Page last updated: 2024-11-13

cobicistat

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

Cross-References

ID SourceID
PubMed CID25151504
CHEMBL ID2095208
CHEBI ID72291
SCHEMBL ID2736227
MeSH IDM0558562

Synonyms (76)

Synonym
HY-10493
tybost (tn)
D09881
cobicistat (jan/usan/inn)
cobicistatum
1,3-thiazol-5-ylmethyl [(2r,5r)-5-{[(2s)-2-({[(2-isopropyl-1,3-thiazol-4-yl)methyl](methyl)carbamoyl}amino)-4-(morpholin-4-yl)butanoyl]amino}-1,6-diphenylhexan-2-yl]carbamate
cobicistat (gs-9350)
CHEMBL2095208
CHEBI:72291 ,
cobicistat on silicon dioxide
cobicistat, (r,r,s)-
gs-9350 ,
cobicistat [usan:inn]
1004316-88-4
gs 9350
thiazol-5-ylmethyl ((1r,4r)-1-benzyl-4-({(2s)-2-((methyl{(2-(1-methylethyl)thiazol-4-yl)methyl}carbamoyl)amino)-4-(morpholin-4-yl)butanoyl}amino)-5-phenylpentyl)carbamate
2,7,10,12-tetraazatridecanoic acid, 12-methyl-13-(2-(1-methylethyl)-4-thiazolyl)-9-(2-(4-morpholinyl)ethyl)-8,11-dioxo-3,6-bis(phenylmethyl)-, 5-thiazolylmethyl ester, (3r,6r,9s)-
lw2e03m5pg ,
cobicistat ,
unii-lw2e03m5pg
tybost
cobicistat component of prezcobix
1,3-thiazol-5-ylmethyl ((2r,5r)-5-(((2s)-2-((methyl((2-(propan-2-yl)-1,3-thiazol-4-yl)methyl)carbamoyl)amino)-4-(morpholin-4-yl)butanoyl)amino)-1,6-diphenylhexan-2-yl)carbamate
evotaz component cobicistat
(1,3-thiazol-5-yl)methyl (5s,8r,11r)-8,11-dibenzyl-2-methyl-5-(2-(morpholin-4-yl)ethyl)-1-(2-(propan-2-yl)-1,3-thiazol-4-yl)-3,6-dioxo-2,4,7,12-tetraazatridecan-13-oate
cobicistat [vandf]
cobicistat [orange book]
cobicistat [usan]
cobicistat component of stribild
symtuza component cobicistat
cobicistat [mi]
rezolsta component cobicistat
cobicistat component of symtuza
cobicistat component of genvoya
cobicistat [inn]
prezcobix component cobicistat
cobicistat component of evotaz
stribild component cobicistat
cobicistat [jan]
genvoya component cobicistat
cobicistat [who-dd]
CS-0742
bdbm50447471
S2900
thiazol-5-ylmethyl n-[(1r,4r)-1-benzyl-4-[[(2s)-2-[[(2-isopropylthiazol-4-yl)methyl-methyl-carbamoyl]amino]-4-morpholino-butanoyl]amino]-5-phenyl-pentyl]carbamate
gtpl7535
1,3-thiazol-5-ylmethyl n-[(2r,5r)-5-[[(2s)-2-[[methyl-[(2-propan-2-yl-1,3-thiazol-4-yl)methyl]carbamoyl]amino]-4-morpholin-4-ylbutanoyl]amino]-1,6-diphenylhexan-2-yl]carbamate
SCHEMBL2736227
thiazol-5-ylmethyl ((2r,5r)-5-((s)-2-(3-((2-isopropylthiazol-4-yl)methyl)-3-methylureido)-4-morpholinobutanamido)-1,6-diphenylhexan-2-yl)carbamate
AC-28961
thiazol-5-ylmethyl (2r,5r)-5-((s)-2-(3-((2-isopropylthiazol-4-yl)methyl)-3-methylureido)-4-morpholinobutanamido)-1,6-diphenylhexan-2-ylcarbamate
AB01566899_01
DTXSID00143269 ,
AKOS025404908
DB09065
mfcd18251449
cobicistat,gs-9350
ZCIGNRJZKPOIKD-CQXVEOKZSA-N
1,3-thiazol-5-ylmethyl n-[(2r,5r)-5-[(2s)-2-{[methyl({[2-(propan-2-yl)-1,3-thiazol-4-yl]methyl})carbamoyl]amino}-4-(morpholin-4-yl)butanamido]-1,6-diphenylhexan-2-yl]carbamate
SW219553-1
BCP06630
EX-A2578
cobicistat; gs-9350
AS-17061
Q5138908
CCG-270459
NCGC00386235-04
2,7,10,12-tetraazatridecanoic acid, 12-methyl-13-[2-(1-methylethyl)-4-thiazolyl]-9-[2-(4-morpholinyl)ethyl]-8,11-dioxo-3,6-bis(phenylmethyl)-, 5-thiazolylmethyl ester, (3r,6r,9s)-
NCGC00386235-06
EN300-7404993
(1,3-thiazol-5-yl)methyl n-[(2r,5r)-5-[(2s)-2-{[methyl({[2-(propan-2-yl)-1,3-thiazol-4-yl]methyl})carbamoyl]amino}-4-(morpholin-4-yl)butanamido]-1,6-diphenylhexan-2-yl]carbamate
1,3-thiazol-5-ylmethyl ((2r,5r)-5-(((2s)-2-((((2-isopropyl-1,3-thiazol-4-yl)methyl)(methyl)carbamoyl)amino)-4-(morpholin-4-yl)butanoyl)amino)-1,6-diphenylhexan-2-yl)carbamate
v03ax03
1,3-thiazol-5-ylmethyl ((2r,5r)-5-((2s)-2-(3-methyl-3-((2-(1-methylethyl)-1,3-thiazol-4-yl)methyl)ureido)-4-(morpholin-4-yl)butanamido)-1,6-diphenylhexan-2-yl)carbamate
dtxcid1065760
thiazol-5-ylmethyl ((1r,4r)-1-benzyl-4-(((2s)-2-((methyl((2-(1-methylethyl)thiazol-4-yl)methyl)carbamoyl)amino)-4-(morpholin-4-yl)butanoyl)amino)-5-phenylpentyl)carbamate

