (64) [back to overview]Change From Baseline in Levels of Serum Procollagen 1 N-Terminal Propeptide (P1NP) at Weeks 24 and 48
NCT02431247 (64) [back to overview]Change From Reference in BMD T-score of Hip and Spine at Week 96
NCT02431247 (64) [back to overview]Number of Participants With ARV Resistance
NCT02431247 (64) [back to overview]Percent Change From Reference in Hip and Spine BMD
NCT02431247 (64) [back to overview]Percentage of Participants With >95% Treatment Adherence Assessed by Drug Accountability
NCT02431247 (64) [back to overview]Percentage of Participants With >95% Treatment Adherence Assessed by Drug Accountability
NCT02431247 (64) [back to overview]Percentage of Participants With >95% Treatment Adherence Assessed by Drug Accountability
NCT02431247 (64) [back to overview]Percentage of Participants With Grade 3 and 4 Adverse Events (AEs), Serious Adverse Events (SAEs), and Premature Discontinuations Due to Adverse Events Through Week 48
NCT02431247 (64) [back to overview]Percentage of Participants With Grade 3 and 4 Adverse Events (AEs), Serious Adverse Events (SAEs), and Premature Discontinuations Due to Adverse Events Through Week 96
NCT02431247 (64) [back to overview]Percentage of Participants With Grade 3 and 4 AEs, SAEs, and Premature Discontinuations Due to Adverse Events Post-Week 96 to End of Extension
NCT02431247 (64) [back to overview]Percentage of Participants With HIV RNA <50, <20, and <200 Copies/mL Post-week 96 to End of Extension
NCT02431247 (64) [back to overview]Percentage of Participants With HIV-1 RNA < 20, 50, and 200 Copies Per mL at Week 48 and 96 Defined by the Time to Loss of Virologic Response (TLOVR) Algorithm
NCT02431247 (64) [back to overview]Percentage of Participants With HIV-1 RNA < 20, 50, and 200 Copies Per mL at Week 48 and 96 Defined by the Time to Loss of Virologic Response (TLOVR) Algorithm
NCT02431247 (64) [back to overview]Percentage of Participants With HIV-1 RNA < 20, 50, and 200 Copies Per mL at Week 48 and 96 Defined by the Time to Loss of Virologic Response (TLOVR) Algorithm
NCT02431247 (64) [back to overview]Percentage of Participants With HIV-1 RNA <20 and 200 Copies Per mL at Weeks 48 and 96 Defined by FDA Snapshot Approach
NCT02431247 (64) [back to overview]Percentage of Participants With HIV-1 RNA <20 and 200 Copies Per mL at Weeks 48 and 96 Defined by FDA Snapshot Approach
NCT02431247 (64) [back to overview]Percentage of Participants With HIV-1 RNA <20 and 200 Copies Per mL at Weeks 48 and 96 Defined by FDA Snapshot Approach
NCT02431247 (64) [back to overview]Percentage of Participants With Non-PDVF by Kaplan-Meier Estimates
NCT02431247 (64) [back to overview]Percentage of Participants With PDVF Post-week 96 to End of Extension
NCT02431247 (64) [back to overview]Percentage of Participants With Protocol-defined Virologic Failure (PDVF)
NCT02431247 (64) [back to overview]Percentage of Participants With Protocol-defined Virologic Failure (PDVF)
NCT02431247 (64) [back to overview]Percentage of Participants With Protocol-defined Virologic Failure (PDVF)
NCT02431247 (64) [back to overview]Percentage of Participants With Time to Treatment Failure by Kaplan-Meier Estimates
NCT02431247 (64) [back to overview]Percentage of Participants With Time to Treatment Failure by Kaplan-Meier Estimates
NCT02431247 (64) [back to overview]Change From Baseline in Serum Creatinine at Week 48
NCT02431247 (64) [back to overview]Percent Change From Baseline in Hip and Spine Bone Mineral Density (BMD)
NCT02431247 (64) [back to overview]Area Under the Plasma Concentration Time Curve Across the Dosing Interval (AUCtau) of Tenofovir Alafenamide
NCT02431247 (64) [back to overview]Area Under the Plasma Concentration-Time Curve From Time of Administration to 24 Hours Post-dose (AUC0-24h) of Darunavir
NCT02431247 (64) [back to overview]Change From Baseline in Cluster of Differentiation-4 (CD4+) Cell Count at Week 48
NCT02431247 (64) [back to overview]Change From Baseline in Estimated Glomerular Filtration Rate Based on Serum Creatinine (eGFRcr) by Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) Formula at Week 48
NCT02431247 (64) [back to overview]Change From Baseline in Estimated Glomerular Filtration Rate Based on Serum Creatinine by (Cockcroft-Gault Formula) at Week 48
NCT02431247 (64) [back to overview]Plasma Concentrations 2 Hours After Dosing (C0-2h) of Tenofovir Alafenamide
NCT02431247 (64) [back to overview]Change From Baseline in Estimated Glomerular Filtration Rate Based on Serum Cystatin C (eGFRcyst) by CKD-EPI Formula at Week 48
NCT02431247 (64) [back to overview]Change From Baseline in log10 HIV-1 RNA Levels at Week 48
NCT02431247 (64) [back to overview]Change From Baseline in Urine Albumin to Creatinine Ratio (UACR) at Week 48
NCT02431247 (64) [back to overview]Change From Baseline in Urine Beta-2 Microglobulin to Creatinine Ratio (UB2MGCR) at Week 48
NCT02431247 (64) [back to overview]Change From Baseline in Urine Protein to Creatinine Ratio (UPCR) at Week 48
NCT02431247 (64) [back to overview]Change From Baseline in Urine Retinol Binding Protein To Creatinine Ratio (URBPCR) at Week 48
NCT02431247 (64) [back to overview]Change From Reference in ALP Levels
NCT02431247 (64) [back to overview]Change From Reference in CD4+ Cell Count at Week 96
NCT02431247 (64) [back to overview]Change From Reference in eGFRcr by CKD-EPI Formula
NCT02431247 (64) [back to overview]Change From Reference in Estimated Glomerular Filtration Rate Based on Serum Creatinine by Cockcroft-Gault Formula
NCT02431247 (64) [back to overview]Change From Reference in Estimated Glomerular Filtration Rate Based on Serum Cystatin C (eGFRcyst) by CKD-EPI Formula
NCT02431247 (64) [back to overview]Change From Reference in Levels of 25-OH Vitamin D
NCT02431247 (64) [back to overview]Change From Reference in Levels of PTH
NCT02431247 (64) [back to overview]Change From Reference in Levels of Serum CTX
NCT02431247 (64) [back to overview]Change From Reference in Levels of Serum P1NP
NCT02431247 (64) [back to overview]Change From Reference in log10 HIV-1 RNA Levels at Week 96
NCT02431247 (64) [back to overview]Change From Reference in Serum Creatinine
NCT02431247 (64) [back to overview]Change From Reference in UACR
NCT02431247 (64) [back to overview]Change From Reference in UB2MGCR
NCT02431247 (64) [back to overview]Change From Reference in UPCR
NCT02431247 (64) [back to overview]Change From Reference in URBPCR
NCT02431247 (64) [back to overview]Percent Change From Baseline in Urine Fractional Excretion of Phosphate (FEPO4) at Week 48
NCT02431247 (64) [back to overview]Percent Change From Reference in Urine FEPO4
NCT02431247 (64) [back to overview]Percentage of Participants With HIV-1 RNA <50 Copies Per mL at Week 96 Defined by FDA Snapshot Approach
NCT02431247 (64) [back to overview]Percentage of Participants With Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA) Less Than (<) 50 Copies Per Milliliter (Copies Per mL) (Virologic Response) at Week 48 Defined by Food and Drug Administration (FDA) Snapshot Approach
NCT02431247 (64) [back to overview]Predose (Trough) Plasma Concentration (C0h) of Darunavir
NCT02431247 (64) [back to overview]CD4+ Cell Count Post-Week From 96 to End of Extension
NCT02625207 (24) [back to overview]Part 1: Time to Reach Maximum Observed Plasma Concentration (Tmax) of Maraviroc
NCT02625207 (24) [back to overview]Part 1: Time to Reach Maximum Observed Plasma Concentration (Tmax) of Metabolites of Maraviroc
NCT02625207 (24) [back to overview]Part 1: Plasma Concentration of Metabolites of Maraviroc at 12 Hour Post-dose
NCT02625207 (24) [back to overview]Part 1: Number of Participants With Treatment Emergent Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT02625207 (24) [back to overview]Part 1: Metabolite to Parent Ratio for Area Under the Concentration-Time Curve From Time 0 to 12 Hours for Maraviroc and Its Metabolites (MRAUC12)
NCT02625207 (24) [back to overview]Part 1: Maximum Observed Plasma Concentration (Cmax) of Metabolites of Maraviroc
NCT02625207 (24) [back to overview]Part 2: Plasma Concentration of Maraviroc at 24 Hours Post-dose
NCT02625207 (24) [back to overview]Part 1: Average Plasma Concentration (Cav) of Metabolites of Maraviroc
NCT02625207 (24) [back to overview]Part 1: Area Under The Plasma Concentration-Time Curve From Time 0 to 12 Hours (AUC [0-12]) of Metabolites of Maraviroc
NCT02625207 (24) [back to overview]Part 2: Time to Reach Maximum Observed Plasma Concentration (Tmax) of Maraviroc
NCT02625207 (24) [back to overview]Part 2: Number of Participants With Laboratory Abnormalities
NCT02625207 (24) [back to overview]Part 2: Number of Participants With Clinically Significant Vital Sign Abnormalities
NCT02625207 (24) [back to overview]Part 2: Number of Participants With 12-Lead Electrocardiogram (ECG) Abnormalities
NCT02625207 (24) [back to overview]Part 1: Area Under The Plasma Concentration-Time Curve From Time 0 to 12 Hours (AUC [0-12]) of Maraviroc
NCT02625207 (24) [back to overview]Part 1: Average Plasma Concentration (Cavg) of Maraviroc
NCT02625207 (24) [back to overview]Part 1: Maximum Observed Plasma Concentration (Cmax) of Maraviroc
NCT02625207 (24) [back to overview]Part 1: Number of Participants With 12-Lead Electrocardiogram (ECG) Abnormalities
NCT02625207 (24) [back to overview]Part 1: Number of Participants With Clinically Significant Vital Sign Abnormalities
NCT02625207 (24) [back to overview]Part 2: Area Under The Plasma Concentration-Time Curve From Time 0 to 24 Hours (AUC [0-24]) of Maraviroc
NCT02625207 (24) [back to overview]Part 2: Maximum Observed Plasma Concentration (Cmax) of Maraviroc
NCT02625207 (24) [back to overview]Part 2: Average Plasma Concentration (Cavg) of Maraviroc
NCT02625207 (24) [back to overview]Part 1: Number of Participants With Laboratory Abnormalities
NCT02625207 (24) [back to overview]Part 1: Plasma Concentration of Maraviroc at 12 Hours Post-dose
NCT02625207 (24) [back to overview]Part 2: Number of Participants With Treatment Emergent Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT03123848 (10) [back to overview]Number of Participants With Adverse Events (AEs)
NCT03123848 (10) [back to overview]Apparent Total Body Clearance of Drug at the Terminal Phase After Extravascular Administration (CL/F)
NCT03123848 (10) [back to overview]Time to Reach the Maximum Plasma Concentration (Tmax)
NCT03123848 (10) [back to overview]Terminal Elimination Half-Life (t1/2)
NCT03123848 (10) [back to overview]Maximum Observed Plasma Concentration (Cmax)
NCT03123848 (10) [back to overview]Elimination Rate Constant (Lambda[z])
NCT03123848 (10) [back to overview]Concentration at Last Quantifiable Time Point (Clast)
NCT03123848 (10) [back to overview]Area Under the Plasma Concentration-Time Curve From Time Zero to Time the Last Quantifiable Time (AUC[0-last])
NCT03123848 (10) [back to overview]Area Under the Plasma Concentration-Time Curve From Time Zero to Infinite Time (AUC0-infinity)
NCT03123848 (10) [back to overview]Apparent Volume of Distribution (Vz/F)
NCT03198884 (7) [back to overview]The Change in Serum Creatinine From Baseline to 48 Weeks.
NCT03198884 (7) [back to overview]Number of Participants With RNA <50 Copies/mL at 48 Weeks
NCT03198884 (7) [back to overview]Number of Participants With RNA <50 Copies/mL at 24, 36, and 48 Weeks
NCT03198884 (7) [back to overview]Change in Mean CD4+ Cell Count From Baseline.
NCT03198884 (7) [back to overview]Analysis of Creatinine Clearance at Time Points 24, 36 and 48 Weeks.
NCT03198884 (7) [back to overview]Incidence of Adverse Events.
NCT03198884 (7) [back to overview]Number of Grade 1 Adverse Events Reported
NCT03864406 (8) [back to overview]Terminal Elimination Half-life (t½) for Rivaroxaban
NCT03864406 (8) [back to overview]Apparent Oral Clearance (CL/F) for Rivaroxaban
NCT03864406 (8) [back to overview]Apparent Volume of Distribution (V/F) for Rivaroxaban
NCT03864406 (8) [back to overview]Time to Maximum Plasma Concentration (Tmax) for Rivaroxaban
NCT03864406 (8) [back to overview]Area Under the Concentration Versus Time Curve (AUC0-24hr) for Rivaroxaban
NCT03864406 (8) [back to overview]Area Under the Concentration Versus Time Curve (AUC0-∞) for Rivaroxaban
NCT03864406 (8) [back to overview]Maximum Total Plasma Concentration (Cmax) for Rivaroxaban
NCT03864406 (8) [back to overview]Minimum Total Plasma Concentration (Cmin) for Rivaroxaban
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: Number of Participants With Clinically Significant Abnormal Electrocardiogram (ECG) Findings
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 GSK3640254
NCT04630002 (54) [back to overview]Cohort 1: Tmax of RTV
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 Grade Increase Post-Baseline Relative to Baseline in Urinalysis Parameters
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 2: Cmax of ETR
NCT04630002 (54) [back to overview]Cohort 2: Ctau of ETR
NCT04630002 (54) [back to overview]Cohort 2: Ctau of GSK3640254
NCT04630002 (54) [back to overview]Cohort 2: Tmax of ETR
NCT04630002 (54) [back to overview]Cohort 2: Tmax of GSK3640254
NCT04630002 (54) [back to overview]Cohort 3: AUC(0-tau) of GSK3640254
NCT04630002 (54) [back to overview]Cohort 3: Cmax of GSK3640254
NCT04630002 (54) [back to overview]Cohort 1: 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 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 Clinically Significant Abnormal ECG Findings
NCT04630002 (54) [back to overview]Cohort 2: Cmax of GSK3640254
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 2: Number of Participants With SAEs and Non-SAEs
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 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 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 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 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 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 Clinically Significant Abnormal ECG Findings
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 AEs Leading to Discontinuations and Deaths
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 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 2: Number of Participants With Vital Sign Values of PCI Criteria
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Hematology Parameters

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]

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]

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]

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 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 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: 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 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]

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: 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]

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: 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]

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]

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]

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]

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]

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: 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: 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]

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 (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 (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)
3rd TrimesterPostpartum
EFV 600mg q.d.55.458.3

[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: 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: 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]

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 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 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 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 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]

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

Mean Change From Baseline in CD4+ Cell Count

The mean change in CD4+ cell count from baseline was calculated with a last observation carried forward method; i.e. the last observed value was carried forward, irrespective of the reason for discontinuation. (NCT00458302)
Timeframe: at week 4, 12, 24, 36, 48, 60, 72, 84, 96, 112, 128, 144

,
Interventionnumber of cells/L (x10^6) (Mean)
week 4week 12week 24week 36week 48week 60week 72week 84week 96week 112week 128week 144
DRV/r-32.9-20.7-35.8-21.1-15.1-2.3-12.3-3.754.887.590.494.9
DRV/r+2NRTIs-16.9-23.6-5.4-1.2-19.0-4.024.134.649.1106.0117.399.3

[back to top]

Resistance Determinations

Number of patients with resistance mutations at any time point when a patient had a viral load > 50 copies/mL after randomization. (NCT00458302)
Timeframe: at each visit from baseline to week 144

,
Interventionnumber of participants (Number)
>= 1 HIV-1 RNA > 50 copies/mL>= 1 successful genotype after baseline>= 1 IAS-USA primary PI mutations>= 1 DRV RAMsNRTI RAMsM184V mutationno primary PI, DRV, NRTI or M184 V mutations
DRV/r4831110030
DRV/r+2NRTIs4223101122

[back to top]

Virological Response [Intent To Treat (ITT), TLOVR - All Switches Included, < 50 Copies/ml, Week 144]

Virological response is defined as the number of patients in the ITT population with a plasma viral load < 50 HIV RNA copies/ml at Week 144. Treatment failure was defined as two consecutive HIV RNA levels ≥ 50 copies/mL, or discontinuation of randomised treatment (known as TLOVR). All switches included means that all data even after any changes of treatment were kept. *Discontinuations and rechallenge with NRTIs are taken into account until start of Week 144 window. (NCT00458302)
Timeframe: Week 144

Interventionparticipants (Number)
DRV/r+2NRTIs106
DRV/r106

[back to top] [back to top]

Virological Response [Intent To Treat (ITT) - TLOVR, < 50 Copies/ml, Week 48]

Virological response is defined as the number of patients in the ITT population with a plasma viral load < 50 HIV RNA copies/ml at Week 48. Treatment failure was defined as two consecutive HIV RNA levels ≥ 50 copies/mL, or discontinuation of randomised treatment (known as TLOVR). In addition, any switch in background nucleoside reverse transcriptase inhibitors (NRTIs) equaled failure* (referred to as a Switch Equals Failure analysis). *Discontinuations and rechallenge with NRTIs are taken into account until start of Week 48 window (NCT00458302)
Timeframe: Week 48

Interventionparticipants (Number)
DRV/r+2NRTIs110
DRV/r107

[back to top]

Virological Response [Per Protocol (PP) - Time to Loss of Virologic Response (TLOVR), < 50 Copies/ml, Week 48]

Virological response is defined as the number of patients in the PP population with a plasma viral load < 50 HIV RNA copies/ml at Week 48. Treatment failure was defined as two consecutive HIV RNA levels ≥ 50 copies/mL, or discontinuation of randomised treatment (known as TLOVR). In addition, any switch in background nucleoside reverse transcriptase inhibitors (NRTIs) equaled failure* (referred to as a Switch Equals Failure analysis). *Discontinuations and rechallenge with NRTIs are taken into account until Week 48 (NCT00458302)
Timeframe: Week 48

Interventionparticipants (Number)
DRV/r+2NRTIs108
DRV/r106

[back to top]

Virological Response [Per Protocol (PP), TLOVR - Switch Equals Failure, < 50 Copies/ml, Week 144]

Virological response is defined as the number of patients in the PP population with a plasma viral load < 50 HIV RNA copies/ml at Week 144. Treatment failure was defined as two consecutive HIV RNA levels ≥ 50 copies/mL, or discontinuation of randomised treatment (known as TLOVR). In addition, any switch in background nucleoside reverse transcriptase inhibitors (NRTIs) equaled failure* (referred to as a Switch Equals Failure analysis). *Discontinuations and rechallenge with NRTIs are taken into account until Week 144 (NCT00458302)
Timeframe: Week 144

Interventionparticipants (Number)
DRV/r+2NRTIs94
DRV/r88

[back to top]

Virological Response [Per Protocol (PP), TLOVR - Switch Equals Failure, <200 Copies/ml, Week 144]

Virological response is defined as the number of patients in the PP population with a plasma viral load < 200 HIV RNA copies/ml at Week 144. Treatment failure was defined as two consecutive HIV RNA levels ≥ 50 copies/mL, or discontinuation of randomised treatment (known as TLOVR). In addition, any switch in background nucleoside reverse transcriptase inhibitors (NRTIs) equaled failure* (referred to as a Switch Equals Failure analysis). *Discontinuations and rechallenge with NRTIs are taken into account until Week 144 (NCT00458302)
Timeframe: week 144

InterventionParticipants (Number)
DRV/r+2NRTIs102
DRV/r95

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

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 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 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]

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]

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 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]

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 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]

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]

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 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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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 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]

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]

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 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]

Number of Participants With Plasma HIV-1 Viral Load < 50 Copies/ml

Number of participants with plasma HIV-1 Viral load < 50 copies/mL at study visit weeks 24, 48, and 96. Closest observed result between 20 and up to 30 weeks (for week 24), between 42 and up to 54 (for week 48), and between 90 and up to 110 (for week 96) used if multiple results available. Missing values excluded. (NCT00537394)
Timeframe: At Weeks 24, 48, 96

,
Interventionparticipants (Number)
Week 24: Number with RNA < 50 c/mL (N=170; N=171)Week 48: Number with RNA < 50 c/mL (N=169; N=165)Week 96: Number with RNA < 50 c/mL (N=158; N=158)
Add NRTIs (Randomized) to Individualized Regimen (cPSS > 2.0)122112107
Omit NRTIs (Randomized) From Individualized Regimen(cPSS > 2)117106109

[back to top]

Time From Randomization to Discontinuation of Randomized NRTI Component of Study Treatment

Discontinuation of Randomized NRTI component of Study Treatment is defined as permanently stopping all NRTIs among those randomized to add NRTIs, or starting any NRTI among those randomized to omit NRTIs. Subjects leaving the study for reasons other than death, relocation, incarceration, or site closure were reviewed for meeting this outcome by a blinded, independent panel. Additionally, any participant failing to start study treatment after randomization and prior to closure was also reviewed. Event times were scheduled study weeks when discontinuation events occurred. Censoring times were latest scheduled study visit weeks with evaluation. (NCT00537394)
Timeframe: From randomization to week 96 study visit

,
Interventionweeks (Number)
1st percentile5th percentile10th percentile
Add NRTIs (Randomized) to Individualized Regimen (cPSS > 2.0)036NA
Omit NRTIs (Randomized) From Individualized Regimen(cPSS > 2)03648

[back to top]

Time From Treatment Dispensation to First Grade 3 or Higher (and at Least One Grade Higher Than Baseline) Signs/Symptom or Laboratory Abnormality

Events following permanent discontinuation of NRTI assignment are excluded (i.e. censoring at time of this event, if applicable). Week 96 study visit could occur up to 110 weeks following randomization. Censoring time was the latest study visit when participant was evaluated or when NRTI assignment was discontinued (when applicable). Event time was the exact number of weeks following treatment initiation when the qualifying sign/symptom started (for those safety events triggered by a sign/symptom), or exact number of weeks following treatment initiation when specimen from qualifying laboratory result was drawn (for those safety events triggered by a laboratory abnormality). (NCT00537394)
Timeframe: From treatment dispensation to week 96 study visit

,
Interventionweeks (Number)
5th percentile25th percentile50th percentile
Add NRTIs (Randomized) to Individualized Regimen (cPSS > 2.0)3.124.7NA
Omit NRTIs (Randomized) From Individualized Regimen(cPSS > 2)3.925.397.7

[back to top]

Time From Treatment Dispensation to First Study ARV Modification (Excluding NRTIs, if Applicable)

First study ARV modification included any discontinuation or substitution of any chosen and initiated ARV for any reason. Events prompting study medication change could include protocol required (e.g. safety), protocol recommended but not required (e.g. virologic failure), or participant motivated (such as non-adherence, loss to follow-up or death; in other words, not protocol recommended or required). Event times were the exact weeks from treatment initiation to the time of qualifying regimen modification. Censoring times were the exact weeks from treatment initiation to the last date of study drugs. The week 96 (final study visit) could occur up through 110 weeks following randomization. (NCT00537394)
Timeframe: From treatment dispensation to week 96 study visit

,
Interventionweeks (Number)
5th percentile10th percentile25th percentile
Add NRTIs (Randomized) to Individualized Regimen (cPSS > 2.0)9.031.198.0
Omit NRTIs (Randomized) From Individualized Regimen(cPSS > 2)24.038.0NA

[back to top]

Time From Treatment Dispensation to Serious Non-AIDS-defining Events

Serious Non-AIDS defining Events were adjudicated by independent and blinded review and possible events included serious diagnoses in the following disease areas: liver, cardiovascular, end-stage renal, non-AIDS malignancy, and diabetes mellitus. Week 96 study visit could take place up to 110 weeks following randomization. Event times were the exact weeks following treatment initiation corresponding to the diagnosis dates of the qualifying serious non-AIDS defining events. Censoring times were the weeks following treatment initiation corresponding to the latest study visit. (NCT00537394)
Timeframe: From treatment initiation to week 96 study visit