Research Excerpts

Overview

Cobicistat is a new pharmacokinetic booster that is a selective inhibitor of cytochrome P450 3A, the main metabolizing pathway of several antiretrovirals. Cobicistat (COBI) is an effective pharmacoenhancer of darunavir when administered as an FDC.

ExcerptReferenceRelevance
"Cobicistat is a cytochrome P450 3A inhibitor and in this case inhibited clearance of intrabursal triamcinolone, leading to exogenous glucocorticoid excess and adrenal suppression."( Exogenous steroid-induced hypoadrenalism in a person living with HIV caused by a drug-drug interaction between cobicistat and intrabursal triamcinolone.
Macpherson, G; Makaram, N; Roberts, SB; Russell, CD; Stevens, J, 2018
)
1.41
"Cobicistat is a new pharmacokinetic booster that is a selective inhibitor of cytochrome P450 3A, the main metabolizing pathway of several antiretrovirals."( Cobicistat: a new boost for the treatment of human immunodeficiency virus infection.
Priano, J; Schafer, JJ; Shah, BM; Squires, KE, 2013
)
2.55
" Cobicistat (COBI) is a pharmacoenhancer with no antiretroviral activity in vitro."( Cobicistat versus ritonavir as a pharmacoenhancer of atazanavir plus emtricitabine/tenofovir disoproxil fumarate in treatment-naive HIV type 1-infected patients: week 48 results.
Andrade-Villanueva, J; Antunes, F; Arastéh, K; Callebaut, C; Cheng, AK; Chetchotisakd, P; DeJesus, E; Fehr, J; Gallant, JE; Koenig, E; Liu, Y; Moyle, G; Rhee, MS; Rizzardini, G; Szwarcberg, J; Zhong, L, 2013
)
2.74
"Cobicistat is a novel cytochrome P450 3A4 (CYP3A4) inhibitor in advanced clinical evaluation for use as a pharmacoenhancer of antiretroviral drugs. "( Cobicistat, a pharmacoenhancer for HIV treatments.
Temesgen, Z, 2013
)
3.28
"Cobicistat (Tybost™) is a mechanism-based inhibitor of cytochrome P450 (CYP) 3A enzymes that is indicated in the EU as a pharmacokinetic enhancer (i.e. "( Cobicistat: a review of its use as a pharmacokinetic enhancer of atazanavir and darunavir in patients with HIV-1 infection.
Deeks, ED, 2014
)
3.29
"Cobicistat is an effective pharmacoenhancer of darunavir when administered as an FDC."( 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
)
1.37
"Cobicistat (COBI) is an alternative pharmacoenhancer to ritonavir. "( A randomized trial in healthy subjects to assess the bioequivalence of an atazanavir/cobicistat fixed-dose combination tablet versus administration as separate agents.
Bertz, R; Jariwala, N; Ravindran, P; Sevinsky, H; Sims, K; Tao, X; Wang, R; Xu, X, 2015
)
2.08
"Cobicistat (COBI) is a pharmacoenhancer that optimizes systemic exposures of protease inhibitors (PIs) such as atazanavir (ATV) and darunavir (DRV)."( Cobicistat-boosted protease inhibitors in HIV-infected patients with mild to moderate renal impairment.