,
Interventionweeks (Number)
1st percentile5th percentile
Add NRTIs (Randomized) to Individualized Regimen (cPSS > 2.0)4.960.0
Omit NRTIs (Randomized) From Individualized Regimen(cPSS > 2)29.3NA

[back to top]

Change in Summarized Quality of Life Score

Quality-of-life score at each evaluation based upon a single question assessing participants' self-report of general health with a range of 0 (representing worst health status) to 100 (representing perfect health). (NCT00537394)
Timeframe: At study entry and Weeks 24, 48, 96

,
Interventionunits on a scale (Median)
Change from baseline to week 24 (N=165; N=165)Change from baseline to week 48 (N=161; N=158)Change from baseline to week 96 (N=155; N=154)
Add NRTIs (Randomized) to Individualized Regimen (cPSS > 2.0)000
Omit NRTIs (Randomized) From Individualized Regimen(cPSS > 2)500.5

[back to top]

Number of Participants Self-reporting Non-adherence to Assigned Study ARVS (Excluding NRTIs, if Applicable)

Results represent self-report of non-adherence during the 4-day period prior to the outcome evaluation visit. Participants in follow-up for whom these data are missing for any reason are inferred as not-adherent. (NCT00537394)
Timeframe: At Weeks 24 and 48

,
Interventionparticipants (Number)
Week 24 (N=170; N=172)Week 48 (N=167; N=163)
Add NRTIs (Randomized) to Individualized Regimen (cPSS > 2.0)2530
Omit NRTIs (Randomized) From Individualized Regimen(cPSS > 2)2626

[back to top]

Time From Randomization to Confirmed Virological Failure

Virologic failure defined as confirmed (two consecutive) plasma HIV-1 RNA meeting 1 of the following 4 criteria: < 1.0 log10 copies/mL reduction from baseline level and >= 200 copies/mL at or after week 12 evaluation; >= 200 copies/mL after 1 measurement < 200 copies/mL; absence of any values < 200 copies/mL by and including week 24 evaluation; >= 200 copies/mL at week 48 evaluation. Event time was the scheduled study visit week when the initial plasma HIV-1 RNA specimen meeting the failure definition was collected. Censoring time was the latest scheduled study visit week when a plasma HIV-1 RNA specimen was collected and tested. (NCT00537394)
Timeframe: From randomization to week 96 study visit

,
Interventionweeks (Number)
5th percentile10th percentile25th percentile
Add NRTIs (Randomized) to Individualized Regimen (cPSS > 2.0)121248
Omit NRTIs (Randomized) From Individualized Regimen(cPSS > 2)121248

[back to top]

Change in Plasma HIV-1 Viral Load From Baseline to Week 1

Method of Kaplan and Meier used to accommodate left-censoring for those whose week 1 levels < 50 copies/mL. (NCT00537394)
Timeframe: From baseline to Week 1 evaluation

Interventionlog10 copies/mL (Median)
Add NRTIs (Randomized) to Individualized Regimen (cPSS > 2.0)1.3
Omit NRTIs (Randomized) From Individualized Regimen(cPSS > 2)1.4

[back to top]

Number of Participants With Change in Virus Co-receptor Tropism Among Those With R5-only Tropic Virus at Study Entry

HIV Co-receptor tropism test result of either dual/mixed or evidence of X4 using virus from sample collected at confirmed virologic failure. (NCT00537394)
Timeframe: From study entry to time of confirmed virological failure (up to 96 weeks)

Interventionparticipants (Number)
Add NRTIs (Randomized) to Individualized Regimen (cPSS > 2.0)3
Omit NRTIs (Randomized) From Individualized Regimen(cPSS > 2)2

[back to top]

Participants With Newly Acquired HIV Drug Resistance Between Study Entry and Confirmed Virologic Failure

Defined among the subgroup of participants experiencing the outcome of confirmed virologic failure. Newly acquired HIV drug resistance is defined as one or more ARVs with partial resistance or resistance when pre-entry resistance was fully sensitive or resistant when pre-entry resistance was fully sensitive or partially sensitive. The ARVs included for resistance acquisition included the following: darunavir/ritonavir; etravirine, tipranavir, tenofovir, emtracitabine, lamivudine, zidovudine, abacavir. (NCT00537394)
Timeframe: Between baseline and confirmed virologic failure (up to 96 weeks)

Interventionparticipants (Number)
Add NRTIs (Randomized) to Individualized Regimen (cPSS > 2.0)18
Omit NRTIs (Randomized) From Individualized Regimen(cPSS > 2)13

[back to top]

Percent of Participants With Regimen Failure, Defined as a Confirmed Virologic Failure or Discontinuation of Randomized NRTI Component of Study Treatment

Virologic failure defined as confirmed plasma HIV-1 RNA meeting 1 of the following 4 criteria: < 1.0 log10 copies/mL reduction from baseline level and >= 200 copies/mL at or after week 12 evaluation; >= 200 copies/mL after 1 measurement < 200 copies/mL; absence of any values < 200 copies/mL by and including week 24 evaluation; >= 200 copies/mL at week 48 evaluation. Discontinuation of Randomized NRTI component of Study Treatment is defined as permanently stopping all NRTIs among those randomized to add NRTIs, or starting any NRTI among those randomized to omit NRTIs. Subjects leaving the study for reasons other than death, relocation, incarceration, or site closure were reviewed for the discontinuation outcome by a blinded, independent panel. Additionally, any participant failing to start study treatment after randomization and prior to closure was also reviewed. Results report percent of participants reaching regimen failure outcome by week 48 evaluation using Kaplan-Meier method. (NCT00537394)
Timeframe: From study entry to end of Week 48 evaluation window

Interventionpercentage of participants (Number)
Add NRTIs (Randomized) to Individualized Regimen (cPSS > 2.0)26.0
Omit NRTIs (Randomized) From Individualized Regimen(cPSS > 2)29.8

[back to top]

Change in Cardiovascular Risk Score From Baseline

Cardiovascular risk score defined by Framingham providing an estimate of the probability of developing cardiovascular disease over the next 10-year period. Persons with a historical cardiovascular event (CAD, cerebro- or peripheral- vascular disorder, MI or stroke), were excluded, and scores were not calculated at follow-up times after individuals had a cardiovascular event. Missing values for input data (e.g. smoking status) resulted in a missing value for Framingham score. (NCT00537394)
Timeframe: At Weeks 24, 48, and 96

,
Interventionunits on a scale (Mean)
Week 24 (N= 150; N=147)Week 48 (N=143; N=144)Week 96 (N=129; N=132)
Add NRTIs (Randomized) to Individualized Regimen (cPSS > 2.0)-0.70.10.5
Omit NRTIs (Randomized) From Individualized Regimen(cPSS > 2)0.30.81.1

[back to top]

Change in CD4 Count From Baseline

Baseline CD4 calculated as average of pre-entry and entry values. Closest observed result between 42 and up to 54 weeks (for week 48) or between 90 and up to 110 weeks (for week 96), used if multiple results available. Missing values excluded. (NCT00537394)
Timeframe: From study entry to Weeks 48 and 96

,
Interventioncells/mm^3 (Median)
Change from entry to week 48 (N=166; N=163)Change from entry to week 96 (N= 154; N=157)
Add NRTIs (Randomized) to Individualized Regimen (cPSS > 2.0)105.5140.8
Omit NRTIs (Randomized) From Individualized Regimen(cPSS > 2)89.5115.5

[back to top]

Change in Fasting Non-HDL Cholesterol From Baseline

Fasting non-HDL cholesterol calculated from difference between fasting total cholesterol and fasting HDL level. Missing values and non-fasting values excluded. (NCT00537394)
Timeframe: From study entry to Weeks 24, 48

,
Interventionmg/dL (Mean)
Change from baseline to week 24 (N=131; N=121)Change from baseline to week 48 (N=125 ; N=117)
Add NRTIs (Randomized) to Individualized Regimen (cPSS > 2.0)4.17.6
Omit NRTIs (Randomized) From Individualized Regimen(cPSS > 2)20.819.8

[back to top]

Economic Impact of a Substitution of Dual Boosted PIs With DRV/r

To assess the economic impact of DRV/r substitution for dual boosted PIs, we compared the average wholesale acquisition costs for the drugs in US Dollars ($) per month. The wholesale acquisition cost in US dollars ($) for each ART regimen was determined and the difference between the cost for the experimental and control groups was calculated and reported as US dollar savings per month. (NCT00543101)
Timeframe: 48 weeks

InterventionUS dollars savings per month (Number)
Switch to DRV/r217
Continue on Current Dual Boosted PI0

[back to top]

The Percentage of Participants With Successful Virologic Suppression

Amount of HIV RNA copies per ml blood collected from subjects as measured by the Ultra-sensitive HIV-1 PCR (Roche Cobas). Successful virologic suppression is defined as < 50 copies/ml blood. The result is the percentage of participants with successful virologic suppression. (NCT00543101)
Timeframe: 24 weeks

InterventionPercentage of Participants (Number)
Switch to DRV/r100
Continue on Current Dual Boosted PI100

[back to top]

Lipid Fraction Results, Mean of the Change From Baseline to Week 24.

We collected fasting total cholesterol, LDL-cholesterol, HDL-cholesterol and triglycerides from all participants in both arms of the study. We calculated the differences between the values at week 24 and baseline for the participants in both arms. We reported the mean of the change from baseline to week 24. (NCT00543101)
Timeframe: baseline and 24 weeks

,
Interventionmg/dL (Number)
Change in Total CholesterolChange in LDL-CholesterolChange in HDL-CholesterolChange in Triglycerides
Continue on Current Dual Boosted PI2210
Switch to DRV/r-6-4-1-13

[back to top]

Treatment Satisfaction (+3, Much More Satisfied Now to -3, Much Less Satisfied Now)

Participants in the experimental arm completed treatment satisfaction questionnaires at 24 weeks, and the control arm at 48 weeks (24 weeks after mid-study crossover to boosted darunavir). The questionnaires used numeric satisfaction scales (+3 much more satisfied now to -3 much less satisfied now). We reported the median and ranges for each question for each study arm. (NCT00543101)
Timeframe: 24 weeks

,
InterventionUnits on a Scale (+3 to -3) (Median)
How satisfied are your with your current treatmentHow satisfied are you with any side-effectsHow convenient is your treatmentHow well does your treatment fit your lifestyleWould you recommend your treatment to someone elseHow likely are you to continue your treatment
Crossover Week 482.52.5332.52.5
Switch to DRV/r323333

[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 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]

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 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]

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 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]

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]

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]

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]

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 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]

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 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 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 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 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 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 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 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]

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 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 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]

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 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]

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 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 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 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]

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]

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]

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]

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 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]

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]

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]

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]

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]

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]

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]

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 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]

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 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]

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]

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]

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]

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]

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]

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 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]

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]

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]

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]

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 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]

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]

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]

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]

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 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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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 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]

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]

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]

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]

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]

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]

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]

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 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]

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 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]

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]

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]

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]

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]

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 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]

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]

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]

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 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]

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]

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]

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 Neurocognitive Impairment at Baseline

(NCT00855413)
Timeframe: Week 2 or 4

InterventionParticipants (Count of Participants)
Acute HIV Infection Treatment Group8

[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]

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]

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]

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]

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 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 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]

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 Week 48

(NCT00855413)
Timeframe: Week 48

InterventionParticipants (Count of Participants)
Acute HIV Infection Treatment Group3

[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 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]

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]

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]

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] [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]

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]

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]

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]

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]

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]

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]

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]

Cmax of Rosuvastatin

(NCT00885495)
Timeframe: 7 days

Interventionng/mL (Geometric Mean)
Rosuvastatin6.70
Rosuvastatin-Darunavir-Ritonavir16.32

[back to top]

AUC of Rosuvastatin

(NCT00885495)
Timeframe: 7 days

Interventionng*hr/mL (Geometric Mean)
Rosuvastatin108.96
Rosuvastatin-Darunavir-Ritonavir161.24

[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]

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]

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 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]

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]

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 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]

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]

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]

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] [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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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 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 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]

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]

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 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]

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 16

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 16

Interventionlog10 Copies/mL (Median)
ETR + DRV/Rtv-2.29

[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]

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]

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]

Geometric Mean of t1/2, of Raltegravir (RAL) at a Dose of 400 mg Twice a Day Plus Darunavir/Ritonavir (DRV/RTV) at a Dose of 800/100 mg Once a Day in HIV-1-infected Patients Were Mesured After 15 Days of Therapy

Geometric mean of t1/2 of raltegravir (RAL) at a dose of 400 mg twice a day plus darunavir/ritonavir (DRV/RTV) at a dose of 800/100 mg once a day in HIV-1-infected patients were mesured after 15 days of therapy. The treating physician chose an NRTI-Geometric mean of t1/2, of raltegravir (RAL) at a dose of 400 mg twice a day plus darunavir/ritonavir (DRV/RTV) at a dose of 800/100 mg once a day in HIV-1-infected patients were mesured after 15 days of therapy.sparing regimen because of toxicity or resistance mutations to NRTIs, which included DRV/RTV 800/100 mg once daily plus RAL 400 mg twice daily. All patients were RAL and DRV naive and had no evidence of protease inhibitor mutations. (NCT01258374)
Timeframe: After at least 15 days on therapy, patients were admitted for a 24-hour PK study. The moorning dose of RAL adn DRV/r was administered in the clinic with. Blood samples were drawn immediatly before breakfast and 0.5, 1,2,3,4,6,8,12 and 24 hours afterward.

Interventionh (Geometric Mean)
t1/2 Darunavirt1/2 Raltegravirt1/2 Ritonavir
Single Arm With Dual Therapy10.912.689.48

[back to top]

Geometric Mean of C-max, of Raltegravir (RAL) at a Dose of 400 mg Twice a Day Plus Darunavir/Ritonavir (DRV/RTV) at a Dose of 800/100 mg Once a Day in HIV-1-infected Patients Were Mesured After 15 Days of Therapy.

Geometric mean of C-max of raltegravir (RAL) at a dose of 400 mg twice a day plus darunavir/ritonavir (DRV/RTV) at a dose of 800/100 mg once a day in HIV-1-infected patients were mesured after 15 days of therapy. The treating physician chose an NRTI-Geometric mean of C-max, of raltegravir (RAL) at a dose of 400 mg twice a day plus darunavir/ritonavir (DRV/RTV) at a dose of 800/100 mg once a day in HIV-1-infected patients were mesured after 15 days of therapy.sparing regimen because of toxicity or resistance mutations to NRTIs, which included DRV/RTV 800/100 mg once daily plus RAL 400 mg twice daily. All patients were RAL and DRV naive and had no evidence of protease inhibitor mutations. (NCT01258374)
Timeframe: After at least 15 days on therapy, patients were admitted for a 24-hour PK study. The moorning dose of RAL adn DRV/r was administered in the clinic with. Blood samples were drawn immediatly before breakfast and 0.5, 1,2,3,4,6,8,12 and 24 hours afterward.

Interventionng/mL (Geometric Mean)
Cmax DarunavirCmax RaltegravirCmax Ritonavir
Single Arm With Dual Therapy7630970490

[back to top]

Geometric Mean of AUC, of Raltegravir (RAL) at a Dose of 400 mg Twice a Day Plus Darunavir/Ritonavir (DRV/RTV) at a Dose of 800/100 mg Once a Day in HIV-1-infected Patients Were Mesured After 15 Days of Therapy.

Geometric mean of AUC0 of raltegravir (RAL) at a dose of 400 mg twice a day plus darunavir/ritonavir (DRV/RTV) at a dose of 800/100 mg once a day in HIV-1-infected patients were mesured after 15 days of therapy. The treating physician chose an NRTI-Geometric mean of C-Trough, of raltegravir (RAL) at a dose of 400 mg twice a day plus darunavir/ritonavir (DRV/RTV) at a dose of 800/100 mg once a day in HIV-1-infected patients were mesured after 15 days of therapy.sparing regimen because of toxicity or resistance mutations to NRTIs, which included DRV/RTV 800/100 mg once daily plus RAL 400 mg twice daily. All patients were RAL and DRV naive and had no evidence of protease inhibitor mutations. (NCT01258374)
Timeframe: After at least 15 days on therapy, patients were admitted for a 24-hour PK study. The moorning dose of RAL adn DRV/r was administered in the clinic with. Blood samples were drawn immediatly before breakfast and 0.5, 1,2,3,4,6,8,12 and 24 hours afterward.

Interventionng.h/mL (Geometric Mean)
AUC RaltegravirAUC DarunavirAUC Ritonavir
Single Arm With Dual Therapy305068,7305470

[back to top]

Geometric Mean of C-Trough, of Raltegravir (RAL) at a Dose of 400 mg Twice a Day Plus Darunavir/Ritonavir (DRV/RTV) at a Dose of 800/100 mg Once a Day in HIV-1-infected Patients Were Mesured After 15 Days of Therapy.

Geometric mean of C-Trough, of raltegravir (RAL) at a dose of 400 mg twice a day plus darunavir/ritonavir (DRV/RTV) at a dose of 800/100 mg once a day in HIV-1-infected patients were mesured after 15 days of therapy.The treating physician chose an NRTI-sparing regimen because of toxicity or resistance mutations to NRTIs, which included DRV/RTV 800/100 mg once daily plus RAL 400 mg twice daily. All patients were RAL and DRV naive and had no evidence of protease inhibitor mutations. (NCT01258374)
Timeframe: After at least 15 days on therapy, patients were admitted for a 24-hour PK study. The moorning dose of RAL adn DRV/r was administered in the clinic with. Blood samples were drawn immediatly before breakfast and 0.5, 1,2,3,4,6,8,12 and 24 hours afterward.

Interventionng/ml (Geometric Mean)
C trough DarunavirC trough RaltegravirC trough Ritonavir
Single Arm With Dual Therapy13304090

[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] [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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

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 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] [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]

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]

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]

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 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]

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]

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 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]

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]

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]

The Ratio of the Maximum Plasma Concentration of Maraviroc Between 20 and 10 Day

On day 10 of the study the maximum concentractions of maraviroc will be measured . On day 20 of the study the maximum plasma concentractions maraviroc will be measured . (NCT01348763)
Timeframe: 10 day, 20 days

Interventionratio (Mean)
Truvada, Darunavir/r and Maraviroc0.99

[back to top]

Number of Participants With Changes in Haematology and Biochemistry Laboratory Tests

Haematology and biochemistry laboratory tests such as full blood count, elelectrolytes and lipids will be measured to assess for changes. (NCT01348763)
Timeframe: 35 days

InterventionParticipants (Count of Participants)
Truvada, Darunavir/r and Maraviroc0

[back to top]

Plasma Pharmacokinetics of COBI: Ctau (Cohort 1)

Ctau was analyzed for Cohort 1 (treatment-naive) and was defined as the observed drug concentration at the end of the dosing interval. (NCT01363011)
Timeframe: Blood samples were collected at 0 (predose), 0.5, 1.0, 2.0, 3.0, 4.0, 5.0, 8.0, 12.0, and 24.0 hours postdose at baseline and Weeks 2, 4, and 24.

Interventionng/mL (Number)
Week 2Week 4Week 24
E/C/F/TDF (Cohort 1)150.537.324.2

[back to top]

Plasma Pharmacokinetics of COBI: Ctau (Cohort 2)

Ctau was analyzed for Cohort 2 (treatment-experienced) and was defined as the observed drug concentration at the end of the dosing interval. (NCT01363011)
Timeframe: Blood samples were collected at 0 (predose), 0.5, 1.0, 2.0, 3.0, 4.0, 5.0, 8.0, 12.0, and 24.0 hours postdose at baseline and Weeks 2, 4, and 24.

Interventionng/mL (Mean)
Week 2 (n = 13)Week 4 (n = 13)Week 24 (n = 11)
COBI+PI+2 NRTIs (Cohort 2)79.971.3139.8

[back to top]

Plasma Pharmacokinetics of COBI: t1/2 (Cohort 1)

t1/2 was analyzed for Cohort 1 (treatment-naive) and was defined as the estimate of the terminal elimination half-life of the drug. (NCT01363011)
Timeframe: Blood samples were collected at 0 (predose), 0.5, 1.0, 2.0, 3.0, 4.0, 5.0, 8.0, 12.0, and 24.0 hours postdose at baseline and Weeks 2, 4, and 24.

Interventionhours (Number)
Week 2Week 4Week 24
E/C/F/TDF (Cohort 1)6.143.573.63

[back to top]

Plasma Pharmacokinetics of COBI: t1/2 (Cohort 2)

t1/2 was analyzed for Cohort 2 (treatment-experienced) and was defined as the estimate of the terminal elimination half-life of the drug. (NCT01363011)
Timeframe: Blood samples were collected at 0 (predose), 0.5, 1.0, 2.0, 3.0, 4.0, 5.0, 8.0, 12.0, and 24.0 hours postdose at baseline and Weeks 2, 4, and 24.

Interventionhours (Median)
Week 2 (n = 13)Week 4 (n = 12)Week 24 (n = 10)
COBI+PI+2 NRTIs (Cohort 2)4.373.983.77

[back to top]

Plasma Pharmacokinetics of COBI: Tmax (Cohort 2)

Tmax was analyzed for Cohort 2 (treatment-experienced) and was defined as the time of Cmax. (NCT01363011)
Timeframe: Blood samples were collected at 0 (predose), 0.5, 1.0, 2.0, 3.0, 4.0, 5.0, 8.0, 12.0, and 24.0 hours postdose at baseline and Weeks 2, 4, and 24.

Interventionhours (Median)
Week 2 (n = 13)Week 4 (n = 13)Week 24 (n = 11)
COBI+PI+2 NRTIs (Cohort 2)3.924.923.00

[back to top]

Change From Baseline in eGFR Using the Modification of Diet in Renal (MDRD) Equation at Week 24 (Cohort 1)

Change from baseline in eGFR-MDRD equation at Week 24 was analyzed in Cohort 1 (treatment-naive). The calculation was normalized to 1.73 m^2 body surface area. (NCT01363011)
Timeframe: Baseline; Week 24

InterventionmL/min/1.73 m^2 (Median)
Baseline (n = 33)Change at Week 24 (n = 30)
E/C/F/TDF (Cohort 1)77.1-7.4

[back to top]

Plasma Pharmacokinetics of COBI: Tmax (Cohort 1)

Tmax was analyzed for Cohort 1 (treatment-naive) and was defined as the time of Cmax. (NCT01363011)
Timeframe: Blood samples were collected at 0 (predose), 0.5, 1.0, 2.0, 3.0, 4.0, 5.0, 8.0, 12.0, and 24.0 hours postdose at baseline and Weeks 2, 4, and 24.

Interventionhours (Number)
Week 2Week 4Week 24
E/C/F/TDF (Cohort 1)4.002.004.00

[back to top]

Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 24 (Cohort 1)

The percentage of participants with HIV-1 RNA < 50 copies/mL at Week 24 was analyzed in Cohort 1 (treatment-naive) using the FDA snapshot analysis algorithm. (NCT01363011)
Timeframe: Week 24

Interventionpercentage of participants (Number)
E/C/F/TDF (Cohort 1)84.8

[back to top]

Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 24 (Cohort 2)

The percentage of participants with HIV-1 RNA < 50 copies/mL at Week 24 was analyzed in Cohort 2 (treatment-experienced) using the FDA snapshot analysis algorithm. (NCT01363011)
Timeframe: Week 24

Interventionpercentage of participants (Number)
COBI+PI+2 NRTIs (Cohort 2)90.4

[back to top]

Change From Baseline in Actual Glomerular Filtration Rate (aGFR) at Weeks 2, 4, and 24 (Cohort 1)

Change from baseline in aGFR at Weeks 2, 4, and 24 was analyzed in Cohort 1 (treatment-naive). aGFR was calculated using iohexol plasma clearance. (NCT01363011)
Timeframe: Baseline; Weeks 2, 4, and 24

InterventionmL/min (Number)
BaselineChange at Week 2Change at Week 4Change at Week 24
E/C/F/TDF (Cohort 1)81.6-12.1-7.3-3.3

[back to top]

Change From Baseline in aGFR at Weeks 2, 4, and 24 (Cohort 2)

Change from baseline in aGFR at Weeks 2, 4, and 24 was analyzed in Cohort 2 (treatment-experienced). aGFR was calculated using iohexol plasma clearance. (NCT01363011)
Timeframe: Baseline; Weeks 2, 4, and 24

InterventionmL/min (Median)
BaselineChange at Week 2 (n=13)Change at Week 4 (n=13)Change at Week 24 (n=11)
COBI+PI+2 NRTIs (Cohort 2)82.51.67.0-4.1

[back to top]

Change From Baseline in eGFR Using the Chronic Kidney Disease, Epidemiology Collaboration (CKD-EPI) Formula Based on Cystatin C Equation at Week 24 (Cohort 1)

Change from baseline in eGFR-CKD-EPI based on cystatin C equation (not adjusted for age, sex, and race) at Week 24 was analyzed in Cohort 1 (treatment-naive). The calculation was normalized to 1.73 m^2 body surface area. (NCT01363011)
Timeframe: Baseline; Week 24

InterventionmL/min/1.73 m^2 (Median)
Baseline (n = 33)Change at Week 24 (n = 30)
E/C/F/TDF (Cohort 1)77.60.3

[back to top]

Change From Baseline in eGFR-CG at Week 24 (Cohort 2)

Change from baseline in eGFR-CG equation at Week 24 was analyzed in Cohort 2 (treatment-experienced). (NCT01363011)
Timeframe: Baseline; Week 24

InterventionmL/min (Median)
Baseline (n = 73)Change at Week 24 (n = 67)
COBI+PI+2 NRTIs (Cohort 2)71.4-3.7

[back to top]

Change From Baseline in eGFR-CG at Weeks 48 and 96 (Cohort 1)

Change from baseline in eGFR-CG at Weeks 48 and 96 were analyzed in Cohort 1 (treatment-naive). This outcome is to measure the long-term effect of COBI-containing regimens on renal parameters. (NCT01363011)
Timeframe: Baseline; Weeks 48 and 96

InterventionmL/min (Median)
Change at Week 48 (n = 28)Change at Week 96 (n = 25)
E/C/F/TDF (Cohort 1)-7.6-7.9

[back to top]

Change From Baseline in eGFR-CG at Weeks 48 and 96 (Cohort 2)

Change from baseline in eGFR-CG at Weeks 48 and 96 were analyzed in Cohort 2 (treatment-experienced). This outcome is to measure the long-term effect of COBI-containing regimens on renal parameters. (NCT01363011)
Timeframe: Baseline; Week 48

InterventionmL/min (Median)
Change at Week 48 (n = 63)Change at Week 96 (n = 50)
COBI+PI+2 NRTIs (Cohort 2)-3.8-5.0

[back to top]

Change From Baseline in eGFR-CKD-EPI Based on Cystatin C Equation (Adjusted) at Weeks 48 and 96 (Cohort 1)

Change from baseline in eGFR-CKD-EPI based on cystatin C equation (adjusted for age, sex, and race) at Weeks 48 and 96 were analyzed in Cohort 1 (treatment-naive). The calculation was normalized to 1.73 m^2 body surface area. This outcome is to measure the long-term effect of COBI-containing regimens on renal parameters. (NCT01363011)
Timeframe: Baseline; Weeks 48 and 96

InterventionmL/min/1.73 m^2 (Median)
Change at Week 48 (n = 28)Change at Week 96 (n = 25)
E/C/F/TDF (Cohort 1)1.612.6

[back to top]

Change From Baseline in eGFR-CKD-EPI Based on Cystatin C Equation (Adjusted) at Weeks 48 and 96 (Cohort 2)

Change from baseline in eGFR-CKD-EPI based on cystatin C equation (adjusted for age, sex, and race) at Weeks 48 and 96 were analyzed in Cohort 2 (treatment-experienced). The calculation was normalized to 1.73 m^2 body surface area. This outcome is to measure the long-term effect of COBI-containing regimens on renal parameters. (NCT01363011)
Timeframe: Baseline; Weeks 48 and 96

InterventionmL/min/1.73 m^2 (Median)
Change at Week 48 (n = 63)Change at Week 96 (n = 50)
COBI+PI+2 NRTIs (Cohort 2)-4.7-2.8

[back to top]

Change From Baseline in eGFR-CKD-EPI Based on Cystatin C Equation at Weeks 48 and 96 (Cohort 1)

Change from baseline in eGFR-CKD-EPI based on cystatin C equation (not adjusted for age, sex, and race) at Weeks 48 and 96 were analyzed in Cohort 1 (treatment-naive). The calculation was normalized to 1.73 m^2 body surface area. This outcome is to measure the long-term effect of COBI-containing regimens on renal parameters. (NCT01363011)
Timeframe: Baseline; Weeks 48 and 96

InterventionmL/min/1.73 m^2 (Median)
Change at Week 48 (n = 28)Change at Week 96 (n = 25)
E/C/F/TDF (Cohort 1)1.912.4

[back to top]

Change From Baseline in eGFR-CKD-EPI Based on Cystatin C Equation at Weeks 48 and 96 (Cohort 2)

Change from baseline in eGFR-CKD-EPI based on cystatin C equation (not adjusted for age, sex, and race) at Weeks 48 and 96 were analyzed in Cohort 2 (treatment-experienced). The calculation was normalized to 1.73 m^2 body surface area. This outcome is to measure the long-term effect of COBI-containing regimens on renal parameters. (NCT01363011)
Timeframe: Baseline; Weeks 48 and 96

InterventionmL/min/1.73 m^2 (Median)
Change at Week 48 (n = 63)Change at Week 96 (n = 50)
COBI+PI+2 NRTIs (Cohort 2)-4.7-2.4

[back to top]

Change From Baseline in eGFR-CKD-EPI Based on Cystatin C Equation, Adjusted at Week 24 (Cohort 1)

Change from baseline in eGFR-CKD-EPI based on cystatin C equation (adjusted for age, sex, and race) at Week 24 was analyzed in Cohort 1 (treatment-naive). The calculation was normalized to 1.73 m^2 body surface area. (NCT01363011)
Timeframe: Baseline; Week 24

InterventionmL/min/1.73 m^2 (Median)
Baseline (n = 33)Change at Week 24 (n = 30)
E/C/F/TDF (Cohort 1)76.90.3

[back to top]

Change From Baseline in eGFR-CKD-EPI Based on Cystatin C Equation, Adjusted at Week 24 (Cohort 2)

Change from baseline in eGFR-CKD-EPI based on cystatin C equation (adjusted for age, sex, and race) at Week 24 was analyzed in Cohort 2 (treatment-experienced). The calculation was normalized to 1.73 m^2 body surface area. (NCT01363011)
Timeframe: Baseline; Week 24

InterventionmL/min/1.73 m^2 (Median)
Baseline (n = 73)Change at Week 24 (n = 67)
COBI+PI+2 NRTIs (Cohort 2)78.2-2.8

[back to top]

Change From Baseline in eGFR-CKD-EPI Formula Based on Cystatin C Equation at Week 24 (Cohort 2)

Change from baseline in eGFR-CKD-EPI based on cystatin C equation (not adjusted for age, sex, and race) at Week 24 was analyzed in Cohort 2 (treatment-experienced). The calculation was normalized to 1.73 m^2 body surface area. (NCT01363011)
Timeframe: Baseline; Week 24

InterventionmL/min/1.73 m^2 (Median)
Baseline (n = 73)Change at Week 24 (n = 67)
COBI+PI+2 NRTIs (Cohort 2)78.6-2.7

[back to top]

Change From Baseline in eGFR-MDRD at Week 24 (Cohort 2)

Change from baseline in eGFR-MDRD equation at Week 24 was analyzed in Cohort 2 (treatment-experienced). The calculation was normalized to 1.73 m^2 body surface area. (NCT01363011)
Timeframe: Baseline; Week 24

InterventionmL/min/1.73 m^2 (Median)
Baseline (n = 73)Change at Week 24 (n = 67)
COBI+PI+2 NRTIs (Cohort 2)65.8-3.4

[back to top]

Change From Baseline in eGFR-MDRD at Weeks 48 and 96 (Cohort 1)

Change from baseline in eGFR-MDRD at Weeks 48 and 96 were analyzed in Cohort 1 (treatment-naive). The calculation was normalized to 1.73 m^2 body surface area. This outcome is to measure the long-term effect of COBI-containing regimens on renal parameters. (NCT01363011)
Timeframe: Baseline; Weeks 48 and 96

InterventionmL/min/1.73 m^2 (Median)
Change at Week 48 (n = 28)Change at Week 96 (n = 25)
E/C/F/TDF (Cohort 1)-12.1-12.9

[back to top]

Change From Baseline in eGFR-MDRD at Weeks 48 and 96 (Cohort 2)

Change from baseline in eGFR-MDRD at Weeks 48 and 96 were analyzed in Cohort 2 (treatment-experienced). The calculation was normalized to 1.73 m^2 body surface area. This outcome is to measure the long-term effect of COBI-containing regimens on renal parameters. (NCT01363011)
Timeframe: Baseline; Weeks 48 and 96

InterventionmL/min/1.73 m^2 (Median)
Change at Week 48 (n = 63)Change at Week 96 (n = 50)
COBI+PI+2 NRTIs (Cohort 2)-3.9-2.8

[back to top]

Change From Baseline in Estimated Glomerular Filtration Rate (eGFR) Using the Cockcroft-Gault (CG) Equation at Week 24 (Cohort 1)

Change from baseline in eGFR-CG equation at Week 24 was analyzed in Cohort 1 (treatment-naive). (NCT01363011)
Timeframe: Baseline; Week 24

InterventionmL/min (Median)
Baseline (n = 33)Change at Week 24 (n = 30)
E/C/F/TDF (Cohort 1)72.9-5.2

[back to top]

Percentage of Participants Who Experienced Adverse Events (Cohort 1)

Adverse events (AEs) occurring from baseline up to 30 days following the last dose of study drug were summarized for Cohort 1 (treatment-naive). A participant was counted once if they had a qualifying event. (NCT01363011)
Timeframe: Up to 147 weeks plus 30 days

Interventionpercentage of participants (Number)
Any AEDrug-related AEGrade 3 or higher AEAE leading to drug discontinuationSerious AEAE of proximal renal tubulopathy
E/C/F/TDF (Cohort 1)100.048.521.212.118.20

[back to top]

Percentage of Participants Who Experienced Adverse Events (Cohort 2)

Adverse events (AEs) occurring from baseline up to 30 days following the last dose of study drug were summarized for Cohort 2 (treatment-experienced). A participant was counted once if they had a qualifying event. (NCT01363011)
Timeframe: Up to 166 weeks plus 30 days

Interventionpercentage of participants (Number)
Any AEDrug-related AEGrade 3 or higher AEAE leading to drug discontinuationSerious AEAE of proximal renal tubulopathy
COBI+PI+2 NRTIs (Cohort 2)93.227.428.811.015.10

[back to top]

Percentage of Participants Who Experienced Graded Laboratory Abnormalities (Cohort 1)

Laboratory abnormalities were summarized for Cohort 1 (treatment-naive) and were defined as values that increased at least one toxicity grade from baseline at any time postbaseline up to and including the date of last dose of study drug plus 30 days. A participant was counted once if they had a qualifying event. (NCT01363011)
Timeframe: Up to 147 weeks plus 30 days

Interventionpercentage of participants (Number)
Any laboratory abnormalityGrade 3 or 4 laboratory abnormality
E/C/F/TDF (Cohort 1)100.039.4

[back to top]

Percentage of Participants Who Experienced Graded Laboratory Abnormalities (Cohort 2)

Laboratory abnormalities were summarized for Cohort 2 (treatment-experienced) and were defined as values that increased at least one toxicity grade from baseline at any time postbaseline up to and including the date of last dose of study drug plus 30 days. A participant was counted once if they had a qualifying event. (NCT01363011)
Timeframe: Up to 166 weeks plus 30 days

Interventionpercentage of participants (Number)
Any laboratory abnormalityGrade 3 or 4 laboratory abnormality
COBI+PI+2 NRTIs (Cohort 2)100.0050.0

[back to top]

Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Weeks 48 and 96 (Cohort 1)

The percentage of participants with HIV-1 RNA < 50 copies/mL at Weeks 48 and 96 were analyzed in Cohort 1 (treatment-naive) using the FDA snapshot analysis algorithm. (NCT01363011)
Timeframe: Weeks 48 and 96

Interventionpercentage of participants (Number)
Week 48 (n = 33)Week 96 (n = 27)
E/C/F/TDF (Cohort 1)78.888.9

[back to top]

Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Weeks 48 and 96 (Cohort 2)

The percentage of participants with HIV-1 RNA < 50 copies/mL at Weeks 48 and 96 were analyzed in Cohort 2 (treatment-experienced) using the FDA snapshot analysis algorithm. (NCT01363011)
Timeframe: Weeks 48 and 96

Interventionpercentage of participants (Number)
Week 48 (n = 73)Week 96 (n = 54)
COBI+PI+2 NRTIs (Cohort 2)82.290.7

[back to top]

Plasma Pharmacokinetics of COBI: AUCtau (Cohort 1)

AUCtau was analyzed for Cohort 1 (treatment-naive) and was defined as the concentration of drug over time (area under the plasma concentration versus time curve over the dosing interval). (NCT01363011)
Timeframe: Blood samples were collected at 0 (predose), 0.5, 1.0, 2.0, 3.0, 4.0, 5.0, 8.0, 12.0, and 24.0 hours postdose at baseline and Weeks 2, 4, and 24.

Interventionh*ng/mL (Number)
Week 2Week 4Week 24
E/C/F/TDF (Cohort 1)16554.712704.19799.7

[back to top]

Plasma Pharmacokinetics of COBI: AUCtau (Cohort 2)

AUCtau was analyzed for Cohort 2 (treatment-experienced) and was defined as the concentration of drug over time (area under the plasma concentration versus time curve over the dosing interval). (NCT01363011)
Timeframe: Blood samples were collected at 0 (predose), 0.5, 1.0, 2.0, 3.0, 4.0, 5.0, 8.0, 12.0, and 24.0 hours postdose at baseline and Weeks 2, 4, and 24.

Interventionh*ng/mL (Mean)
Week 2 (n = 13)Week 4 (n = 13)Week 24 (n = 11)
COBI+PI+2 NRTIs (Cohort 2)12458.011165.313980.5

[back to top]

Plasma Pharmacokinetics of COBI: Cmax (Cohort 1)

Cmax was analyzed for Cohort 1 (treatment-naive) and was defined as the maximum observed concentration of drug in plasma. (NCT01363011)
Timeframe: Blood samples were collected at 0 (predose), 0.5, 1.0, 2.0, 3.0, 4.0, 5.0, 8.0, 12.0, and 24.0 hours postdose at baseline and Weeks 2, 4, and 24.

Interventionng/mL (Number)
Week 2Week 4Week 24
E/C/F/TDF (Cohort 1)1734.61522.91266.4

[back to top]

Plasma Pharmacokinetics of COBI: Cmax (Cohort 2)

Cmax was analyzed for Cohort 2 (treatment-experienced) and was defined as the maximum observed concentration of drug in plasma. (NCT01363011)
Timeframe: Blood samples were collected at 0 (predose), 0.5, 1.0, 2.0, 3.0, 4.0, 5.0, 8.0, 12.0, and 24.0 hours postdose at baseline and Weeks 2, 4, and 24.

Interventionng/mL (Mean)
Week 2 (n = 13)Week 4 (n = 13)Week 24 (n = 11)
COBI+PI+2 NRTIs (Cohort 2)1366.71297.71568.6

[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]

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]

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]

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]

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 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 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 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 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 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 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 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 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 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 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 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 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 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 48 in Precursors of Circulating Endothelial Cells

(NCT01391013)
Timeframe: Baseline to Week 48

,
InterventionEndothelial cells (Median)
BaselineWeek 48
Combination Therapy18108
Monotherapy16120

[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 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]

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]

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]

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 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]

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]

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 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]

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]

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]

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]

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]

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]

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 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]

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]

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 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]

Number of Participants With at Least One Adverse Event.

(NCT01422369)
Timeframe: 16 Days

InterventionParticipants (Number)
All Subjects2

[back to top]

NK-104 AUC

(NCT01422369)
Timeframe: 16 Days

Interventionng * h/mL (Mean)
NK-104192.00

[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]

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]

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]

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]

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]

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]

Percentage of Participants With Onset of Any Treatment-emergent Grade 3 or 4 Adverse Event Between Baseline and Week 24

(NCT01440569)
Timeframe: Up to 24 weeks

Interventionpercentage of participants (Number)
Treatment-Naive5.4
Treatment-Experienced11.1

[back to top]

Change From Baseline in CD4+ Cell Count at Week 24

(NCT01440569)
Timeframe: Baseline; Week 24

Interventioncells/μL (Mean)
Treatment-Naive145
Treatment-Experienced99

[back to top]

Change From Baseline in CD4+ Cell Count at Week 48

(NCT01440569)
Timeframe: Baseline; Week 48

Interventioncells/µL (Mean)
Treatment-Naive194
Treatment-Experienced121

[back to top]

Percentage of Participants Achieving HIV-1 RNA < 50 Copies/mL at Week 24 (Snapshot Analysis)

(NCT01440569)
Timeframe: Week 24

Interventionpercentage of participants (Number)
Treatment-Naive83.7
Treatment-Experienced61.1

[back to top]

Percentage of Participants Achieving HIV-1 RNA < 50 Copies/mL at Week 48 (Snapshot Analysis)

(NCT01440569)
Timeframe: Week 48

Interventionpercentage of participants (Number)
Treatment-Naive82.7
Treatment-Experienced50.0

[back to top]

Percentage of Participants Experiencing Any Treatment-emergent Adverse Event and Any Treatment-emergent Adverse Event Leading to Discontinuation of Study Drug Through Week 24

(NCT01440569)
Timeframe: Up to 24 weeks

,
Interventionpercentage of participants (Number)
Any treatment-emergent adverse event (TEAE)Any TEAE that led to study drug discontinuation
Treatment-Experienced83.30
Treatment-Naive88.15.1

[back to top]

Percentage of Participants Experiencing Any Treatment-emergent Adverse Event and Any Treatment-emergent Adverse Event Leading to Discontinuation of Study Drug Through Week 48

(NCT01440569)
Timeframe: Up to 48 weeks

,
Interventionpercentage of participants (Number)
Any treatment-emergent adverse event (TEAE)Any TEAE that led to study drug discontinuation
Treatment-Experienced88.90
Treatment-Naive91.55.4

[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]

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]

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 (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]

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]

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]

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 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]

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]

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]

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 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 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]

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 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]

Change From Baseline in HIV-1 RNA at Week 48

(NCT01565850)
Timeframe: Baseline; Week 48

Interventionlog10 copies/mL (Mean)
D/C/F/TAF-3.27
DRV+COBI+FTC/TDF-3.26

[back to top]

Change From Baseline in HIV-1 RNA at Week 24

(NCT01565850)
Timeframe: Baseline; Week 24

Interventionlog10 copies/mL (Mean)
D/C/F/TAF-3.20
DRV+COBI+FTC/TDF-3.18

[back to top]

Change From Baseline in CD4+ Cell Count at Week 48

(NCT01565850)
Timeframe: Baseline; Week 48

Interventioncells/µL (Mean)
D/C/F/TAF231
DRV+COBI+FTC/TDF212

[back to top]

Change From Baseline in CD4+ Cell Count at Week 24

(NCT01565850)
Timeframe: Baseline; Week 24

Interventioncells/µL (Mean)
D/C/F/TAF186
DRV+COBI+FTC/TDF139

[back to top]

Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 48

The snapshot algorithm was used which defines a patient's virologic response status using only the viral load at the predefined time point within an allowed window of time, along with study drug discontinuation status. (NCT01565850)
Timeframe: Week 48

Interventionpercentage of participants (Number)
D/C/F/TAF76.7
DRV+COBI+FTC/TDF84.0

[back to top]

Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 24

The snapshot algorithm was used which defines a patient's virologic response status using only the viral load at the predefined time point within an allowed window of time, along with study drug discontinuation status. (NCT01565850)
Timeframe: Week 24

Interventionpercentage of participants (Number)
D/C/F/TAF74.8
DRV+COBI+FTC/TDF74.0

[back to top]

Time to Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study

Virologic failure was confirmed by the next HIV-1 RNA measurement ≥1000 copies/mL (irrespective of time between initial and confirmatory measure [specimens on separate dates] and treatment status). A week 24 measurement included HIV-1 RNA obtained ≥7*22=154 days after study entry (to allow for 14 day window for scheduling the visit). HIV-1 RNA measurements through and including 21 November 2016 were considered in identifying an initial failing measurement, allowing HIV-1 RNA at the close-out visit between 22 November 2016 and 13 February 2017 to be a confirmatory measure. Event times were the scheduled week of the initial failing measurement (RNA scheduled at week 0, 12, 24, 48 and every 24 weeks after). Censoring times were the scheduled week of the last RNA result. Length of follow-up varied by Cohort. (NCT01641367)
Timeframe: From week 24 through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

,,,,,
Interventionweeks (Number)
1st percentile5th percentile10th percentile25th percentile50th percentile
Experimental: Cohort A2424242460
Experimental: Cohort C2448120NANA
Experimental: Cohort D242424NANA
Experimental: Sub-cohort B12448NANANA
Experimental: Sub-cohort B22472144NANA
Experimental: Sub-cohort B3NANANANANA

[back to top]

Percent of Participants That Developed Immune Reconstitution Inflammatory Syndrome (IRIS) by Week 48 [CPI+SOC v SOC]

Results report percent of participants reaching outcome by week 48 using Kaplan-Meier method. Event time was the exact week of the diagnosis. Censoring time was the earlier of last contact week and the week of the last step 1/2 visit. (NCT01641367)
Timeframe: From study entry to Week 48

Interventionpercentage of participants (Number)
Cell Phone Intervention (CPI) + Standard of Care (SOC)0.4
Standard of Care (SOC)1.1

[back to top]

Number of Participants With Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study

Virologic failure was confirmed by the next HIV-1 RNA measurement ≥1000 copies/mL (irrespective of time between initial and confirmatory measure [specimens on separate dates] and treatment status). A week 24 measurement included HIV-1 RNA obtained ≥7*22=154 days after study entry (to allow for 14 day window for scheduling the visit). HIV-1 RNA measurements through and including 21 November 2016 were considered in identifying an initial failing measurement, allowing HIV-1 RNA at the close-out visit between 22 November 2016 and 13 February 2017 to be a confirmatory measure. Length of follow-up varied by Cohort. (NCT01641367)
Timeframe: From week 24 through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

InterventionParticipants (Count of Participants)
Experimental: Cohort A145
Experimental: Sub-cohort B16
Experimental: Sub-cohort B24
Experimental: Sub-cohort B30
Experimental: Cohort C5
Experimental: Cohort D6

[back to top]

Number of Participants With Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study [CPI+SOC v SOC]

Virologic failure was confirmed by the next HIV-1 RNA measurement ≥1000 copies/mL (irrespective of time between initial and confirmatory measure [specimens on separate dates] and treatment status). A week 24 measurement included HIV-1 RNA obtained ≥7*22=154 days after study entry (to allow for 14 day window for scheduling the visit). HIV-1 RNA measurements through and including 21 November 2016 were considered in identifying an initial failing measurement, allowing HIV-1 RNA at the close-out visit between 22 November 2016 and 13 February 2017 to be a confirmatory measure. (NCT01641367)
Timeframe: From week 24 through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

InterventionParticipants (Count of Participants)
Cell Phone Intervention (CPI) + Standard of Care (SOC)66
Standard of Care (SOC)89

[back to top]

Number of Participants With Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study, With a New Resistance-associated Mutation Detected in Population-based Sequencing

Virologic failure was confirmed by the next HIV-1 RNA measurement ≥1000 copies/mL (irrespective of time between initial and confirmatory measure[specimens on separate dates] and treatment status). A week 24 measurement included HIV-1 RNA obtained ≥7*22=154 days after study entry (to allow for 14 day window for scheduling the visit).HIV-1 RNA measurements through and including 21 November 2016 were considered in identifying an initial failing measurement, allowing HIV-1 RNA at the close-out visit between 22 November 2016 and 13 February 2017 to be a confirmatory measure. Length of follow-up varied by Cohort. A new resistance-associated mutation is defined as one not present in the genotype prior to entry. (NCT01641367)
Timeframe: From week 24 through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

InterventionParticipants (Count of Participants)
Experimental: Cohort A48
Experimental: Sub-cohort B11
Experimental: Sub-cohort B22
Experimental: Sub-cohort B30
Experimental: Cohort C1
Experimental: Cohort D5

[back to top]

Percent of Participants With a Dose Modification Due to Grade 3 or 4 Toxicity by Week 48

Results report percent of participants reaching outcome by week 48 using Kaplan-Meier method. Event time was the exact week of the modification. Censoring time was the earliest time point between last dose week and the week of the last step 1/2 visit. DAIDS AE Grading Table, Version 1.0 was used. (NCT01641367)
Timeframe: From study entry to week 48

Interventionpercentage of participants (Number)
Experimental: Cohort A1.0
Experimental: Sub-cohort B10
Experimental: Sub-cohort B20
Experimental: Sub-cohort B30
Experimental: Cohort C0
Experimental: Cohort D0

[back to top]

Time to First Dose Modification Due to Grade 3 or 4 Toxicity [CPI+SOC v SOC]

Event time was the exact week of the modification. Censoring time was the earliest time point between last dose week and the week of the last step 1/2 visit. DAIDS AE Grading Table, Version 1.0 was used. (NCT01641367)
Timeframe: From study entry through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

,
Interventionweeks (Number)
1st percentile5th percentile
Cell Phone Intervention (CPI) + Standard of Care (SOC)45.7NA
Standard of Care (SOC)NANA

[back to top]

Time to First Dose Modification Due to Grade 3 or 4 Toxicity

Event time was the exact week of the modification. Censoring time was the earliest time point between last dose week and the week of the last step 1/2 visit. Length of follow-up varied by Cohort. DAIDS AE Grading Table, Version 1.0 was used. (NCT01641367)
Timeframe: From study entry through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

,,,,,
Interventionweeks (Number)
1st percentile5th percentile
Experimental: Cohort A45.7NA
Experimental: Cohort CNANA
Experimental: Cohort DNANA
Experimental: Sub-cohort B163.3NA
Experimental: Sub-cohort B2NANA
Experimental: Sub-cohort B3NANA

[back to top]

Time to Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study, With a New Resistance-associated Mutation Detected in Population-based Sequencing [CPI+SOC v SOC]

Virologic failure was confirmed by the next HIV-1 RNA measurement ≥1000 copies/mL (irrespective of time between initial and confirmatory measure [specimens on separate dates] and treatment status). A week 24 measurement included HIV-1 RNA obtained ≥7*22=154 days after study entry (to allow for 14 day window for scheduling the visit). HIV-1 RNA measurements through and including 21 November 2016 were considered in identifying an initial failing measurement, allowing HIV-1 RNA at the close-out visit between 22 November 2016 and 13 February 2017 to be a confirmatory measure. Event times were the scheduled week of the initial failing measurement (RNA scheduled at week 0, 12, 24, 48 and every 24 weeks after). Censoring times were the scheduled week of the last RNA result. A new resistance-associated mutation is defined as one not present in the genotype prior to entry. (NCT01641367)
Timeframe: From week 24 through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

,
Interventionweeks (Number)
1st percentile5th percentile10th percentile25th percentile
Cell Phone Intervention (CPI) + Standard of Care (SOC)2424NANA
Standard of Care (SOC)242448NA

[back to top]

Time to Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study, With a New Resistance-associated Mutation Detected in Population-based Sequencing

Virologic failure was confirmed by the next HIV-1 RNA measurement ≥1000 copies/mL (irrespective of time between initial and confirmatory measure [specimens on separate dates] and treatment status). A week 24 measurement included HIV-1 RNA obtained ≥7*22=154 days after study entry (to allow for 14 day window for scheduling the visit). HIV-1 RNA measurements through and including 21 November 2016 were considered in identifying an initial failing measurement, allowing HIV-1 RNA at the close-out visit between 22 November 2016 and 13 February 2017 to be a confirmatory measure. Event times were the scheduled week of the initial failing measurement (RNA scheduled at week 0, 12, 24, 48 and every 24 weeks after). Censoring times were the scheduled week of the last RNA result. Length of follow-up varied by Cohort. A new resistance-associated mutation is defined as one not present in the genotype prior to entry. (NCT01641367)
Timeframe: From week 24 through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

,,,,,
Interventionweeks (Number)
1st percentile5th percentile10th percentile25th percentile
Experimental: Cohort A242424144
Experimental: Cohort C48NANANA
Experimental: Cohort D242424NA
Experimental: Sub-cohort B124NANANA
Experimental: Sub-cohort B224NANANA
Experimental: Sub-cohort B3NANANANA

[back to top]

Time to Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study [CPI+SOC v SOC]

Virologic failure was confirmed by the next HIV-1 RNA measurement ≥1000 copies/mL (irrespective of time between initial and confirmatory measure [specimens on separate dates] and treatment status). A week 24 measurement included HIV-1 RNA obtained ≥7*22=154 days after study entry (to allow for 14 day window for scheduling the visit). HIV-1 RNA measurements through and including 21 November 2016 were considered in identifying an initial failing measurement, allowing HIV-1 RNA at the close-out visit between 22 November 2016 and 13 February 2017 to be a confirmatory measure. Event times were the scheduled week of the initial failing measurement (RNA scheduled at week 0, 12, 24, 48 and every 24 weeks after). Censoring times were the scheduled week of the last RNA result. (NCT01641367)
Timeframe: From week 24 through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

,
Interventionweeks (Number)
1st percentile5th percentile10th percentile25th percentile50th percentile
Cell Phone Intervention (CPI) + Standard of Care (SOC)24242460NA
Standard of Care (SOC)24242424NA

[back to top]

Change From Baseline in Fasting Values of Triglycerides

Baseline is defined as the last measurement obtained on or before the earlier of the following two dates: the date of entry/randomization plus three days (this is the time allowed in the protocol for starting study-defined ART) and the date of starting the study-defined ARV regimen (this second date applies only to Cohorts B, C and D as there is no change of regimen for patients in Cohort A) (NCT01641367)
Timeframe: Baseline, week 24, 48 and 72

,,,,,
Interventionmg/dL (Mean)
Change from baseline at week 24Change from baseline at week 48Change from baseline at week 72
Experimental: Cohort A15.412.217.5
Experimental: Cohort C15.49.911.8
Experimental: Cohort D28.924.46.7
Experimental: Sub-cohort B1-3.6-11.5-31.3
Experimental: Sub-cohort B227.619.918.9
Experimental: Sub-cohort B336.022.220.7

[back to top]

Time From Study Entry/Randomization to Treatment Modification or Discontinuation.

Treatment modification is defined as the first occurrence of a substitution or subtraction of one or more drugs in the study regimen, a temporary hold lasting 7 days or longer, or the addition of a new drug to the regimen. This would not include splitting any fixed dose combination medications if the participant continues on the active drugs of the combination. Event time was the exact week of the modification or discontinuation. Censoring time was the earlier of last contact week and the week of the last step 1/2 visit. Length of follow-up varied by Cohort. (NCT01641367)
Timeframe: From study entry through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

,,,,,
Interventionweeks (Number)
1st percentile5th percentile10th percentile25th percentile50th percentile
Experimental: Cohort A1.08.425.358.6NA
Experimental: Cohort C0.14.129.7NANA
Experimental: Cohort D2.45.147.698.9NA
Experimental: Sub-cohort B13.133.459.0NANA
Experimental: Sub-cohort B24.436.038.6165.6NA
Experimental: Sub-cohort B34.44.44.4NANA

[back to top]

Time From Study Entry/Randomization to Treatment Modification or Discontinuation Due to Toxicity [CPI+SOC v SOC]

Treatment modification is defined as the first occurrence of a substitution or subtraction of one or more drugs in the study regimen, a temporary hold lasting 7 days or longer, or the addition of a new drug to the regimen, due to an adverse event. This would not include splitting any fixed dose combination medications if the participant continues on the active drugs of the combination. Event time was the exact week of the modification or discontinuation. Censoring time was the earliest time point between last dose week and the week of the last step 1/2 visit. DAIDS AE Grading Table, Version 1.0 was used. (NCT01641367)
Timeframe: From study entry through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

,
Interventionweeks (Number)
1st percentile5th percentile
Cell Phone Intervention (CPI) + Standard of Care (SOC)3.1NA
Standard of Care (SOC)9.0NA

[back to top]

Time From Study Entry/Randomization to Treatment Modification or Discontinuation Due to Toxicity

Treatment modification is defined as the first occurrence of a substitution or subtraction of one or more drugs in the study regimen, a temporary hold lasting 7 days or longer, or the addition of a new drug to the regimen, due to an adverse event. This would not include splitting any fixed dose combination medications if the participant continues on the active drugs of the combination. Event time was the exact week of the modification or discontinuation. Censoring time was the earliest time point between last dose week and the week of the last step 1/2 visit. Length of follow-up varied by Cohort. DAIDS AE Grading Table, Version 1.0 was used. (NCT01641367)
Timeframe: From study entry through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

,,,,,
Interventionweeks (Number)
1st percentile5th percentile10th percentile
Experimental: Cohort A2.1106.1NA
Experimental: Cohort C0.1NANA
Experimental: Cohort DNANANA
Experimental: Sub-cohort B115.0NANA
Experimental: Sub-cohort B26.4134.0NA
Experimental: Sub-cohort B3NANANA

[back to top]

Time From Study Entry/Randomization to Treatment Modification or Discontinuation [CPI+SOC v SOC]

Treatment modification is defined as the first occurrence of a substitution or subtraction of one or more drugs in the study regimen, a temporary hold lasting 7 days or longer, or the addition of a new drug to the regimen. This would not include splitting any fixed dose combination medications if the participant continues on the active drugs of the combination. Event time was the exact week of the modification or discontinuation. Censoring time was the earlier of last contact week and the week of the last step 1/2 visit. (NCT01641367)
Timeframe: From study entry through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

,
Interventionweeks (Number)
1st percentile5th percentile10th percentile25th percentile
Cell Phone Intervention (CPI) + Standard of Care (SOC)1.05.123.684.0
Standard of Care (SOC)2.422.338.9111.1

[back to top]

Time From Study Entry/Randomization to the First of Death, an AIDS-defining Event or a Non-AIDS-defining Event [CPI+SOC v SOC]

Event time was the exact week of death, AIDS-defining event or non-AIDS defining event. Censoring time was the earlier of last contact week and the week of the last step 1/2 visit. Only new events were considered, an event that was also reported at or prior to study entry was not included. If a participant experienced multiple events, then the time of the first event was used in the analysis. AIDS defining events included parasitic, fungal, bacterial, and viral infections as well as neoplastic diseases, and neurological disorders. Non-AIDS defining events included malignancies, diabetes, neuropathies, cardiac and renal events. (NCT01641367)
Timeframe: From study entry through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

,
Interventionweeks (Number)
1st percentile5th percentile10th percentile25th percentile
Cell Phone Intervention (CPI) + Standard of Care (SOC)4.027.660.6NA
Standard of Care (SOC)4.042.377.9NA

[back to top]

Time From Study Entry/Randomization to the First of Death, an AIDS-defining Event or a Non-AIDS-defining Event

Event time was the exact week of death, AIDS-defining event or non-AIDS defining event. Censoring time was the earlier of last contact week and the week of the last step 1/2 visit. Only new events were considered, an event that was also reported at or prior to study entry was not included. If a participant experienced multiple events, then the time of the first event was used in the analysis. AIDS defining events included parasitic, fungal, bacterial, and viral infections as well as neoplastic diseases, and neurological disorders. Non-AIDS defining events included malignancies, diabetes, neuropathies, cardiac and renal events. Length of follow-up varied by Cohort. (NCT01641367)
Timeframe: From study entry through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

,,,,,
Interventionweeks (Number)
1st percentile5th percentile10th percentile25th percentile
Experimental: Cohort A4.027.657.9NA
Experimental: Cohort C3.336.077.9142.4
Experimental: Cohort D2.424.048.496.3
Experimental: Sub-cohort B13.136.084.0NA
Experimental: Sub-cohort B216.350.3120.0NA
Experimental: Sub-cohort B3NANANANA

[back to top]

Time From Study Entry/Randomization to the First of Death or Hospitalization.

Event time was the exact week of death or hospitalization. Censoring time was the earlier of last contact week and the week of the last step 1/2 visit. If a participant experienced multiple events, then the time of the first event was used in the analysis. Length of follow-up varied by Cohort. (NCT01641367)
Timeframe: From study entry through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

,,,,,
Interventionweeks (Number)
1st percentile5th percentile10th percentile25th percentile50th percentile
Experimental: Cohort A2.413.432.6120.1168.9
Experimental: Cohort C2.07.777.9NANA
Experimental: Cohort D2.35.696.1NANA
Experimental: Sub-cohort B12.320.380.7NANA
Experimental: Sub-cohort B23.028.049.7NANA
Experimental: Sub-cohort B316.416.416.4NANA

[back to top]

Time From Study Entry/Randomization to the Development of Immune Reconstitution Inflammatory Syndrome (IRIS) [CPI+SOC v SOC]

Event time was the exact week of the diagnosis. Censoring time was the earlier of last contact week and the week of the last step 1/2 visit. (NCT01641367)
Timeframe: From study entry through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

,
Interventionweeks (Number)
1st percentile5th percentile
Cell Phone Intervention (CPI) + Standard of Care (SOC)NANA
Standard of Care (SOC)25.0NA

[back to top]

Time From Study Entry/Randomization to the Development of Immune Reconstitution Inflammatory Syndrome (IRIS)

Event time was the exact week of the diagnosis. Censoring time was the earlier of last contact week and the week of the last step 1/2 visit. Length of follow-up varied by Cohort. (NCT01641367)
Timeframe: From study entry through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

,,,,,
Interventionweeks (Number)
1st percentile5th percentile
Experimental: Cohort ANANA
Experimental: Cohort CNANA
Experimental: Cohort D13.0NA
Experimental: Sub-cohort B1NANA
Experimental: Sub-cohort B225.0NA
Experimental: Sub-cohort B3NANA

[back to top]

Time From Study Entry/Randomization to Death [CPI+SOC v SOC]

Event time was the exact week of death. Censoring time was the earlier of last contact week and the week of the last step 1/2 visit. (NCT01641367)
Timeframe: From study entry through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

,
Interventionweeks (Number)
1st percentile5th percentile10th percentile
Cell Phone Intervention (CPI) + Standard of Care (SOC)11.3NANA
Standard of Care (SOC)15.982.1NA

[back to top]

Time From Study Entry/Randomization to Death

Event time was the exact week of death. Censoring time was the earlier of last contact week and the week of the last step 1/2 visit. Length of follow-up varied by Cohort. (NCT01641367)
Timeframe: From study entry through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

,,,,,
Interventionweeks (Number)
1st percentile5th percentile10th percentile
Experimental: Cohort A11.362.4NA
Experimental: Cohort C77.9NANA
Experimental: Cohort D2.4NANA
Experimental: Sub-cohort B13.1NANA
Experimental: Sub-cohort B244.6NANA
Experimental: Sub-cohort B3NANANA

[back to top]

Change From Baseline in Fasting Values of Triglycerides [CPI+SOC v SOC]

Baseline is defined as the last measurement obtained on or before the earlier of the following two dates: the date of entry/randomization plus three days (this is the time allowed in the protocol for starting study-defined ART) and the date of starting the study-defined ARV regimen. (NCT01641367)
Timeframe: Baseline, week 24, 48, and 72

,
Interventionmg/dL (Mean)
Change from baseline at week 24Change from baseline at week 48Change from baseline at week 72
Cell Phone Intervention (CPI) + Standard of Care (SOC)15.32.213.7
Standard of Care (SOC)18.719.67.1

[back to top]

Change From Baseline in Fasting Values of Total Cholesterol [CPI+SOC v SOC]

Baseline is defined as the last measurement obtained on or before the earlier of the following two dates: the date of entry/randomization plus three days (this is the time allowed in the protocol for starting study-defined ART) and the date of starting the study-defined ARV regimen. (NCT01641367)
Timeframe: Baseline, week 24, 48, and 72

,
Interventionmg/dL (Mean)
Change from baseline at week 24Change from baseline at week 48Change from baseline at week 72
Cell Phone Intervention (CPI) + Standard of Care (SOC)10.115.117.4
Standard of Care (SOC)14.414.118.7

[back to top]

Change From Baseline in Fasting Values of Total Cholesterol

Baseline is defined as the last measurement obtained on or before the earlier of the following two dates: the date of entry/randomization plus three days (this is the time allowed in the protocol for starting study-defined ART) and the date of starting the study-defined ARV regimen (this second date applies only to Cohorts B, C and D as there is no change of regimen for patients in Cohort A) (NCT01641367)
Timeframe: Baseline, week 24, 48 and 72

,,,,,
Interventionmg/dL (Mean)
Change from baseline at week 24Change from baseline at week 48Change from baseline at week 72
Experimental: Cohort A5.74.47.6
Experimental: Cohort C16.520.022.1
Experimental: Cohort D7.919.124.5
Experimental: Sub-cohort B116.719.722.6
Experimental: Sub-cohort B232.540.440.4
Experimental: Sub-cohort B312.49.928.2

[back to top]

Change From Baseline in Fasting Values of High-density Lipoprotein Cholesterol [CPI+SOC v SOC]

Baseline is defined as the last measurement obtained on or before the earlier of the following two dates: the date of entry/randomization plus three days (this is the time allowed in the protocol for starting study-defined ART) and the date of starting the study-defined ARV regimen. (NCT01641367)
Timeframe: Baseline, week 24, 48, and 72

,
Interventionmg/dL (Mean)
Change from baseline at week 24Change from baseline at week 48Change from baseline at week 72
Cell Phone Intervention (CPI) + Standard of Care (SOC)2.95.66.0
Standard of Care (SOC)3.63.76.6

[back to top]

Change From Baseline in Fasting Values of High-density Lipoprotein Cholesterol

Baseline is defined as the last measurement obtained on or before the earlier of the following two dates: the date of entry/randomization plus three days (this is the time allowed in the protocol for starting study-defined ART) and the date of starting the study-defined ARV regimen (this second date applies only to Cohorts B, C and D as there is no change of regimen for patients in Cohort A)] (NCT01641367)
Timeframe: Baseline, week 24, 48 and 72

,,,,,
Interventionmg/dL (Mean)
Change from baseline at week 24Change from baseline at week 48Change from baseline at week 72
Experimental: Cohort A2.83.54.7
Experimental: Cohort C1.02.35.8
Experimental: Cohort D-2.21.83.4
Experimental: Sub-cohort B13.25.34.4
Experimental: Sub-cohort B211.413.415.7
Experimental: Sub-cohort B32.13.84.6

[back to top]

Change From Baseline in Fasting Values of Glucose [CPI+SOC v SOC]

Baseline is defined as the last measurement obtained on or before the earlier of the following two dates: the date of entry/randomization plus three days (this is the time allowed in the protocol for starting study-defined ART) and the date of starting the study-defined ARV regimen. (NCT01641367)
Timeframe: Baseline, week 24, 48, and 72

,
Interventionmg/dL (Mean)
Change from baseline at week 24Change from baseline at week 48Change from baseline at week 72
Cell Phone Intervention (CPI) + Standard of Care (SOC)3.73.63.6
Standard of Care (SOC)3.75.11.5

[back to top]

Change From Baseline in Fasting Values of Glucose

Baseline is defined as the last measurement obtained on or before the earlier of the following two dates: the date of entry/randomization plus three days (this is the time allowed in the protocol for starting study-defined ART) and the date of starting the study-defined ARV regimen (this second date applies only to Cohorts B, C and D as there is no change of regimen for patients in Cohort A) (NCT01641367)
Timeframe: Baseline, week 24, 48 and 72

,,,,,
Interventionmg/dL (Mean)
Change from baseline at week 24Change from baseline at week 48Change from baseline at week 72
Experimental: Cohort A1.92.13.0
Experimental: Cohort C2.13.0-0.9
Experimental: Cohort D3.24.27.8
Experimental: Sub-cohort B18.89.36.8
Experimental: Sub-cohort B26.16.2-5.2
Experimental: Sub-cohort B36.61.74.3

[back to top]

Change From Baseline in Fasting Values of Calculated Low-density Lipoprotein Cholesterol [CPI+SOC v SOC]

Baseline is defined as the last measurement obtained on or before the earlier of the following two dates: the date of entry/randomization plus three days (this is the time allowed in the protocol for starting study-defined ART) and the date of starting the study-defined ARV regimen. (NCT01641367)
Timeframe: Baseline, week 24, 48, and 72

,
Interventionmg/dL (Mean)
Change from baseline at week 24Change from baseline at week 48Change from baseline at week 72
Cell Phone Intervention (CPI) + Standard of Care (SOC)5.512.011.9
Standard of Care (SOC)9.510.112.8

[back to top]

Time From Study Entry/Randomization to the First of Death or Hospitalization [CPI+SOC v SOC]

Event time was the exact week of death or hospitalization. Censoring time was the earlier of last contact week and the week of the last step 1/2 visit. If a participant experienced multiple events, then the time of the first event was used in the analysis. (NCT01641367)
Timeframe: From study entry through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

,
Interventionweeks (Number)
1st percentile5th percentile10th percentile25th percentile
Cell Phone Intervention (CPI) + Standard of Care (SOC)2.311.344.6168.9
Standard of Care (SOC)2.320.345.3NA

[back to top]

Percent of Participants With Treatment Modification or Discontinuation by Week 48 [CPI+SOC v SOC]

Results report percent of participants reaching outcome by week 48 using Kaplan-Meier method. Treatment modification is defined as the first occurrence of a substitution or subtraction of one or more drugs in the study regimen, a temporary hold lasting 7 days or longer, or the addition of a new drug to the regimen. This would not include splitting any fixed dose combination medications if the participant continues on the active drugs of the combination. Event time was the exact week of the modification or discontinuation. Censoring time was the earliest time point between last dose week and the week of the last step 1/2 visit. (NCT01641367)
Timeframe: From study entry to Week 48

Interventionpercentage of participants (Number)
Cell Phone Intervention (CPI) + Standard of Care (SOC)19.1
Standard of Care (SOC)13.6

[back to top]

Change From Baseline in Fasting Values of Calculated Low-density Lipoprotein Cholesterol

Baseline is defined as the last measurement obtained on or before the earlier of the following two dates: the date of entry/randomization plus three days (this is the time allowed in the protocol for starting study-defined ART) and the date of starting the study-defined ARV regimen (this second date applies only to Cohorts B, C and D as there is no change of regimen for patients in Cohort A) (NCT01641367)
Timeframe: Baseline, week 24, 48 and 72

,,,,,
Interventionmg/dL (Mean)
Change from baseline at week 24Change from baseline at week 48Change from baseline at week 72
Experimental: Cohort A1.33.43.9
Experimental: Cohort C12.215.313.6
Experimental: Cohort D9.914.419.7
Experimental: Sub-cohort B113.316.322.4
Experimental: Sub-cohort B221.528.227.8
Experimental: Sub-cohort B3-0.50.316.6

[back to top]

Change From Baseline in CD4+ T-cell Count [CPI+SOC v SOC]

Baseline is defined as the last measurement obtained on or before the earlier of the following two dates: the date of entry/randomization plus three days (this is the time allowed in the protocol for starting study-defined ART) and the date of starting the study-defined ARV regimen. (NCT01641367)
Timeframe: Baseline, week 24, 48, and 72

,
Interventioncells/mm^3 (Mean)
Change from baseline at week 24Change from baseline at week 48Change from baseline at week 72
Cell Phone Intervention (CPI) + Standard of Care (SOC)72112145
Standard of Care (SOC)74107134

[back to top]

Change From Baseline in CD4+ T-cell Count

Baseline is defined as the last measurement obtained on or before the earlier of the following two dates: the date of entry/randomization plus three days (this is the time allowed in the protocol for starting study-defined ART) and the date of starting the study-defined ARV regimen (this second date applies only to Cohorts B, C and D as there is no change of regimen for patients in Cohort A) (NCT01641367)
Timeframe: Baseline, week 24, 48, and 72

,,,,,
Interventioncells/mm^3 (Mean)
Change from baseline at week 24Change from baseline at week 48Change from baseline at week 72
Experimental: Cohort A396587
Experimental: Cohort C100160185
Experimental: Cohort D90135165
Experimental: Sub-cohort B1109157182
Experimental: Sub-cohort B2116158197
Experimental: Sub-cohort B314286238

[back to top]

Proportion of Participants With Plasma HIV-1 RNA ≤200 Copies/mL at 72 Weeks [CPI+SOC v SOC]

"The measurement closest to exactly 72 weeks (ie, 7x72=504 days) after the date of entry, within the window of 72 weeks ± 6 weeks (specifically 463 to 546 days, inclusive).~The analysis in the protocol and in the Analysis Plan involved estimating the proportion of participants in the overall study population with HIV-1 RNA ≤200 copies/mL at week 72 with a 95% confidence interval calculated via a Wald approach. Death or lost to follow-up before week 72 was considered as HIV-1 RNA>200 copies/mL at week 72. If a result was expected, missing results at week 72 were considered as HIV-1 RNA >200 copies/mL at week 72 unless the immediately preceding and succeeding HIV-1 RNA measurements were ≤200 copies/mL." (NCT01641367)
Timeframe: 72 weeks after the date of entry

Interventionproportion of participants (Number)
Cell Phone Intervention (CPI) + Standard of Care (SOC)0.69
Standard of Care (SOC)0.62

[back to top]

Proportion of Participants With Plasma HIV-1 RNA ≤200 Copies/mL at 72 Weeks

"The measurement closest to exactly 72 weeks (ie, 7x72=504 days) after the date of entry, within the window of 72 weeks ± 6 weeks (specifically 463 to 546 days, inclusive).~The analysis in the protocol and in the Analysis Plan involved estimating the proportion of participants in the overall study population with HIV-1 RNA ≤200 copies/mL at week 72 with a 95% confidence interval calculated via a Wald approach. Death or lost to follow-up before week 72 was considered as HIV-1 RNA>200 copies/mL at week 72. If a result was expected, missing results at week 72 were considered as HIV-1 RNA >200 copies/mL at week 72 unless the immediately preceding and succeeding HIV-1 RNA measurements were ≤200 copies/mL. Since the primary analysis was on the total study population, overall results were also submitted. All participants in B3 had HIV-1 RNA ≤200 copies/mL at week 72. Therefore, Wald confidence interval could not be computed for B3 and Clopper-Pearson Exact confidence interval is provided." (NCT01641367)
Timeframe: 72 weeks after the date of entry

Interventionproportion of participants (Number)
Overall Study0.64
Experimental: Cohort A0.44
Experimental: Sub-cohort B10.92
Experimental: Sub-cohort B20.87
Experimental: Sub-cohort B31.00
Experimental: Cohort C0.85
Experimental: Cohort D0.77

[back to top]

Proportion of Participants With Plasma HIV-1 RNA ≤200 Copies/mL at 48 Weeks [CPI+SOC v SOC]

"The measurement closest to exactly 48 weeks (ie, 7x48=336 days) after the date of entry, within the window of 48 weeks ± 6 weeks (specifically 295 to 378 days after randomization, inclusive).~The analysis in the protocol and in the Stat. Analysis Plan involved estimating the proportion of participants in the overall study population with HIV-1 RNA ≤200 copies/mL at week 48 with a 95% confidence interval calculated via a Wald approach. Death or lost to follow-up before week 48 was considered as HIV-1 RNA>200 copies/mL at week 48. Missing results at week 48 were considered as HIV-1 RNA >200 copies/mL at week 48 unless the immediately preceding and succeeding HIV-1 RNA measurements were ≤200 copies/mL." (NCT01641367)
Timeframe: 48 weeks after the date of entry

Interventionproportion of participants (Number)
Cell Phone Intervention (CPI) + Standard of Care (SOC)0.66
Standard of Care (SOC)0.62

[back to top]

Proportion of Participants With Plasma HIV-1 RNA ≤200 Copies/mL at 48 Weeks

"The measurement closest to exactly 48 weeks (ie, 7x48=336 days) after the date of entry, within the window of 48 weeks ± 6 weeks (specifically 295 to 378 days after randomization, inclusive).~The analysis in the protocol and in the Stat. Analysis Plan involved estimating the proportion of participants in the overall study population with HIV-1 RNA ≤200 copies/mL at week 48 with a 95% confidence interval calculated via a Wald approach. Death or lost to follow-up before week 48 was considered as HIV-1 RNA>200 copies/mL at week 48. Missing results at week 48 were considered as HIV-1 RNA >200 copies/mL at week 48 unless the immediately preceding and succeeding HIV-1 RNA measurements were ≤200 copies/mL. Since the primary analysis was on the total study population, overall results were also submitted. All participants in B3 had HIV-1 RNA ≤200 copies/mL at week 48. Therefore, Wald confidence interval could not be computed for B3 and Clopper-Pearson Exact confidence interval is provided." (NCT01641367)
Timeframe: 48 weeks after the date of entry

Interventionproportion of participants (Number)
Overall Study0.64
Experimental: Cohort A0.44
Experimental: Sub-cohort B10.88
Experimental: Sub-cohort B20.88
Experimental: Sub-cohort B31.00
Experimental: Cohort C0.90
Experimental: Cohort D0.74

[back to top]

Proportion of Participants With Plasma HIV-1 RNA ≤200 Copies/mL at 24 Weeks [CPI+SOC v SOC]

"The measurement closest to exactly 24 weeks (ie, 7x24=168 days) after the date of entry, within the window of 24 weeks ± 6 weeks (specifically 127 to 210 days after randomization, inclusive).~The analysis in the protocol and in the Stat. Analysis Plan involved estimating the proportion of participants in the overall study population with HIV-1 RNA ≤200 copies/mL at week 24 with a 95% confidence interval calculated via a Wald approach. Death or lost to follow-up before week 24 was considered as HIV-1 RNA>200 copies/mL at week 24. Missing results at week 24 were considered as HIV-1 RNA >200 copies/mL at week 24 unless the immediately preceding and succeeding HIV-1 RNA measurements were ≤200 copies/mL." (NCT01641367)
Timeframe: 24 weeks after the date of entry

Interventionproportion of participants (Number)
Cell Phone Intervention (CPI) + Standard of Care (SOC)0.68
Standard of Care (SOC)0.61

[back to top]

Proportion of Participants With Plasma HIV-1 RNA ≤200 Copies/mL at 24 Weeks

"The measurement closest to exactly 24 weeks (ie, 7x24=168 days) after the date of entry, within the window of 24 weeks ± 6 weeks (specifically 127 to 210 days after randomization, inclusive).~The analysis in the protocol and in the Stat. Analysis Plan involved estimating the proportion of participants in the overall study population with HIV-1 RNA ≤200 copies/mL at week 24 with a 95% confidence interval calculated via a Wald approach. Death or lost to follow-up before week 24 was considered as HIV-1 RNA>200 copies/mL at week 24. Missing results at week 24 were considered as HIV-1 RNA >200 copies/mL at week 24 unless the immediately preceding and succeeding HIV-1 RNA measurements were ≤200 copies/mL. Since the primary analysis was on the total study population, overall results were also submitted. All participants in B3 had HIV-1 RNA ≤200 copies/mL at week 24. Therefore, Wald confidence interval could not be computed for B3 and Clopper-Pearson Exact confidence interval is provided." (NCT01641367)
Timeframe: 24 weeks after the date of entry

Interventionproportion of participants (Number)
Overall Study0.64
Experimental: Cohort A0.43
Experimental: Sub-cohort B10.89
Experimental: Sub-cohort B20.88
Experimental: Sub-cohort B31.00
Experimental: Cohort C0.90
Experimental: Cohort D0.74

[back to top]

Percent of Participants With Treatment Modification or Discontinuation Due to Toxicity by Week 48 [CPI+SOC v SOC]

Results report percent of participants reaching outcome by week 48 using Kaplan-Meier method. Treatment modification is defined as the first occurrence of a substitution or subtraction of one or more drugs in the study regimen, a temporary hold lasting 7 days or longer, or the addition of a new drug to the regimen, due to an adverse event. This would not include splitting any fixed dose combination medications if the participant continues on the active drugs of the combination. Event time was the exact week of the modification or discontinuation. Censoring time was the earliest time point between last dose week and the week of the last step 1/2 visit. DAIDS AE Grading Table, Version 1.0 was used. (NCT01641367)
Timeframe: From study entry to Week 48

Interventionpercentage of participants (Number)
Cell Phone Intervention (CPI) + Standard of Care (SOC)2.8
Standard of Care (SOC)2.3

[back to top]

Percent of Participants With Treatment Modification or Discontinuation Due to Toxicity by Week 48

Results report percent of participants reaching outcome by week 48 using Kaplan-Meier method. Treatment modification is defined as the first occurrence of a substitution or subtraction of one or more drugs in the study regimen, a temporary hold lasting 7 days or longer, or the addition of a new drug to the regimen, due to an adverse event. This would not include splitting any fixed dose combination medications if the participant continues on the active drugs of the combination. Event time was the exact week of the modification or discontinuation. Censoring time was the earliest time point between last dose week and the week of the last step 1/2 visit. DAIDS AE Grading Table, Version 1.0 was used. (NCT01641367)
Timeframe: From study entry to Week 48

Interventionpercentage of participants (Number)
Experimental: Cohort A3.2
Experimental: Sub-cohort B12.7
Experimental: Sub-cohort B21.4
Experimental: Sub-cohort B30
Experimental: Cohort C4.3
Experimental: Cohort D0

[back to top]

Percent of Participants Experiencing Death, AIDS-defining Event or a Non-AIDS-defining Event by Week 48

Results report percent of participants reaching outcome by week 48 using Kaplan-Meier method. Event time was the exact week of death, AIDS-defining event or non-AIDS defining event. Censoring time was the earlier of last contact week and the week of the last step 1/2 visit. Only new events were considered, an event that was also reported at or prior to study entry was not included. If a participant experienced multiple events, then the time of the first event was used in the analysis. AIDS defining events included parasitic, fungal, bacterial, and viral infections as well as neoplastic diseases, and neurological disorders. Non-AIDS defining events included malignancies, diabetes, neuropathies, cardiac and renal events. (NCT01641367)
Timeframe: From study entry to Week 48

Interventionpercentage of participants (Number)
Experimental: Cohort A8.8
Experimental: Sub-cohort B15.4
Experimental: Sub-cohort B24.2
Experimental: Sub-cohort B30
Experimental: Cohort C5.8
Experimental: Cohort D5.9

[back to top]

Percent of Participants With Treatment Modification or Discontinuation by Week 48

Results report percent of participants reaching outcome by week 48 using Kaplan-Meier method. Treatment modification is defined as the first occurrence of a substitution or subtraction of one or more drugs in the study regimen, a temporary hold lasting 7 days or longer, or the addition of a new drug to the regimen. This would not include splitting any fixed dose combination medications if the participant continues on the active drugs of the combination. Event time was the exact week of the modification or discontinuation. Censoring time was the earliest time point between last dose week and the week of the last step 1/2 visit. (NCT01641367)
Timeframe: From study entry to Week 48

Interventionpercentage of participants (Number)
Experimental: Cohort A19.9
Experimental: Sub-cohort B16.8
Experimental: Sub-cohort B219.4
Experimental: Sub-cohort B312.5
Experimental: Cohort C14.3
Experimental: Cohort D11.8

[back to top]

Percent of Participants With Death or Hospitalization by Week 48 [CPI+SOC v SOC]

Results report percent of participants reaching outcome by week 48 using Kaplan-Meier method. Event time was the exact week of death or hospitalization. Censoring time was the earlier of last contact week and the week of the last step 1/2 visit. If a participant experienced multiple events, then the time of the first event was used in the analysis. (NCT01641367)
Timeframe: From study entry to Week 48

Interventionpercentage of participants (Number)
Cell Phone Intervention (CPI) + Standard of Care (SOC)10.6
Standard of Care (SOC)10.6

[back to top]

Percent of Participants With Death or Hospitalization by Week 48

Results report percent of participants reaching outcome by week 48 using Kaplan-Meier method. Event time was the exact week of death or hospitalization. Censoring time was the earlier of last contact week and the week of the last step 1/2 visit. If a participant experienced multiple events, then the time of the first event was used in the analysis. (NCT01641367)
Timeframe: From study entry to Week 48

Interventionpercentage of participants (Number)
Experimental: Cohort A12.3
Experimental: Sub-cohort B18.1
Experimental: Sub-cohort B29.7
Experimental: Sub-cohort B312.5
Experimental: Cohort C5.7
Experimental: Cohort D5.9

[back to top]

Percent of Participants With Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study, With a New Resistance-associated Mutation Detected in Population-based Sequencing, by Week 48 [CPI+SOC v SOC]

Results report percent of participants reaching outcome by week 48 using Kaplan-Meier method. Virologic failure was confirmed by the next HIV-1 RNA measurement ≥1000 copies/mL (irrespective of time between initial and confirmatory measure[specimens on separate dates] and treatment status). A week 24 measurement included HIV-1 RNA obtained ≥7*22=154 days after study entry(to allow for 14 day window for scheduling the visit).HIV-1 RNA measurements through and including 21 November 2016 were considered in identifying an initial failing measurement,allowing HIV-1 RNA at the close-out visit between 22 November 2016 and 13 February 2017 to be a confirmatory measure.Event times were the scheduled week of the initial failing measurement(RNA scheduled at week 0, 12, 24, 48 and every 24 weeks after).Censoring times were the scheduled week of the last RNA result.A new resistance-associated mutation is defined as one not present in the genotype prior to entry. (NCT01641367)
Timeframe: From week 24 to Week 48

Interventionpercentage of participants (Number)
Cell Phone Intervention (CPI) + Standard of Care (SOC)7.8
Standard of Care (SOC)12.1

[back to top]

Percent of Participants With Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study, With a New Resistance-associated Mutation Detected in Population-based Sequencing, by Week 48

Results report percent of participants reaching outcome by week 48 using Kaplan-Meier method. Virologic failure was confirmed by the next HIV-1 RNA measurement ≥1000 copies/mL (irrespective of time between initial and confirmatory measure[specimens on separate dates] and treatment status). A week 24 measurement included HIV-1 RNA obtained ≥7*22=154 days after study entry(to allow for 14 day window for scheduling the visit).HIV-1 RNA measurements through and including 21 November 2016 were considered in identifying an initial failing measurement,allowing HIV-1 RNA at the close-out visit between 22 November 2016 and 13 February 2017 to be a confirmatory measure.Event times were the scheduled week of the initial failing measurement(RNA scheduled at week 0, 12, 24, 48 and every 24 weeks after).Censoring times were the scheduled week of the last RNA result.Length of follow-up varied by Cohort.A new resistance-associated mutation is defined as one not present in the genotype prior to entry. (NCT01641367)
Timeframe: From week 24 to Week 48

Interventionpercentage of participants (Number)
Experimental: Cohort A16.6
Experimental: Sub-cohort B11.4
Experimental: Sub-cohort B21.4
Experimental: Sub-cohort B30
Experimental: Cohort C1.5
Experimental: Cohort D15.4

[back to top]

Percent of Participants With Confirmed Virologic Failure by Week 48 [CPI+SOC v SOC]

Results report percent of participants reaching outcome by week 48 using Kaplan-Meier method. Virologic failure was confirmed by the next HIV-1 RNA measurement ≥1000 copies/mL (irrespective of time between initial and confirmatory measure [specimens on separate dates] and treatment status). A week 24 measurement included HIV-1 RNA obtained ≥7*22=154 days after study entry(to allow for 14 day window for scheduling the visit). HIV-1 RNA measurements through and including 21 November 2016 were considered in identifying an initial failing measurement,allowing HIV-1 RNA at the close-out visit between 22 November 2016 and 13 February 2017 to be a confirmatory measure. Event times were the scheduled week of the initial failing measurement (RNA scheduled at week 0, 12, 24, 48 and every 24 weeks after). Censoring times were the scheduled week of the last RNA result. (NCT01641367)
Timeframe: From week 24 to Week 48

Interventionpercentage of participants (Number)
Cell Phone Intervention (CPI) + Standard of Care (SOC)24.9
Standard of Care (SOC)32.2

[back to top]

Number of Participants With Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study, With a New Resistance-associated Mutation Detected in Population-based Sequencing [CPI+SOC v SOC]

Virologic failure was confirmed by the next HIV-1 RNA measurement ≥1000 copies/mL (irrespective of time between initial and confirmatory measure[specimens on separate dates] and treatment status). A week 24 measurement included HIV-1 RNA obtained ≥7*22=154 days after study entry (to allow for 14 day window for scheduling the visit).HIV-1 RNA measurements through and including 21 November 2016 were considered in identifying an initial failing measurement, allowing HIV-1 RNA at the close-out visit between 22 November 2016 and 13 February 2017 to be a confirmatory measure. A new resistance-associated mutation is defined as one not present in the genotype prior to entry. (NCT01641367)
Timeframe: From week 24 through Step 1/2 follow-up; median (IQR) step 1/2 follow-up was 72 (72,108) weeks

InterventionParticipants (Count of Participants)
Cell Phone Intervention (CPI) + Standard of Care (SOC)20
Standard of Care (SOC)32

[back to top]

Number of Weeks of Follow-up

All participants were followed on step 1/2 until 48 weeks after the last participant was enrolled to step 1 regardless of virologic status or treatment switches. Length of follow-up varied by Cohort. (NCT01641367)
Timeframe: From study entry through Step 1/2 follow-up

Interventionweeks (Median)
Experimental: Cohort A72
Experimental: Sub-cohort B196
Experimental: Sub-cohort B284
Experimental: Sub-cohort B396
Experimental: Cohort C72
Experimental: Cohort D96

[back to top]

Percent of Participants With a Dose Modification Due to Grade 3 or 4 Toxicity by Week 48 [CPI+SOC v SOC]

Results report percent of participants reaching outcome by week 48 using Kaplan-Meier method. Event time was the exact week of the modification. Censoring time was the earliest time point between last dose week and the week of the last step 1/2 visit. DAIDS AE Grading Table, Version 1.0 was used. (NCT01641367)
Timeframe: From study entry to Week 48

Interventionpercentage of participants (Number)
Cell Phone Intervention (CPI) + Standard of Care (SOC)1.2
Standard of Care (SOC)0

[back to top]

Number of Weeks of Follow-up [CPI+SOC v SOC]

All participants were followed on step 1/2 until 48 weeks after the last participant was enrolled to step 1 regardless of virologic status or treatment switches. (NCT01641367)
Timeframe: From study entry through Step 1/2 follow-up

Interventionweeks (Median)
Cell Phone Intervention (CPI) + Standard of Care (SOC)72
Standard of Care (SOC)72

[back to top]

Percent of Participants Experiencing Death by Week 48

Results report percent of participants reaching outcome by week 48 using Kaplan-Meier method. Event time was the exact week of death. Censoring time was the earlier of last contact week and the week of the last step 1/2 visit. (NCT01641367)
Timeframe: From study entry to Week 48

Interventionpercentage of participants (Number)
Experimental: Cohort A3.9
Experimental: Sub-cohort B11.4
Experimental: Sub-cohort B21.4
Experimental: Sub-cohort B30
Experimental: Cohort C0
Experimental: Cohort D2.9

[back to top]

Percent of Participants Experiencing Death by Week 48 [CPI+SOC v SOC]

Results report percent of participants reaching outcome by week 48 using Kaplan-Meier method. Event time was the exact week of death. Censoring time was the earlier of last contact week and the week of the last step 1/2 visit. (NCT01641367)
Timeframe: From study entry to Week 48

Interventionpercentage of participants (Number)
Cell Phone Intervention (CPI) + Standard of Care (SOC)3.9
Standard of Care (SOC)1.5

[back to top]

Percent of Participants With Confirmed Virologic Failure by Week 48

Results report percent of participants reaching outcome by week 48 using Kaplan-Meier method. Virologic failure was confirmed by the next HIV-1 RNA measurement ≥1000 copies/mL (irrespective of time between initial and confirmatory measure [specimens on separate dates] and treatment status). A week 24 measurement included HIV-1 RNA obtained ≥7*22=154 days after study entry(to allow for 14 day window for scheduling the visit). HIV-1 RNA measurements through and including 21 November 2016 were considered in identifying an initial failing measurement,allowing HIV-1 RNA at the close-out visit between 22 November 2016 and 13 February 2017 to be a confirmatory measure. Event times were the scheduled week of the initial failing measurement (RNA scheduled at week 0, 12, 24, 48 and every 24 weeks after). Censoring times were the scheduled week of the last RNA result. Length of follow-up varied by Cohort. (NCT01641367)
Timeframe: From week 24 to Week 48

Interventionpercentage of participants (Number)
Experimental: Cohort A48.9
Experimental: Sub-cohort B18.2
Experimental: Sub-cohort B22.9
Experimental: Sub-cohort B30
Experimental: Cohort C5.8
Experimental: Cohort D18.6

[back to top]

Percent of Participants Experiencing Death, AIDS-defining Event or a Non-AIDS-defining Event by Week 48 [CPI+SOC v SOC]

Results report percent of participants reaching outcome by week 48 using Kaplan-Meier method. Event time was the exact week of death, AIDS-defining event or non-AIDS defining event. Censoring time was the earlier of last contact week and the week of the last step 1/2 visit. Only new events were considered, an event that was also reported at or prior to study entry was not included. If a participant experienced multiple events, then the time of the first event was used in the analysis. AIDS defining events included parasitic, fungal, bacterial, and viral infections as well as neoplastic diseases, and neurological disorders. Non-AIDS defining events included malignancies, diabetes, neuropathies, cardiac and renal events. (NCT01641367)
Timeframe: From study entry to Week 48

Interventionpercentage of participants (Number)
Cell Phone Intervention (CPI) + Standard of Care (SOC)8.2
Standard of Care (SOC)6.5

[back to top]

Percent of Participants That Developed Immune Reconstitution Inflammatory Syndrome (IRIS) by Week 48

Results report percent of participants reaching outcome by week 48 using Kaplan-Meier method. Event time was the exact week of the diagnosis. Censoring time was the earlier of last contact week and the week of the last step 1/2 visit. (NCT01641367)
Timeframe: From study entry to week 48

Interventionpercentage of participants (Number)
Experimental: Cohort A0.7
Experimental: Sub-cohort B10
Experimental: Sub-cohort B21.4
Experimental: Sub-cohort B30
Experimental: Cohort C0
Experimental: Cohort D3.0

[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 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]

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]

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]

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 <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]

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 <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]

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]

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]

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]

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 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 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]

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]

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 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]

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]

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]

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 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]

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]

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]

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 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]

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]

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]

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]

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 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]

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]

Change From Baseline in Alkaline Phosphatase (ALP) Levels at Weeks 24 and 48

Change from baseline in ALP at Weeks 24 and 48 was reported. (NCT02431247)
Timeframe: Baseline, Weeks 24 and 48

,
InterventionUnits per liter (U/L) (Mean)
Week 24Week 48
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])-3.2-1.1
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)12.015.1

[back to top]

Change From Baseline in BMD T-score of Hip and Spine

BMD status was assessed using BMD T-scores; normal bone status was defined by a BMD T-score >= -1, osteopenia by a T-score >= -2.5 to <-1.0, and osteoporosis by a T-score <-2.5. (NCT02431247)
Timeframe: Baseline, Weeks 24 and 48

,
InterventionBMD T-score (Mean)
Hip region BMD T-score (Week 24)Spine region BMD T-score (Week 24)Hip region BMD T-score (Week 48)Spine region BMD T-score (Week 48)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])0.019-0.1210.015-0.061
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)-0.109-0.322-0.177-0.225

[back to top]

Change From Baseline in Levels of 25-Hydroxyvitamin D (25-OH Vitamin D), at Week 24 and 48

Change from baseline in 25-OH Vitamin D levels at Week 24 and 48 were reported. (NCT02431247)
Timeframe: Baseline, Weeks 24 and 48

,
Interventionnanomol per liter (nmol/L) (Mean)
Week 24Week 48
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])12.716.9
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)22.128.3

[back to top]

Change From Baseline in Levels of Parathyroid Hormone (PTH) at Weeks 24 and 48

Change from baseline in PTH at Weeks 24 and 48 were reported. (NCT02431247)
Timeframe: Baseline, Weeks 24 and 48

,
InterventionPicomol per liter (pmol/L) (Mean)
Week 24Week 48
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])0.113-0.004
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)0.7770.633

[back to top]

Change From Baseline in Levels of Serum Collagen Type 1 Beta Carboxy Telopeptide (CTX) at Weeks 24 and 48

Change from baseline in serum CTX at Weeks 24 and 48 were reported. (NCT02431247)
Timeframe: Baseline, Weeks 24 and 48

,
Interventionmcg/L (Mean)
Week 24Week 48
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])0.0470.046
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)0.2830.226

[back to top]

Change From Baseline in Levels of Serum Procollagen 1 N-Terminal Propeptide (P1NP) at Weeks 24 and 48

Change from baseline in serum P1NP at Weeks 24 and 48 were reported. (NCT02431247)
Timeframe: Baseline, Weeks 24 and 48

,
Interventionmicrogram per liter (mcg/L) (Mean)
Week 24Week 48
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])1.8920.065
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)24.67924.251

[back to top]

Change From Reference in BMD T-score of Hip and Spine at Week 96

BMD status was assessed using BMD T-scores; normal bone status was defined by a BMD T-score >= -1, osteopenia by a T-score >= -2.5 to <-1.0, and osteoporosis by a T-score <-2.5. Here, reference 1 is the comparative phase baseline value in Week 48 analysis for test group and reference 2 is the last value prior to the switch for Control group. (NCT02431247)
Timeframe: From Reference 1 to Week 96 for D/C/F/TAF Group and Reference 2 to Week 96 for Switch to D/C/F/TAF

,
InterventionBMD T-score (Mean)
Hip region BMD T-scoreSpine region BMD T-score
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])-0.016-0.090
Switch to D/C/F/TAF0.0250.034

[back to top]

Number of Participants With ARV Resistance

Number of participants with DRV, FTC, TDF/TAF resistance were reported. (NCT02431247)
Timeframe: Baseline to end of extension (up to 4 years)

,,
InterventionParticipants (Count of Participants)
DRV resistance-associated mutations (RAMs)TFV RAMsFTC RAMs
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])002
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)001
Switch to D/C/F/TAF002

[back to top]

Percent Change From Reference in Hip and Spine BMD

"The BMD is the amount of mineral in gram per square centimeter of bone, which was assessed by DEXA scan. Positive values are best values and negative values are worst values of change. Percent change from reference in hip and spine BMD was assessed. Here, reference 1 is the comparative phase baseline value in Week 48 analysis for test group and reference 2 is the last value prior to the switch for Control group." (NCT02431247)
Timeframe: From Reference 1 to Week 96 for D/C/F/TAF Group and Reference 2 to Week 96 for Switch to D/C/F/TAF

,
InterventionPercent change in BMD (Mean)
Hip region BMDSpine region BMD
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])-0.2565-0.9349
Switch to D/C/F/TAF0.54670.4829

[back to top]

Percentage of Participants With >95% Treatment Adherence Assessed by Drug Accountability

Treatment adherence assessed by drug accountability (based on pill count) from start of treatment/switch to last study drug intake by determination of the cumulative treatment adherence in participants who returned all dispensed bottles prior to or at the last visit in the study. Adherent participants were defined as having an adherence >95% as assessed by drug accountability. (NCT02431247)
Timeframe: Baseline to Switch and switch to EOE to Open-Label D/C/F/TAF (Up to 3 years)

Interventionpercentage of participants (Number)
Baseline to Switch (double-blind treatment)
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)82.6

[back to top]

Percentage of Participants With >95% Treatment Adherence Assessed by Drug Accountability

Treatment adherence assessed by drug accountability (based on pill count) from start of treatment/switch to last study drug intake by determination of the cumulative treatment adherence in participants who returned all dispensed bottles prior to or at the last visit in the study. Adherent participants were defined as having an adherence >95% as assessed by drug accountability. (NCT02431247)
Timeframe: Baseline to Switch and switch to EOE to Open-Label D/C/F/TAF (Up to 3 years)

Interventionpercentage of participants (Number)
Switch to End of Extension (open-label D/C/F/TAF)
Switch to D/C/F/TAF88.7

[back to top]

Percentage of Participants With >95% Treatment Adherence Assessed by Drug Accountability

Treatment adherence assessed by drug accountability (based on pill count) from start of treatment/switch to last study drug intake by determination of the cumulative treatment adherence in participants who returned all dispensed bottles prior to or at the last visit in the study. Adherent participants were defined as having an adherence >95% as assessed by drug accountability. (NCT02431247)
Timeframe: Baseline to Switch and switch to EOE to Open-Label D/C/F/TAF (Up to 3 years)

Interventionpercentage of participants (Number)
Baseline to Switch (double-blind treatment)Switch to End of Extension (open-label D/C/F/TAF)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])87.292.2

[back to top]

Percentage of Participants With Grade 3 and 4 Adverse Events (AEs), Serious Adverse Events (SAEs), and Premature Discontinuations Due to Adverse Events Through Week 48

AE is any untoward medical occurrence in participant who received study drug without regard to possibility of causal relationship. Events with Grade 3 or higher (3=Severe; 4=life-threatening; 5=fatal) are events that significantly interrupt usual daily activity, require systemic drug therapy/other treatment and are, in many situations, considered unacceptable or intolerable events. SAE is any adverse event (AE) that results in: death, persistent or significant disability/incapacity, requires inpatient hospitalization or prolongation of existing hospitalization, is life-threatening experience, is a congenital anomaly/birth defect and may jeopardize participant and/or may require medical or surgical intervention to prevent one of the outcomes listed above. (NCT02431247)
Timeframe: Up to Weeks 48

,
Interventionpercentage of participants (Number)
Grade 3 AEsGrade 4 AEsSAEsPremature discontinuations due to AEs
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])4.70.64.71.9
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)4.41.75.84.4

[back to top]

Percentage of Participants With Grade 3 and 4 Adverse Events (AEs), Serious Adverse Events (SAEs), and Premature Discontinuations Due to Adverse Events Through Week 96

AE is any untoward medical occurrence in participant who received study drug without regard to possibility of causal relationship. Events with Grade 3 or higher (3=Severe; 4=life-threatening; 5=fatal) are events that significantly interrupt usual daily activity, require systemic drug therapy/other treatment and are, in many situations, considered unacceptable or intolerable events. SAE is any adverse event (AE) that results in: death, persistent or significant disability/incapacity, requires inpatient hospitalization or prolongation of existing hospitalization, is life-threatening experience, is a congenital anomaly/birth defect and may jeopardize participant and/or may require medical or surgical intervention to prevent one of the outcomes listed above. (NCT02431247)
Timeframe: Up to Week 96

,
Interventionpercentage of participants (Number)
Grade 3 AEsGrade 4 AEsSAEsPremature discontinuations due to AEs
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])11.60.810.82.8
Switch to D/C/F/TAF3.71.42.70.3

[back to top]

Percentage of Participants With Grade 3 and 4 AEs, SAEs, and Premature Discontinuations Due to Adverse Events Post-Week 96 to End of Extension

AE is any untoward medical occurrence in participant who received study drug without regard to possibility of causal relationship. Events with Grade 3 or higher (3=Severe; 4=life-threatening; 5=fatal) are events that significantly interrupt usual daily activity, require systemic drug therapy/other treatment and are, in many situations, considered unacceptable or intolerable events. SAE is any adverse event (AE) that results in: death, persistent or significant disability/incapacity, requires inpatient hospitalization or prolongation of existing hospitalization, is life-threatening experience, is a congenital anomaly/birth defect and may jeopardize participant and/or may require medical or surgical intervention to prevent one of the outcomes listed above. (NCT02431247)
Timeframe: From Week 96 to end of extension (up to 3 years)

,
Interventionpercentage of participants (Number)
Grade 3 AEsGrade 4 AEsSAEsPremature discontinuations due to AEs
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])3.503.21.0
Switch to D/C/F/TAF5.21.34.81.3

[back to top]

Percentage of Participants With HIV RNA <50, <20, and <200 Copies/mL Post-week 96 to End of Extension

Percentage of participants with HIV RNA <50, <20, and <200 copies/mL post Week 96 to end of extension were reported. (NCT02431247)
Timeframe: Week 96 to end of extension (up to 3 years)

,
Interventionpercentage of participants (Number)
Week 96 + 6 months (<50 copies/mL)Week 96 + 12 months (<50 copies/mL)Week 96 + 18 months (<50 copies/mL)Week 96 + 24 months (<50 copies/mL)Week 96 + 30 months (<50 copies/mL)Week 96 + 36 months (<50 copies/mL)Week 96 + 6 months (<20 copies/mL)Week 96 + 12 months (<20 copies/mL)Week 96 + 18 months (<20 copies/mL)Week 96 + 24 months (<20 copies/mL)Week 96 + 30 months (<20 copies/mL)Week 96 + 36 months (<20 copies/mL)Week 96 + 6 months (<200 copies/mL)Week 96 + 12 months (<200 copies/mL)Week 96 + 18 months (<200 copies/mL)Week 96 + 24 months (<200 copies/mL)Week 96 + 30 months (<200 copies/mL)Week 96 + 36 months (<200 copies/mL)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])97.799.098.197.594.7100.085.889.792.490.189.594.799.7100.098.797.596.5100.0
Switch to D/C/F/TAF96.396.798.295.791.468.888.291.692.887.084.562.599.399.598.297.898.387.5

[back to top]

Percentage of Participants With HIV-1 RNA < 20, 50, and 200 Copies Per mL at Week 48 and 96 Defined by the Time to Loss of Virologic Response (TLOVR) Algorithm

Percentage of participants with HIV-1 RNA less than (<) 20, 50, and 200 copies per mL at Weeks 48 and 96 based on TLOVR algorithm were assessed. TLOVR requires sustained HIV-1 RNA < 50 copies per mL; confirmed HIV-1 RNA >= 50 copies per mL is considered as non-response (rebound); participant is considered non-responder after permanent discontinuation. (NCT02431247)
Timeframe: At Week 48 and 96

Interventionpercentage of participants (Number)
At Week 48: < 20 Copies per mLAt Week 48: <50 Copies per mLAt Week 48: <200 Copies per mL
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)79.988.791.7

[back to top]

Percentage of Participants With HIV-1 RNA < 20, 50, and 200 Copies Per mL at Week 48 and 96 Defined by the Time to Loss of Virologic Response (TLOVR) Algorithm

Percentage of participants with HIV-1 RNA less than (<) 20, 50, and 200 copies per mL at Weeks 48 and 96 based on TLOVR algorithm were assessed. TLOVR requires sustained HIV-1 RNA < 50 copies per mL; confirmed HIV-1 RNA >= 50 copies per mL is considered as non-response (rebound); participant is considered non-responder after permanent discontinuation. (NCT02431247)
Timeframe: At Week 48 and 96

Interventionpercentage of participants (Number)
At Week 96: <20 Copies per mLAt Week 96: <50 Copies per mLAt Week 96: <200 Copies per mL
Switch to D/C/F/TAF78.493.896.9

[back to top]

Percentage of Participants With HIV-1 RNA < 20, 50, and 200 Copies Per mL at Week 48 and 96 Defined by the Time to Loss of Virologic Response (TLOVR) Algorithm

Percentage of participants with HIV-1 RNA less than (<) 20, 50, and 200 copies per mL at Weeks 48 and 96 based on TLOVR algorithm were assessed. TLOVR requires sustained HIV-1 RNA < 50 copies per mL; confirmed HIV-1 RNA >= 50 copies per mL is considered as non-response (rebound); participant is considered non-responder after permanent discontinuation. (NCT02431247)
Timeframe: At Week 48 and 96

Interventionpercentage of participants (Number)
At Week 48: < 20 Copies per mLAt Week 48: <50 Copies per mLAt Week 48: <200 Copies per mLAt Week 96: <20 Copies per mLAt Week 96: <50 Copies per mLAt Week 96: <200 Copies per mL
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])82.691.293.173.285.186.7

[back to top]

Percentage of Participants With HIV-1 RNA <20 and 200 Copies Per mL at Weeks 48 and 96 Defined by FDA Snapshot Approach

Percentage of participants with HIV-1 RNA < 20/200 copies per mL using FDA snapshot approach were reported. The snapshot approach classified participants into 3 outcome categories: 1) virologic success (HIV RNA < 20/50/200 copies per mL at Week 48 and 96), 2) virologic failure (HIV RNA >= 20/50/200 copies per mL at Week 48 and 96), 3) no viral load data in the Week 48 and 96 visit window (discontinued due to adverse event/death/other reason). The missing HIV-1 RNA is considered as non-response. (NCT02431247)
Timeframe: At Weeks 48 and 96

Interventionpercentage of participants (Number)
At week 48: <20 Copies per mLAt week 48: <200 Copies per mL
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)79.390.6

[back to top]

Percentage of Participants With HIV-1 RNA <20 and 200 Copies Per mL at Weeks 48 and 96 Defined by FDA Snapshot Approach

Percentage of participants with HIV-1 RNA < 20/200 copies per mL using FDA snapshot approach were reported. The snapshot approach classified participants into 3 outcome categories: 1) virologic success (HIV RNA < 20/50/200 copies per mL at Week 48 and 96), 2) virologic failure (HIV RNA >= 20/50/200 copies per mL at Week 48 and 96), 3) no viral load data in the Week 48 and 96 visit window (discontinued due to adverse event/death/other reason). The missing HIV-1 RNA is considered as non-response. (NCT02431247)
Timeframe: At Weeks 48 and 96

Interventionpercentage of participants (Number)
At week 96: <20 Copies per mLAt week 96: <200 Copies per mL
Switch to D/C/F/TAF83.596.9

[back to top]

Percentage of Participants With HIV-1 RNA <20 and 200 Copies Per mL at Weeks 48 and 96 Defined by FDA Snapshot Approach

Percentage of participants with HIV-1 RNA < 20/200 copies per mL using FDA snapshot approach were reported. The snapshot approach classified participants into 3 outcome categories: 1) virologic success (HIV RNA < 20/50/200 copies per mL at Week 48 and 96), 2) virologic failure (HIV RNA >= 20/50/200 copies per mL at Week 48 and 96), 3) no viral load data in the Week 48 and 96 visit window (discontinued due to adverse event/death/other reason). The missing HIV-1 RNA is considered as non-response. (NCT02431247)
Timeframe: At Weeks 48 and 96

Interventionpercentage of participants (Number)
At week 48: <20 Copies per mLAt week 48: <200 Copies per mLAt week 96: <20 Copies per mLAt week 96: <200 Copies per mL
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])82.692.876.286.2

[back to top]

Percentage of Participants With Non-PDVF by Kaplan-Meier Estimates

Percentage of participants with non-PDVF by Kaplan-Meier Estimates were reported. PDVF was defined as having virologic non-response (HIV-1 RNA <1 log10 reduction from baseline and >=50 copies/mL at the Week 8 visit, confirmed at the next visit), virologic rebound (confirmed HIV-1 RNA >=50 copies/mL after confirmed consecutive HIV-1 RNA <50 copies/mL or confirmed >1 log10 increase in HIV-1 RNA from nadir), or viremic at final time point (final available on treatment HIV-1 RNA >=400 copies/mL). (NCT02431247)
Timeframe: From Week 96 to end of extension (up to 2 years and 6 months)

,
Interventionpercentage of participants (Number)
Week 96Week 96 + 6 monthsWeek 96 + 12 monthsWeek 96 + 18 monthsWeek 96 + 24 monthsWeek 96 + 30 months
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])10099.699.698.697.397.3
Switch to D/C/F/TAF10099.299.297.897.892.5

[back to top]

Percentage of Participants With PDVF Post-week 96 to End of Extension

Percentage of participants with PDVF were reported. Protocol-defined virologic failure was defined as having virologic non-response (HIV-1 RNA <1 log10 reduction from baseline and >=50 copies/mL at the Week 8 visit, confirmed at the next visit), virologic rebound (confirmed HIV-1 RNA >=50 copies/mL after confirmed consecutive HIV-1 RNA <50 copies/mL or confirmed >1 log10 increase in HIV-1 RNA from nadir), or viremic at final time point (final available on treatment HIV-1 RNA >=400 copies/mL). (NCT02431247)
Timeframe: Week 96 to end of extension (up to 3 years)

,
Interventionpercentage of participants (Number)
Participants who met PDVFVirologic non-responseVirologic reboundViremic at final time point
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])1.001.00
Switch to D/C/F/TAF2.101.40.7

[back to top]

Percentage of Participants With Protocol-defined Virologic Failure (PDVF)

Percentage of participants with PDVF were reported. Protocol-defined virologic failure was defined as having virologic non-response (HIV-1 RNA <1 log10 reduction from baseline and >=50 copies/mL at the Week 8 visit, confirmed at the next visit), virologic rebound (confirmed HIV-1 RNA >=50 copies/mL after confirmed consecutive HIV-1 RNA <50 copies/mL or confirmed >1 log10 increase in HIV-1 RNA from nadir), or viremic at final time point (final available on treatment HIV-1 RNA >=400 copies/mL). (NCT02431247)
Timeframe: From Baseline up to Week 96

Interventionpercentage of participants (Number)
Participants who met PDVF (Baseline - Switch)Virologic non-response (Baseline - Switch)Virologic rebound (Baseline - Switch)Viremic at final time point (Baseline - Switch)
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)4.403.90.6

[back to top]

Percentage of Participants With Protocol-defined Virologic Failure (PDVF)

Percentage of participants with PDVF were reported. Protocol-defined virologic failure was defined as having virologic non-response (HIV-1 RNA <1 log10 reduction from baseline and >=50 copies/mL at the Week 8 visit, confirmed at the next visit), virologic rebound (confirmed HIV-1 RNA >=50 copies/mL after confirmed consecutive HIV-1 RNA <50 copies/mL or confirmed >1 log10 increase in HIV-1 RNA from nadir), or viremic at final time point (final available on treatment HIV-1 RNA >=400 copies/mL). (NCT02431247)
Timeframe: From Baseline up to Week 96

Interventionpercentage of participants (Number)
Participants who met PDVF (Baseline - Week 96)Virologic non-response (Baseline - Week 96)Virologic rebound (Baseline-Week 96)Viremic at final time point (Baseline-Week 96)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])4.10.60.30.6

[back to top]

Percentage of Participants With Protocol-defined Virologic Failure (PDVF)

Percentage of participants with PDVF were reported. Protocol-defined virologic failure was defined as having virologic non-response (HIV-1 RNA <1 log10 reduction from baseline and >=50 copies/mL at the Week 8 visit, confirmed at the next visit), virologic rebound (confirmed HIV-1 RNA >=50 copies/mL after confirmed consecutive HIV-1 RNA <50 copies/mL or confirmed >1 log10 increase in HIV-1 RNA from nadir), or viremic at final time point (final available on treatment HIV-1 RNA >=400 copies/mL). (NCT02431247)
Timeframe: From Baseline up to Week 96

Interventionpercentage of participants (Number)
Participants who met PDVF (Switch - Week 96)Virologic non-response (Switch - Week 96)Virologic rebound (Switch - Week 96)Viremic at final time point (Switch - Week 96)
Switch to D/C/F/TAF1.101.10

[back to top]

Percentage of Participants With Time to Treatment Failure by Kaplan-Meier Estimates

Percentage of participants with time to treatment failure by Kaplan-Meier Estimates were reported. Treatment failure was defined as having either protocol-defined virologic failure or having discontinued for reasons other than alternate access to D/C/F/TAF (or other ARVs). (NCT02431247)
Timeframe: From Week 96 to end of extension (up to 3 years)

Interventionpercentage of participants (Number)
Week 96Week 96 + 6 monthsWeek 96 + 12 monthsWeek 96 + 18 monthsWeek 96 + 24 monthsWeek 96 + 30 months
Switch to D/C/F/TAF10097.494.189.586.479.1

[back to top]

Percentage of Participants With Time to Treatment Failure by Kaplan-Meier Estimates

Percentage of participants with time to treatment failure by Kaplan-Meier Estimates were reported. Treatment failure was defined as having either protocol-defined virologic failure or having discontinued for reasons other than alternate access to D/C/F/TAF (or other ARVs). (NCT02431247)
Timeframe: From Week 96 to end of extension (up to 3 years)

Interventionpercentage of participants (Number)
Week 96Week 96 + 6 monthsWeek 96 + 12 monthsWeek 96 + 18 monthsWeek 96 + 24 monthsWeek 96 + 30 monthsWeek 96 + 36 months
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])10098.995.690.687.184.884.8

[back to top]

Change From Baseline in Serum Creatinine at Week 48

Change from baseline in serum creatinine at Week 48 was reported. (NCT02431247)
Timeframe: Baseline and Week 48

Interventionmilligram per deciliter (mg/dL) (Least Squares Mean)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])0.05
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)0.09

[back to top]

Percent Change From Baseline in Hip and Spine Bone Mineral Density (BMD)

"The BMD is the amount of mineral in gram per square centimeter of bone, which was assessed by dual energy x-ray absorptiometry (DEXA) scan. Positive values are best values and negative values are worst values of change. Percent change from baseline in hip and spine BMD was assessed." (NCT02431247)
Timeframe: Baseline, Weeks 24 and 48

,
InterventionPercent change (Least Squares Mean)
Hip region BMD (Week 24)Spine region BMD (Week 24)Hip region BMD (Week 48)Spine region BMD (Week 48)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])0.29-1.340.17-0.68
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)-1.66-3.43-2.69-2.38

[back to top]

Area Under the Plasma Concentration Time Curve Across the Dosing Interval (AUCtau) of Tenofovir Alafenamide

The AUCtau is the measure of the plasma drug concentration from time zero to end of dosing interval. It is used to characterize drug absorption. (NCT02431247)
Timeframe: 30 minutes to 4 hours postdose at Weeks 2, 4, 12, 24 and 48 and at 2 timepoints with at least 2.5 hours in between sampling at Week 8 and 36 (first sample between 1 and 4 hours postdose)

Interventionh*ng/mL (Mean)
Tenofovir Alafenamide 10 mg [D/C/F/TAF (Test)]132.3117

[back to top]

Area Under the Plasma Concentration-Time Curve From Time of Administration to 24 Hours Post-dose (AUC0-24h) of Darunavir

AUC (0-24) is the area under the plasma concentration-time curve from time zero to 24 hours post-dose. (NCT02431247)
Timeframe: 0 to 24 hours post dose

Interventionhours*nanogram per milliliter (h*ng/mL) (Mean)
Darunavir 800 mg [D/C/F/TAF]87909.3282

[back to top]

Change From Baseline in Cluster of Differentiation-4 (CD4+) Cell Count at Week 48

The immunologic change was determined by changes in Cluster of CD4+ cell count. Change from baseline in CD4+ cell count at Week 48 were assessed. (NCT02431247)
Timeframe: Baseline and Week 48

InterventionCells per millimeter cube (cells/mm^3) (Least Squares Mean)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])190.49
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)172.01

[back to top]

Change From Baseline in Estimated Glomerular Filtration Rate Based on Serum Creatinine (eGFRcr) by Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) Formula at Week 48

Change from baseline in eGFRcr was calculated using the CKD-EPI equation as per which Stage 1 (normal or high GFR [>=90 mL/min]); Stage 2 (Mild CKD [60 to 90 mL/min]); Stage 3 (Moderate CKD [30 to 59mL/min]); Stage 4 (Severe CKD [15 to 29 mL/min]); Stage 5 (End Stage CKD [<15 mL/min]). The eGFRcr was assessed by calculating serum creatinine (Scr) using the equation: eGFRcr milliliter per minute per 1.72 meter square (mL/min/1.73m^2) = 144*(Scr/0.7)^-0.329*0.993^age (Scr =< 0.7 mg/dL) and eGFRcr mL/min/1.73m^2 = 144*(Scr/0.7)^-1.209*0.993^age (Scr >0.7 mg/dL) for female participants and eGFRcr mL/min/1.73m^2 = 141*(Scr/0.9)^-0.411*0.993^age (Scr =<0.9 mg/dL) and eGFRcr mL/min/1.73m^2 = 141*(Scr/0.9)^-1.209*0.993^age (Scr >0.9 mg/dL) for male participants. (NCT02431247)
Timeframe: Baseline and Week 48

InterventionmL/min/1.73 m^2 (Least Squares Mean)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])-6.04
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)-9.16

[back to top]

Change From Baseline in Estimated Glomerular Filtration Rate Based on Serum Creatinine by (Cockcroft-Gault Formula) at Week 48

Change from baseline in eGFRcr by cockcroft-gault formula at Week 48. The eGFRcr was assessed by calculated creatinine clearance (CrCl) using the Cockcroft-Gault formula, and was assessed using CrCl [mL/min] = (140 - A) * W / (72 * C) * R. Where A is age at sample date [years], W is body weight at specific visit (kilogram [kg]), C is the serum concentration of creatinine [mg/dL], R = 1 if the participant is male and = 0.85 if female. (NCT02431247)
Timeframe: Baseline and Week 48

Interventionmilliliter per minute (mL/min) (Least Squares Mean)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])-5.16
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)-11.20

[back to top]

Plasma Concentrations 2 Hours After Dosing (C0-2h) of Tenofovir Alafenamide

C0-2h is defined as the plasma concentrations 2 hours after dosing. (NCT02431247)
Timeframe: 0 to 2 hours post dose

Interventionng/mL (Mean)
Tenofovir Alafenamide 10 mg [D/C/F/TAF (Test)]11.9785

[back to top]

Change From Baseline in Estimated Glomerular Filtration Rate Based on Serum Cystatin C (eGFRcyst) by CKD-EPI Formula at Week 48

Change from baseline in eGFRcyst was calculated using the CKD-EPI equation as per which Stage 1 (normal or high GFR) >= 90 indicates normal kidney function; Stage 2 (Mild CKD): 60 to 89 mL/min indicates mildly reduced kidney function; Stage 3 (Moderate CKD): 30 to 59 mL/min indicates moderately reduced kidney function; Stage 4 (Severe CKD): 15 to 29 mL/min indicates severely reduced kidney function; Stage 5 (End Stage of CKD): <15 mL/min indicate very severe or end stage kidney failure. The eGFRcyst was assessed by calculated serum cystatin C (Scyst) using the equation: eGFRcyst mL/min/1.73m^2 = 133 * (Scyst/0,8)^-0.499 * 0.996^age [* 0.932 if female] (Scyst =<0.8 mg/L) and eGFRcr mL/min/1.73m^2 = 133 * (Scyst/0,8)^-1.328 * 0.996^age [* 0.932 if male] (Scyst >0.8 mg/L). (NCT02431247)
Timeframe: Baseline and Week 48

InterventionmL/min/1.73 m^2 (Least Squares Mean)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])5.32
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)2.92

[back to top]

Change From Baseline in log10 HIV-1 RNA Levels at Week 48

Change from baseline in log10 HIV-1 RNA levels were reported. (NCT02431247)
Timeframe: Baseline and Week 48

Interventionlog10 HIV-1 RNA copies per mL (Least Squares Mean)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])-2.95
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)-2.91

[back to top]

Change From Baseline in Urine Albumin to Creatinine Ratio (UACR) at Week 48

Change from baseline in UACR at Week 48 was reported. (NCT02431247)
Timeframe: Baseline and Week 48

Interventionmg/g (Median)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])-0.58
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)-0.15

[back to top]

Change From Baseline in Urine Beta-2 Microglobulin to Creatinine Ratio (UB2MGCR) at Week 48

Change from baseline in UB2MGCR at Week 48 were reported. (NCT02431247)
Timeframe: Baseline and Week 48

Interventionmcg/g (Median)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])-30.42
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)18.36

[back to top]

Change From Baseline in Urine Protein to Creatinine Ratio (UPCR) at Week 48

Change from baseline in UPCR at Week 48 was reported. (NCT02431247)
Timeframe: Baseline and Week 48

Interventionmilligram per gram (mg/g) (Median)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])-15.72
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)-10.53

[back to top]

Change From Baseline in Urine Retinol Binding Protein To Creatinine Ratio (URBPCR) at Week 48

Change from baseline in URBPCR at Week 48 were reported. (NCT02431247)
Timeframe: Baseline and Week 48

Interventionmicrogram per gram (mcg/g) (Median)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])7.00
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)35.02

[back to top]

Change From Reference in ALP Levels

Change from reference in ALP levels at Week 96 was reported. Here, reference 1 is the comparative phase baseline value in Week 48 analysis for test group and reference 2 is the last value prior to the switch for Control group. (NCT02431247)
Timeframe: From Reference 1 to Week 96 for D/C/F/TAF Group and Reference 2 to Week 96 for Switch to D/C/F/TAF

InterventionU/L (Mean)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])-0.9
Switch to D/C/F/TAF-9.7

[back to top]

Change From Reference in CD4+ Cell Count at Week 96

The immunologic change was determined by changes in cluster of differentiation (CD4+) cell count. Change from reference in CD4+ cell count at Week 96 was assessed. Here, reference 1 is the comparative phase baseline value in Week 48 analysis for test group and reference 2 is the last value prior to the switch for Control group. (NCT02431247)
Timeframe: From Reference 1 to Week 96 for D/C/F/TAF Group and Reference 2 to Week 96 for Switch to D/C/F/TAF

Interventioncells/mm^3 (Least Squares Mean)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])228.85
Switch to D/C/F/TAF27.01

[back to top]

Change From Reference in eGFRcr by CKD-EPI Formula

Change from reference in eGFRcr was calculated using the CKD-EPI equation as per Stage 1 (normal or high GFR) to Stage 5 (End Stage of CKD). The eGFRcr was assessed by calculating serum creatinine (Scr) using the CKD-EPI equation: eGFRcr milliliter per minute per 1.72 meter square (mL/min/1.73m^2) = 144 * (Scr/0.7)^-0.329 * 0.993^age (Scr =< 0.7 mg/dL) and eGFRcr mL/min/1.73m^2 = 144 * (Scr/0.7)^-1.209 * 0.993^age (Scr >0.7 mg/dL) for female participants and eGFRcr mL/min/1.73m^2 = 141 x (Scr/0.9)^-0.411 x 0.993^age (Scr =<0.9 mg/dL) and eGFRcr mL/min/1.73m^2 = 141 * (Scr/0.9)^-1.209 x 0.993^age (Scr >0.9 mg/dL) for male participants. Here, reference 1 is the comparative phase baseline value in Week 48 analysis for test group and reference 2 is the last value prior to the switch for Control group. (NCT02431247)
Timeframe: From Reference 1 to Week 96 for D/C/F/TAF Group and Reference 2 to Week 96 for Switch to D/C/F/TAF

InterventionmL/min/1.73 m^2 (Median)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])-5.6
Switch to D/C/F/TAF2.3

[back to top]

Change From Reference in Estimated Glomerular Filtration Rate Based on Serum Creatinine by Cockcroft-Gault Formula

Change from reference in eGFRcr by cockcroft-gault formula was reported. The eGFRcr was assessed by calculated creatinine clearance (CrCl) using the Cockcroft-Gault formula, and was assessed using CrCl [mL/min] = (140 - A) * W / (72 * C) * R. Where A is age at sample date [years], W is body weight at specific visit (kilogram [kg]), C is the serum concentration of creatinine [mg/dL], R = 1 if the participant is male and = 0.85 if female. Here, reference 1 is the comparative phase baseline value in Week 48 analysis for test group and reference 2 is the last value prior to the switch for Control group. (NCT02431247)
Timeframe: From Reference 1 to Week 96 for D/C/F/TAF Group and Reference 2 to Week 96 for Switch to D/C/F/TAF

InterventionmL/min (Median)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])-5.2
Switch to D/C/F/TAF4.6

[back to top]

Change From Reference in Estimated Glomerular Filtration Rate Based on Serum Cystatin C (eGFRcyst) by CKD-EPI Formula

Change from reference in eGFRcyst was calculated using the CKD-EPI equation as per Stage 1 (normal or high GFR) to Stage 5 (End Stage of CKD): <15 mL/min indicate very severe or end stage kidney failure. The eGFRcyst was assessed by calculated serum cystatin C (Scyst) using the CKD-EPI equation: eGFRcyst mL/min/1.73m^2 = 133 * (Scyst/0,8)^-0.499 * 0.996^age [x 0.932 if female] (Scyst =<0.8 mg/L) and eGFRcr mL/min/1.73m^2 = 133 * (Scyst/0,8)^-1.328 * 0.996^age [* 0.932 if male] (Scyst >0.8 mg/L). Here, reference 1 is the comparative phase baseline value in Week 48 analysis for test group and reference 2 is the last value prior to the switch for Control group. (NCT02431247)
Timeframe: From Reference 1 to Week 96 for D/C/F/TAF Group and Reference 2 to Week 96 for Switch to D/C/F/TAF

InterventionmL/min/1.73 m^2 (Median)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])4.4
Switch to D/C/F/TAF0

[back to top]

Change From Reference in Levels of 25-OH Vitamin D

Change from reference in 25-OH Vitamin D were reported. Here, reference 1 is the comparative phase baseline value in Week 48 analysis for test group and reference 2 is the last value prior to the switch for Control group. (NCT02431247)
Timeframe: From Reference 1 to Week 96 for D/C/F/TAF Group and Reference 2 to Week 96 for Switch to D/C/F/TAF

Interventionnmol/L (Mean)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])21.3
Switch to D/C/F/TAF-10.3

[back to top]

Change From Reference in Levels of PTH

Change from reference in PTH levels were reported. Here, reference 1 is the comparative phase baseline value in Week 48 analysis for test group and reference 2 is the last value prior to the switch for Control group. (NCT02431247)
Timeframe: From Reference 1 to Week 96 for D/C/F/TAF Group and Reference 2 to Week 96 for Switch to D/C/F/TAF

Interventionpmol/L (Mean)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])-0.290
Switch to D/C/F/TAF-1.283

[back to top]

Change From Reference in Levels of Serum CTX

Change from reference in serum CTX levels were reported. Here, reference 1 is the comparative phase baseline value in Week 48 analysis for test group and reference 2 is the last value prior to the switch for Control group. (NCT02431247)
Timeframe: From Reference 1 to Week 96 for D/C/F/TAF Group and Reference 2 to Week 96 for Switch to D/C/F/TAF

Interventionmcg/L (Mean)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])0.041
Switch to D/C/F/TAF-0.162

[back to top]

Change From Reference in Levels of Serum P1NP

Change from reference in serum P1NP levels were reported. Here, reference 1 is the comparative phase baseline value in Week 48 analysis for test group and reference 2 is the last value prior to the switch for Control group. (NCT02431247)
Timeframe: From Reference 1 to Week 96 for D/C/F/TAF Group and Reference 2 to Week 96 for Switch to D/C/F/TAF

Interventionmcg/L (Mean)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])2.817
Switch to D/C/F/TAF-11.963

[back to top]

Change From Reference in log10 HIV-1 RNA Levels at Week 96

Change from reference in log10 HIV-1 RNA levels were reported. Here, reference 1 is the comparative phase baseline value in Week 48 analysis for Test group and reference 2 is the last value prior to the switch for Control group. (NCT02431247)
Timeframe: From Reference 1 to Week 96 for D/C/F/TAF Group and Reference 2 to Week 96 for Switch to D/C/F/TAF

Interventionlog10 HIV-1 RNA copies per mL (Least Squares Mean)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])-2.72
Switch to D/C/F/TAF-0.0027

[back to top]

Change From Reference in Serum Creatinine

Change from reference in serum creatinine was reported. Here, reference 1 is the comparative phase baseline value in Week 48 analysis for test group and reference 2 is the last value prior to the switch for Control group. (NCT02431247)
Timeframe: From Reference 1 to Week 96 for D/C/F/TAF Group and Reference 2 to Week 96 for Switch to D/C/F/TAF

Interventionmg/dL (Median)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])0.045
Switch to D/C/F/TAF-0.034

[back to top]

Change From Reference in UACR

Change from reference in UACR at Week 96 was reported. Here, reference 1 is the comparative phase baseline value in Week 48 analysis for test group and reference 2 is the last value prior to the switch for Control group. (NCT02431247)
Timeframe: From Reference 1 to Week 96 for D/C/F/TAF Group and Reference 2 to Week 96 for Switch to D/C/F/TAF

Interventionmg/g (Median)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])-0.70
Switch to D/C/F/TAF-0.49

[back to top]

Change From Reference in UB2MGCR

Change from reference in UB2MGCR was reported. Here, reference 1 is the comparative phase baseline value in Week 48 analysis for test group and reference 2 is the last value prior to the switch for Control group. (NCT02431247)
Timeframe: From Reference 1 to Week 96 for D/C/F/TAF Group and Reference 2 to Week 96 for Switch to D/C/F/TAF

Interventionmcg/g (Median)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])-27.04
Switch to D/C/F/TAF-40.53

[back to top]

Change From Reference in UPCR

Change from reference in UPCR was reported. Here, reference 1 is the comparative phase baseline value in Week 48 analysis for test group and reference 2 is the last value prior to the switch for Control group. (NCT02431247)
Timeframe: From Reference 1 to Week 96 for D/C/F/TAF+ FTC/TDF Group and Reference 2 to Week 96 for Switch to D/C/F/TAF

Interventionmg/g (Median)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])-15.46
Switch to D/C/F/TAF-1.40

[back to top]

Change From Reference in URBPCR

Change from reference in URBPCR at Week 96 were reported. Here, reference 1 is the comparative phase baseline value in Week 48 analysis for test group and reference 2 is the last value prior to the switch for Control group. (NCT02431247)
Timeframe: From Reference 1 to Week 96 for D/C/F/TAF Group and Reference 2 to Week 96 for Switch to D/C/F/TAF

Interventionmcg/g (Median)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])13.70
Switch to D/C/F/TAF-35.53

[back to top]

Percent Change From Baseline in Urine Fractional Excretion of Phosphate (FEPO4) at Week 48

Percent change from baseline in FEPO4 at Week 48 was reported. (NCT02431247)
Timeframe: Baseline and Week 48

InterventionPercent change (Median)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])16.00
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)22.55

[back to top]

Percent Change From Reference in Urine FEPO4

Percent change from reference in FEPO4 at Week 96 were reported. Here, reference 1 is the comparative phase baseline value in Week 48 analysis for test group and reference 2 is the last value prior to the switch for Control group. (NCT02431247)
Timeframe: From Reference 1 to Week 96 for D/C/F/TAF Group and Reference 2 to Week 96 for Switch to D/C/F/TAF

InterventionPercent change in urine FEPO4 (Median)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])18.52
Switch to D/C/F/TAF-7.51

[back to top]

Percentage of Participants With HIV-1 RNA <50 Copies Per mL at Week 96 Defined by FDA Snapshot Approach

Percentage of participants with a HIV-1 RNA < 50 copies per mL were assessed using FDA snapshot approach which defines a participant's virologic response status using only the viral load at the predefined time point within an allowed window of time, along with study drug discontinuation status. The snapshot approach classified participants into 3 outcome categories: 1) virologic success (HIV RNA < 20/50/200 copies per mL at Week 96), 2) virologic failure (HIV RNA greater than or equal to [>=] 20/50/200 copies per mL at Week 96), 3) no viral load data in the Week 96 visit window (discontinued due to adverse event/death/other reason). The missing HIV-1 RNA is considered as non-response. (NCT02431247)
Timeframe: At Week 96

Interventionpercentage of participants (Number)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])85.1
Switch to D/C/F/TAF94.2

[back to top]

Percentage of Participants With Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA) Less Than (<) 50 Copies Per Milliliter (Copies Per mL) (Virologic Response) at Week 48 Defined by Food and Drug Administration (FDA) Snapshot Approach

Percentage of participants with a HIV-1 RNA < 50 copies per mL were assessed using FDA snapshot approach which defines a participant's virologic response status using only the viral load at the predefined time point within an allowed window of time, along with study drug discontinuation status. The snapshot approach classified participants into 3 outcome categories: 1) virologic success (HIV RNA < 20/50/200 copies per mL at Week 48), 2) virologic failure (HIV RNA greater than or equal to [>=] 20/50/200 copies per mL at Week 48), 3) no viral load data in the Week 48 visit window (discontinued due to adverse event/death/other reason). The missing HIV-1 RNA is considered as non-response. (NCT02431247)
Timeframe: At Week 48

Interventionpercentage of participants (Number)
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])91.4
DRV/COBI+ FTC/TDF (Control) (Baseline to Switch)88.4

[back to top]

Predose (Trough) Plasma Concentration (C0h) of Darunavir

C0h is defined as the predose (trough) plasma concentration or concentration just prior to study drug administration. (NCT02431247)
Timeframe: 30 minutes to 4 hours postdose at Weeks 2, 4, 12, 24 and 48 and at 2 timepoints with at least 2.5 hours in between sampling at Week 8 and 36 (first sample between 1 and 4 hours postdose)

Interventionnanogram per milliliter (ng/mL) (Mean)
Darunavir 800 mg [D/C/F/TAF (Test)]1898.9100

[back to top]

CD4+ Cell Count Post-Week From 96 to End of Extension

The immunologic change was determined by Cluster of CD4+ cell count. CD4+ cell count post-Week 96 to end of extension were assessed. (NCT02431247)
Timeframe: Week 96 to end of extension (up to 3 years)

,
Interventioncells/mm^3 (Mean)
Week 96 + 6 monthsWeek 96 + 12 monthsWeek 96 + 18 monthsWeek 96 + 24 monthsWeek 96 + 30 monthsWeek 96 + 36 months
D/C/F/TAF (Test) (Baseline to End of Extension [EOE])790.2779.4789.8781.9741.6784.7
Switch to D/C/F/TAF749.7774.3758.4784.1736.7778.4

[back to top]

Part 1: Time to Reach Maximum Observed Plasma Concentration (Tmax) of Maraviroc

(NCT02625207)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, 9, 12 hours post-dose on Day 5

Interventionhour (Median)
Cohort 1- No CYP3A5*1 Alleles3.00
Cohort 2- One CYP3A5*1 Allele2.00
Cohort 3- Two CYP3A5*1 Alleles2.00
Cohort 4- No CYP3A5*1 Alleles2.01

[back to top]

Part 1: Time to Reach Maximum Observed Plasma Concentration (Tmax) of Metabolites of Maraviroc

Metabolites of maraviroc included PF-6857639, PF-6857640, PF-06927572 and PF-06927573. (NCT02625207)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, 9, 12 hours post-dose on Day 5

,,,
Interventionhour (Median)
PF-6857639PF-6857640PF-06927572PF-06927573
Cohort 1- No CYP3A5*1 Alleles3.003.003.003.00
Cohort 2- One CYP3A5*1 Allele2.002.002.002.00
Cohort 3- Two CYP3A5*1 Alleles2.002.001.522.00
Cohort 4- No CYP3A5*1 Alleles3.003.003.003.00

[back to top]

Part 1: Plasma Concentration of Metabolites of Maraviroc at 12 Hour Post-dose

Metabolites of maraviroc included PF-6857639, PF-6857640, PF-06927572 and PF-06927573. (NCT02625207)
Timeframe: 12 hour post-dose on Day 5

,,,
Interventionng/mL (Geometric Mean)
PF-6857639PF-6857640PF-06927572PF-06927573
Cohort 1- No CYP3A5*1 Alleles0.62490.62900.62420.5396
Cohort 2- One CYP3A5*1 Allele2.7481.6581.7521.629
Cohort 3- Two CYP3A5*1 Alleles2.2261.1341.0710.8550
Cohort 4- No CYP3A5*1 Alleles0.92131.3001.3851.175

[back to top]

Part 1: Number of Participants With Treatment Emergent Adverse Events (AEs) and Serious Adverse Events (SAEs)

An AE was any untoward medical occurrence in a participant who received study drug without regard to possibility of causal relationship. SAE was 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. Treatment-emergent are events between first dose of study drug and up to 6 days that were absent before treatment or that worsened relative to pre-treatment state. (NCT02625207)
Timeframe: Baseline up to end of study (up to 6 days)

,,,
Interventionparticipants (Number)
AEsSAEs
Cohort 1- No CYP3A5*1 Alleles30
Cohort 2- One CYP3A5*1 Allele00
Cohort 3- Two CYP3A5*1 Alleles00
Cohort 4- No CYP3A5*1 Alleles30

[back to top]

Part 1: Metabolite to Parent Ratio for Area Under the Concentration-Time Curve From Time 0 to 12 Hours for Maraviroc and Its Metabolites (MRAUC12)

MRAUC12 is the ratio of AUC12 of maraviroc to AUC12 of maraviroc's metabolites. Metabolites of Maraviroc included PF-6857639, PF-6857640, PF-06927572 and PF-06927573. AUC12 is the area under the plasma concentration-time profile from time 0 to 12 hours post-dose. (NCT02625207)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, 9, 12 hours post-dose on Day 5

,,,
Interventionratio (Geometric Mean)
PF-6857639PF-6857640PF-06927572PF-06927573
Cohort 1- No CYP3A5*1 Alleles0.021720.025770.027660.02048
Cohort 2- One CYP3A5*1 Allele0.035640.026210.029080.02427
Cohort 3- Two CYP3A5*1 Alleles0.043120.027970.027950.02040
Cohort 4- No CYP3A5*1 Alleles0.015850.023380.027740.02099

[back to top]

Part 1: Maximum Observed Plasma Concentration (Cmax) of Metabolites of Maraviroc

Metabolites of maraviroc included PF-6857639, PF-6857640, PF-06927572 and PF-06927573. (NCT02625207)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, 9, 12 hours post-dose on Day 5

,,,
Interventionng/mL (Geometric Mean)
PF-6857639PF-6857640PF-06927572PF-06927573
Cohort 1- No CYP3A5*1 Alleles18.1421.9522.8817.39
Cohort 2- One CYP3A5*1 Allele24.1818.9620.6717.64
Cohort 3- Two CYP3A5*1 Alleles20.5215.0015.3110.86
Cohort 4- No CYP3A5*1 Alleles11.2216.4819.9115.27

[back to top]

Part 2: Plasma Concentration of Maraviroc at 24 Hours Post-dose

(NCT02625207)
Timeframe: 24 hours post-dose on Day 10

Interventionng/mL (Geometric Mean)
Cohort 1- No CYP3A5*1 Alleles56.56
Cohort 3- Two CYP3A5*1 Alleles56.34

[back to top]

Part 1: Average Plasma Concentration (Cav) of Metabolites of Maraviroc

Cavg is the average plasma concentration of metabolites of maraviroc during the 0 to 12 hour time period. It was calculated as area under the plasma concentration-time curve from 0 to 12 hours (AUC [0-12]) divided by 12. Metabolites of maraviroc included PF-6857639, PF-6857640, PF-06927572 and PF-06927573. (NCT02625207)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, 9, 12 hours post-dose on Day 5

,,,
Interventionng/mL (Geometric Mean)
PF-6857639PF-6857640PF-06927572PF-06927573
Cohort 1- No CYP3A5*1 Alleles6.4207.6158.1796.056
Cohort 2- One CYP3A5*1 Allele9.0496.6587.3836.165
Cohort 3- Two CYP3A5*1 Alleles8.0745.2395.2323.821
Cohort 4- No CYP3A5*1 Alleles4.0185.9307.0295.323

[back to top]

Part 1: Area Under The Plasma Concentration-Time Curve From Time 0 to 12 Hours (AUC [0-12]) of Metabolites of Maraviroc

AUC (0-12) is the area under the plasma concentration versus time curve from time zero (pre-dose) to 12 hours post-dose. Metabolites of maraviroc included PF-6857639, PF-6857640, PF-06927572 and PF-06927573. (NCT02625207)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, 9, 12 hours post-dose on Day 5

,,,
Interventionng*hr/mL (Geometric Mean)
PF-6857639PF-6857640PF-06927572PF-06927573
Cohort 1- No CYP3A5*1 Alleles77.0991.4298.1472.67
Cohort 2- One CYP3A5*1 Allele108.779.9288.6373.92
Cohort 3- Two CYP3A5*1 Alleles96.9862.8662.8045.86
Cohort 4- No CYP3A5*1 Alleles48.2471.1584.3263.88

[back to top]

Part 2: Time to Reach Maximum Observed Plasma Concentration (Tmax) of Maraviroc

(NCT02625207)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, 9, 12, 24 hours post-dose on Day 10

Interventionhour (Median)
Cohort 1- No CYP3A5*1 Alleles3.00
Cohort 3- Two CYP3A5*1 Alleles3.02

[back to top]

Part 2: Number of Participants With Laboratory Abnormalities

Criteria: Hemoglobin; hematocrit; red blood cell count: <0.8*LLN, mean corpuscular volume; mean corpuscular hemoglobin concentration; mean platelet volume: <0.9*LLN or >1.1*ULN, platelet: <0.5*LLN or >1.75*ULN, white blood cells <0.6*LLN or >1.5*ULN, lymphocyte; neutrophil: <0.8*LLN or >1.2*ULN, basophil; eosinophil; monocyte: >1.2*ULN, bilirubin (total, direct, indirect) >1.5*ULN, aspartate aminotransferase; alanine aminotransferase; alkaline phosphatase: >3.0*ULN, total protein; albumin: <0.8*LLN or >1.2*ULN; creatinine: >1.3*ULN, uric acid >1.2*ULN, sodium<0.95*LLN or >1.05*ULN, potassium; chloride; calcium; bicarbonate:<0.9*LLN or >1.1*ULN, glucose <0.6*LLN or >1.5*ULN, urine specific gravity <1.003, urine pH <4.5 or >8, urine glucose or ketones (qualitative) >=1, urine protein; urine blood/hemoglobin >=1, urobilinogen; bilirubin; nitrite; leukocyte esterase >=1. (NCT02625207)
Timeframe: Baseline up to Day 11

Interventionparticipants (Number)
Cohort 1- No CYP3A5*1 Alleles1
Cohort 3- Two CYP3A5*1 Alleles3

[back to top]

Part 2: Number of Participants With Clinically Significant Vital Sign Abnormalities

Criteria for clinically significant vital sign abnormalities included supine/sitting pulse rate of <40 bpm or >120 bpm, standing pulse rate of <40 bpm or >140 bpm, supine SBP and standing SBP of <90 mm Hg, >=30 mm Hg, supine DBP and standing DBP of <50 mm Hg, >=20 mm Hg. (NCT02625207)
Timeframe: Baseline up to Day 11

Interventionparticipants (Number)
Cohort 1- No CYP3A5*1 Alleles0
Cohort 3- Two CYP3A5*1 Alleles0

[back to top]

Part 2: Number of Participants With 12-Lead Electrocardiogram (ECG) Abnormalities

Criteria for ECG abnormalities: Maximum PR interval of >=300 msec, maximum QRS interval >=140 msec, maximum QTCF interval (Fridericia's correction) of 450 to <480 msec, 480 to <500 msec and >=500 msec, maximum increase of >=25 percent for baseline values of >200 msec and >=50 percent for baseline values of <=200 msec for PR interval, maximum increase from baseline of >=50 percent for QRS interval, maximum increase from baseline of >=30 msec to <60 msec and maximum increase from baseline of >60 msec in QTCF interval (Fridericia's Correction). (NCT02625207)
Timeframe: Baseline up to Day 11

Interventionparticipants (Number)
Cohort 1- No CYP3A5*1 Alleles0
Cohort 3- Two CYP3A5*1 Alleles0

[back to top]

Part 1: Area Under The Plasma Concentration-Time Curve From Time 0 to 12 Hours (AUC [0-12]) of Maraviroc

AUC (0-12) is the area under the plasma concentration versus time curve from time zero (pre-dose) to 12 hours post-dose. (NCT02625207)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, 9, 12 hours post-dose on Day 5

Interventionnanogram*hour per milliliter (ng*hr/mL) (Geometric Mean)
Cohort 1- No CYP3A5*1 Alleles3441
Cohort 2- One CYP3A5*1 Allele2954
Cohort 3- Two CYP3A5*1 Alleles2181
Cohort 4- No CYP3A5*1 Alleles2947

[back to top]

Part 1: Average Plasma Concentration (Cavg) of Maraviroc

Cavg is the average plasma concentration of maraviroc during the 0 to 12 hour time period. It was calculated as area under the plasma concentration-time curve from 0 to 12 hours (AUC [0-12]) divided by 12. (NCT02625207)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, 9, 12 hours post-dose on Day 5

Interventionnanogram per milliliter (ng/mL) (Geometric Mean)
Cohort 1- No CYP3A5*1 Alleles286.8
Cohort 2- One CYP3A5*1 Allele246.2
Cohort 3- Two CYP3A5*1 Alleles181.6
Cohort 4- No CYP3A5*1 Alleles245.8

[back to top]

Part 1: Maximum Observed Plasma Concentration (Cmax) of Maraviroc

(NCT02625207)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, 9, 12 hours post-dose on Day 5

Interventionng/mL (Geometric Mean)
Cohort 1- No CYP3A5*1 Alleles863.9
Cohort 2- One CYP3A5*1 Allele754.0
Cohort 3- Two CYP3A5*1 Alleles529.0
Cohort 4- No CYP3A5*1 Alleles731.0

[back to top]

Part 1: Number of Participants With 12-Lead Electrocardiogram (ECG) Abnormalities

Criteria for ECG abnormalities: Maximum PR interval of >=300 milliseconds (msec), maximum QRS interval >=140 msec, maximum QTCF interval (Fridericia's correction) of 450 to <480 msec, 480 to <500 msec and >=500 msec, maximum increase of >=25 percent for baseline values of >200 msec and >=50 percent for baseline values of less than or equal to (<=) 200 msec for PR interval, maximum increase from baseline of >=50 percent for QRS interval, maximum increase from baseline of >=30 msec to <60 msec and maximum increase from baseline of >60 msec in QTCF interval (Fridericia's Correction). (NCT02625207)
Timeframe: Baseline up to Day 6

Interventionparticipants (Number)
Cohort 1- No CYP3A5*1 Alleles0
Cohort 2- One CYP3A5*1 Allele0
Cohort 3- Two CYP3A5*1 Alleles0
Cohort 4- No CYP3A5*1 Alleles0

[back to top]

Part 1: Number of Participants With Clinically Significant Vital Sign Abnormalities

Criteria for clinically significant vital sign abnormalities included supine/sitting pulse rate of less than (<) 40 beats per minute (bpm) or greater than (>)120 bpm, standing pulse rate of <40 bpm or >140 bpm, supine systolic blood pressure (SBP) and standing SBP of <90 millimeter of mercury (mm Hg), greater than or equal to (>=) 30 mm Hg, supine diastolic blood pressure (DBP) and standing DBP of <50 mm Hg, >=20 mm Hg. (NCT02625207)
Timeframe: Baseline up to Day 6

Interventionparticipants (Number)
Cohort 1- No CYP3A5*1 Alleles0
Cohort 2- One CYP3A5*1 Allele0
Cohort 3- Two CYP3A5*1 Alleles0
Cohort 4- No CYP3A5*1 Alleles0

[back to top]

Part 2: Area Under The Plasma Concentration-Time Curve From Time 0 to 24 Hours (AUC [0-24]) of Maraviroc

AUC (0-24) is the area under the plasma concentration versus time curve from time zero (pre-dose) to 24 hours post-dose. (NCT02625207)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, 9, 12, 24 hours post-dose on Day 10

Interventionng*hr/mL (Geometric Mean)
Cohort 1- No CYP3A5*1 Alleles4413
Cohort 3- Two CYP3A5*1 Alleles3645

[back to top]

Part 2: Maximum Observed Plasma Concentration (Cmax) of Maraviroc

(NCT02625207)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, 9, 12, 24 hours post-dose on Day 10

Interventionng/mL (Geometric Mean)
Cohort 1- No CYP3A5*1 Alleles633.6
Cohort 3- Two CYP3A5*1 Alleles432.9

[back to top]

Part 2: Average Plasma Concentration (Cavg) of Maraviroc

Cavg is the average plasma concentration of maraviroc during the 0 to 24 hour time period. It was calculated as area under the plasma concentration-time curve from 0 to 24 hours (AUC [0-24]) divided by 24. (NCT02625207)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, 9, 12, 24 hours post-dose on Day 10

Interventionng/mL (Geometric Mean)
Cohort 1- No CYP3A5*1 Alleles184.1
Cohort 3- Two CYP3A5*1 Alleles151.7

[back to top]

Part 1: Number of Participants With Laboratory Abnormalities

Criteria: Hemoglobin; hematocrit; red blood cell count: <0.8*lower limit of normal, (LLN), mean corpuscular volume; mean corpuscular hemoglobin concentration; mean platelet volume:<0.9*LLN or >1.1* upper limit of normal (ULN), platelet: <0.5*LLN or >1.75*ULN, white blood cells <0.6*LLN or >1.5*ULN, lymphocyte; neutrophil: <0.8*LLN or >1.2*ULN, basophil; eosinophil; monocyte:>1.2*ULN, bilirubin (total, direct, indirect) >1.5*ULN, aspartate aminotransferase; alanine aminotransferase; alkaline phosphatase:>3.0*ULN, total protein; albumin:<0.8*LLN or >1.2*ULN; creatinine: >1.3*ULN, uric acid>1.2*ULN, sodium<0.95*LLN or >1.05*ULN, potassium; chloride; calcium; bicarbonate:<0.9*LLN or >1.1*ULN, glucose <0.6*LLN or >1.5*ULN, urine specific gravity <1.003, urine pH <4.5 or >8, urine glucose or ketones (qualitative) >=1, urine protein; urine blood/hemoglobin >=1, urobilinogen; bilirubin; nitrite; leukocyte esterase >=1. (NCT02625207)
Timeframe: Baseline up to Day 6

Interventionparticipants (Number)
Cohort 1- No CYP3A5*1 Alleles1
Cohort 2- One CYP3A5*1 Allele5
Cohort 3- Two CYP3A5*1 Alleles0
Cohort 4- No CYP3A5*1 Alleles2

[back to top]

Part 1: Plasma Concentration of Maraviroc at 12 Hours Post-dose

(NCT02625207)
Timeframe: 12 hours post-dose on Day 5

Interventionng/mL (Geometric Mean)
Cohort 1- No CYP3A5*1 Alleles59.84
Cohort 2- One CYP3A5*1 Allele63.09
Cohort 3- Two CYP3A5*1 Alleles45.32
Cohort 4- No CYP3A5*1 Alleles63.10

[back to top]

Part 2: Number of Participants With Treatment Emergent Adverse Events (AEs) and Serious Adverse Events (SAEs)

An AE was any untoward medical occurrence in a participant who received study drug without regard to possibility of causal relationship. SAE was 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. Treatment-emergent are events between first dose of study drug and up to 11 days that were absent before treatment or that worsened relative to pretreatment state. (NCT02625207)
Timeframe: Baseline up to end of study (up to 11 days)

,
Interventionparticipants (Number)
AEsSAEs
Cohort 1- No CYP3A5*1 Alleles20
Cohort 3- Two CYP3A5*1 Alleles10

[back to top]

Number of Participants With Adverse Events (AEs)

An AE is any untoward medical occurrence in a participant participating in a clinical study that does not necessarily have a causal relationship with the pharmaceutical/biological agent under study. (NCT03123848)
Timeframe: Up to approximately 1 month

InterventionParticipants (Number)
Darunavir 800 mg/Cobicistat 150 mg as FDC (Prezcobix)0

[back to top]

Apparent Total Body Clearance of Drug at the Terminal Phase After Extravascular Administration (CL/F)

CL/F is the apparent total body clearance of drug at the terminal phase after extravascular administration. (NCT03123848)
Timeframe: Predose, 0.5, 1, 1.5, 2, 2.5, 3, 4, 5, 6, 9, 12, 16, 20, 24, 36, 48, 60, 72 hours postdose

Interventionmilliliter per hour (mL/h) (Mean)
Darunavir 800 mg (FDC: Darunavir 800/Cobicistat 150 mg)16974
Cobicistat 150 mg (FDC: Darunavir 800/Cobicistat 150 mg)33573

[back to top]

Time to Reach the Maximum Plasma Concentration (Tmax)

Tmax is defined as the time to reach the maximum plasma concentration. (NCT03123848)
Timeframe: Predose, 0.5, 1, 1.5, 2, 2.5, 3, 4, 5, 6, 9, 12, 16, 20, 24, 36, 48, 60, 72 hours postdose

Interventionhour (h) (Median)
Darunavir 800 mg (FDC: Darunavir 800/Cobicistat 150 mg)4.00
Cobicistat 150 mg (FDC: Darunavir 800/Cobicistat 150 mg)4.00

[back to top]

Terminal Elimination Half-Life (t1/2)

t1/2 is the time measured for the plasma concentration to decrease by 1 half to its original concentration. It is associated with the terminal slope of the semi logarithmic drug concentration-time curve, and is calculated as 0.693/lambda(z). (NCT03123848)
Timeframe: Predose, 0.5, 1, 1.5, 2, 2.5, 3, 4, 5, 6, 9, 12, 16, 20, 24, 36, 48, 60, 72 hours postdose

InterventionHour (Mean)
Darunavir 800 mg (FDC: Darunavir 800/Cobicistat 150 mg)4.4
Cobicistat 150 mg (FDC: Darunavir 800/Cobicistat 150 mg)3.5

[back to top]

Maximum Observed Plasma Concentration (Cmax)

The Cmax is the maximum observed plasma concentration. (NCT03123848)
Timeframe: Predose, 0.5, 1, 1.5, 2, 2.5, 3, 4, 5, 6, 9, 12, 16, 20, 24, 36, 48, 60, 72 hours postdose

Interventionnanogram per milliliter (ng/mL) (Mean)
Darunavir 800 mg (FDC: Darunavir 800/Cobicistat 150 mg)5496
Cobicistat 150 mg (FDC: Darunavir 800/Cobicistat 150 mg)832

[back to top]

Elimination Rate Constant (Lambda[z])

Lambda(z) is first-order rate constant associated with the terminal portion of the curve, determined as the negative slope of the terminal log-linear phase of the drug concentration-time curve. (NCT03123848)
Timeframe: Predose, 0.5, 1, 1.5, 2, 2.5, 3, 4, 5, 6, 9, 12, 16, 20, 24, 36, 48, 60, 72 hours postdose

Interventionper hour (1/h) (Mean)
Darunavir 800 mg (FDC: Darunavir 800/Cobicistat 150 mg)0.173
Cobicistat 150 mg (FDC: Darunavir 800/Cobicistat 150 mg)0.199

[back to top]

Concentration at Last Quantifiable Time Point (Clast)

Clast is defined as concentration at last quantifiable time point. (NCT03123848)
Timeframe: Predose, 0.5, 1, 1.5, 2, 2.5, 3, 4, 5, 6, 9, 12, 16, 20, 24, 36, 48, 60, 72 hours postdose

Interventionng/mL (Mean)
Darunavir 800 mg (FDC: Darunavir 800/Cobicistat 150 mg)33.3
Cobicistat 150 mg (FDC: Darunavir 800/Cobicistat 150 mg)8.59

[back to top]

Area Under the Plasma Concentration-Time Curve From Time Zero to Time the Last Quantifiable Time (AUC[0-last])

AUC(0-last) is defined as area under the plasma concentration-time curve from time zero to time the last quantifiable time, calculated by linear trapezoidal summation. (NCT03123848)
Timeframe: Predose, 0.5, 1, 1.5, 2, 2.5, 3, 4, 5, 6, 9, 12, 16, 20, 24, 36, 48, 60, 72 hours postdose

Interventionhour*nanogram per milliliter (h*ng/mL) (Mean)
Darunavir 800 mg (FDC: Darunavir 800/Cobicistat 150 mg)51274
Cobicistat 150 mg (FDC: Darunavir 800/Cobicistat 150 mg)5667

[back to top]

Area Under the Plasma Concentration-Time Curve From Time Zero to Infinite Time (AUC0-infinity)

AUC(0-infinity) is defined as area under the plasma concentration-time curve from time zero to infinite time. (NCT03123848)
Timeframe: Predose, 0.5, 1, 1.5, 2, 2.5, 3, 4, 5, 6, 9, 12, 16, 20, 24, 36, 48, 60, 72 hours postdose

Interventionh*ng/mL (Mean)
Darunavir 800 mg (FDC: Darunavir 800/Cobicistat 150 mg)51460
Cobicistat 150 mg (FDC: Darunavir 800/Cobicistat 150 mg)5710

[back to top]

Apparent Volume of Distribution (Vz/F)

Vz/F is defined as the apparent volume of distribution at the terminal phase after extravascular administration. (NCT03123848)
Timeframe: Predose, 0.5, 1, 1.5, 2, 2.5, 3, 4, 5, 6, 9, 12, 16, 20, 24, 36, 48, 60, 72 hours postdose

Interventionmilliliter (mL) (Mean)
Darunavir 800 mg (FDC: Darunavir 800/Cobicistat 150 mg)111970
Cobicistat 150 mg (FDC: Darunavir 800/Cobicistat 150 mg)166460

[back to top]

The Change in Serum Creatinine From Baseline to 48 Weeks.

A second primary endpoint was evaluating the change in serum creatinine from baseline to 48 weeks for all subjects. (NCT03198884)
Timeframe: 48 weeks

Interventionmg/dL (Mean)
Retrospective Chart Review73.4

[back to top]

Number of Participants With RNA <50 Copies/mL at 48 Weeks

Our first primary endpoint evaluated the percent of study subjects with an RNA <50 copies/mL at 48 weeks after initiation of the once daily two-drug regimen. (NCT03198884)
Timeframe: 48 weeks

InterventionParticipants (Count of Participants)
Retrospective Chart Review19

[back to top]

Number of Participants With RNA <50 Copies/mL at 24, 36, and 48 Weeks

"This secondary outcome measure analyzed the percentage of subjects with < 50 copies/mL RNA at time points 24, 36 and 48 weeks.~The percent of subjects with an RNA < 50 copies/mL at each time point was analyzed using McNemar's test following the guidelines of the Snapshot algorithm. Missing RNA data was considered a treatment failure." (NCT03198884)
Timeframe: 48 weeks

InterventionParticipants (Count of Participants)
Analysis of HIV RNA at Week 2419
Analysis of HIV RNA at Week 3614
Analysis of HIV RNA at Week 4819

[back to top]

Change in Mean CD4+ Cell Count From Baseline.

A secondary endpoint included changes from baseline in CD4+ cell counts. (NCT03198884)
Timeframe: 48 weeks

Interventioncells/μL (Mean)
Week 24454
Week 36428
Week 48456

[back to top]

Analysis of Creatinine Clearance at Time Points 24, 36 and 48 Weeks.

(NCT03198884)
Timeframe: 48 weeks

Interventionmg/dL (Mean)
Analysis of Serum Creatinine at Week 2475.8
Analysis of Serum Creatinine at Week 3669.1
Analyis of Serum Creatinine at Week 4877.7

[back to top]

Incidence of Adverse Events.

10 study subjects reported an adverse event. (NCT03198884)
Timeframe: 48 weeks

InterventionParticipants (Count of Participants)
InsomniaDiarrheaHeadache
Retrospective Chart Review643

[back to top]

Number of Grade 1 Adverse Events Reported

"10 study subjects reported adverse events. All adverse events reported (insomnia, diarrhea, headache) were of Grade 1 severity.~There were no adverse events that led to discontinuation of the study regimen." (NCT03198884)
Timeframe: 48 weeks

InterventionAdverse Events (Number)
InsomniaDiarrheaHeadache
Adverse Events643

[back to top]

Terminal Elimination Half-life (t½) for Rivaroxaban

Terminal elimination half-life (t½) for Rivaroxaban following a single dose of rivaroxaban 10 mg orally on days 1, 7, and 13 was analyzed using noncompartmental methods with Phoenix WinNonlin® (version 8.0; Pharsight Corporation, Mountain View, CA). The apparent elimination rate constant (λZ) was determined by calculating the absolute value of the slope of the log-linear regression of at least 3 points of the plasma concentration-time plot. The t½ was calculated as 0.693/apparent elimination rate constant (λZ). (NCT03864406)
Timeframe: Up to 24 hours postdose on days 1, 7, and 13

InterventionHours (Mean)
Day 1 (phase 1)Day 7 (phase 2)Day 13 (phase 3)
Pharmacokinetic Study in Healthy Volunteers5.658.197.49

[back to top]

Apparent Oral Clearance (CL/F) for Rivaroxaban

Apparent oral clearance (CL/F) for Rivaroxaban following a single dose of rivaroxaban 10 mg orally on days 1, 7, and 13 was analyzed using noncompartmental methods with Phoenix WinNonlin® (version 8.0; Pharsight Corporation, Mountain View, CA). CL/F was calculated as 10 mg ÷ AUC0-tau for rivaroxaban. (NCT03864406)
Timeframe: Up to 24 hours postdose on days 1, 7, and 13

InterventionL/hr (Mean)
Day 1 (phase 1)Day 7 (phase 2)Day 13 (phase 3)
Pharmacokinetic Study in Healthy Volunteers11.044.925.19

[back to top]

Apparent Volume of Distribution (V/F) for Rivaroxaban

Apparent volume of distribution (V/F) for Rivaroxaban following a single dose of rivaroxaban 10 mg orally on days 1, 7, and 13 was analyzed using noncompartmental methods with Phoenix WinNonlin® (version 8.0; Pharsight Corporation, Mountain View, CA). (NCT03864406)
Timeframe: Up to 24 hours postdose on days 1, 7, and 13

InterventionLiter (Mean)
Day 1 (phase 1)Day 7 (phase 2)Day 13 (phase 3)
Pharmacokinetic Study in Healthy Volunteers89.9456.2252.84

[back to top]

Time to Maximum Plasma Concentration (Tmax) for Rivaroxaban

Time to maximum plasma concentration for Rivaroxaban following a single dose of rivaroxaban 10 mg orally on days 1, 7, and 13 was analyzed using noncompartmental methods with Phoenix WinNonlin® (version 8.0; Pharsight Corporation, Mountain View, CA). Tmax for rivaroxaban was obtained directly by visual inspection of the plasma concentration versus time over 24 hours postdose. (NCT03864406)
Timeframe: Up to 24 hours postdose on days 1, 7, and 13

InterventionHours (Mean)
Day 1 (phase 1)Day 7 (phase 2)Day 13 (phase 3)
Pharmacokinetic Study in Healthy Volunteers2.54.13.02

[back to top]

Area Under the Concentration Versus Time Curve (AUC0-24hr) for Rivaroxaban

Area under the concentration versus time curve from (AUC0-24hr) following a single dose of rivaroxaban 10 mg orally on days 1, 7, and 13 to measure total drug exposure was analyzed by the linear up/log down trapezoidal rule using noncompartmental methods with Phoenix WinNonlin® (version 8.0; Pharsight Corporation, Mountain View, CA). (NCT03864406)
Timeframe: 0 to 24 hours postdose on days 1, 7, and 13

Interventionhr*ng/ml (Mean)
Day 1 (phase 1)Day 7 (phase 2)Day 13 (phase 3)
Pharmacokinetic Study in Healthy Volunteers991.251929.781939

[back to top]

Area Under the Concentration Versus Time Curve (AUC0-∞) for Rivaroxaban

Area under the concentration versus time curve from 0 to infinity (AUC0-∞) following a single dose of rivaroxaban 10 mg orally on days 1, 7, and 13 to measure the total drug exposure was analyzed by the linear up/log down trapezoidal rule using noncompartmental methods with Phoenix WinNonlin® (version 8.0; Pharsight Corporation, Mountain View, CA). (NCT03864406)
Timeframe: Total drug exposure at time point zero to infinity on days 1, 7, & 13

Interventionhr*ng/ml (Mean)
Day 1 (phase 1)Day 7 (phase 2)Day 13 (phase 3)
Pharmacokinetic Study in Healthy Volunteers1049.192274.32226.38

[back to top]

Maximum Total Plasma Concentration (Cmax) for Rivaroxaban

Maximum total plasma concentration (Cmax) following a single dose of rivaroxaban 10 mg orally on days 1, 7, and 13 was analyzed using noncompartmental methods with Phoenix WinNonlin® (version 8.0; Pharsight Corporation, Mountain View, CA). Cmax for rivaroxaban was obtained directly by visual inspection of the plasma concentration versus time over 24 hours postdose. (NCT03864406)
Timeframe: Up to 24 hours postdose on days 1, 7, and 13

Interventionng/mL (Mean)
Day 1 (phase 1)Day 7 (phase 2)Day 13 (phase 3)
Pharmacokinetic Study in Healthy Volunteers128.6192.25196.17

[back to top]

Minimum Total Plasma Concentration (Cmin) for Rivaroxaban

Minimum total plasma concentration (Cmin) for Rivaroxaban following a single dose of rivaroxaban 10 mg orally on days 1, 7, and 13 was analyzed using noncompartmental methods with Phoenix WinNonlin® (version 8.0; Pharsight Corporation, Mountain View, CA). Cmin for rivaroxaban was obtained directly by visual inspection of the plasma concentration versus times over 24 hours postdose. (NCT03864406)
Timeframe: Up to 24 hours postdose on days 1, 7, and 13

Interventionng/mL (Mean)
Day 1 (phase 1)Day 7 (phase 2)Day 13 (phase 3)
Pharmacokinetic Study in Healthy Volunteers6.9327.3224.43

[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: 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 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 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]

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 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 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 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 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 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: 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: 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: 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 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 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 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 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 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 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 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 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 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 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 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 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 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 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 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 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: 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 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 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 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 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 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]