Abram, ME; Curley, J; Custodio, J; Fisher, M; Graham, H; Laplante, F; Liu, Y; Martorell, C; McDonald, CK; Post, FA; Ramgopal, M; Rhee, MS; Szwarcberg, J,
)
3.02
"Cobicistat is a boosting agent for the integrase inhibitor elvitegravir and the protease inhibitors atazanavir and darunavir."( Pharmacokinetic enhancement in HIV antiretroviral therapy: a comparison of ritonavir and cobicistat.
Bow, D; Ng, J; Norton, M; Renjifo, B; Salem, AH; van Wyk, J,
)
1.07
"Cobicistat is an alternative agent to ritonavir for boosting plasma drug levels of darunavir among other antiretrovirals."( Darunavir/cobicistat once daily for the treatment of HIV.
Crauwels, H; de Smedt, G; Iterbeke, K; Kakuda, TN; Opsomer, M; Tomaka, F; van de Casteele, T; Vanveggel, S, 2015
)
1.54
"Cobicistat (COBI) is a pharmacoenhancer with no antiretroviral activity."( Brief Report: Cobicistat Compared With Ritonavir as a Pharmacoenhancer for Atazanavir in Combination With Emtricitabine/Tenofovir Disoproxil Fumarate: Week 144 Results.
Abram, ME; Andrade-Villanueva, JF; Antunes, F; Arastéh, K; Cao, H; Chetchotisakd, P; DeJesus, E; Fehr, J; Gallant, JE; Koenig, E; Liu, HC; Rizzardini, G; Szwarcberg, J, 2015
)
2.22
"Cobicistat is a PK enhancer lacking antiviral activity that, via selective cytochrome P-450 (CYP) 3A inhibition, inhibits the metabolism of certain antiretroviral medications and is used for prolonging their effect. "( Cobicistat: Review of a Pharmacokinetic Enhancer for HIV Infection.
Gauthier, TP; Schafer, JJ; Sherman, EM; Unger, NR; Worley, MV, 2015
)
3.3
"Cobicistat is an alternative agent to ritonavir for boosting plasma drug levels for several antiretrovirals."( Darunavir/cobicistat for the treatment of HIV-1: a new era for compact drugs with high genetic barrier to resistance.
Capetti, A; Cossu, MV; Rizzardini, G, 2015
)
1.54
"Cobicistat (COBI) is a pharmacoenhancer for antiretroviral therapy."( Biotransformation of Cobicistat: Metabolic Pathways and Enzymes.
Liu, K; Lu, J; Ma, X; Shehu, AI; Wang, P, 2016
)
2.2

Effects

Cobicistat has been studied as a booster for elvitegravir, a second-generation integrase inhibitor, and protease inhibitors. Cobicistat was recently approved in the USA as a fixed-dose combination.

ExcerptReferenceRelevance
"Cobicistat has a lower potential for off-target drug interactions than the standard boosting agent ritonavir, due to its more selective inhibition of CYP3A and lower likelihood for enzymatic induction, and is devoid of anti-HIV activity."( Cobicistat: a review of its use as a pharmacokinetic enhancer of atazanavir and darunavir in patients with HIV-1 infection.
Deeks, ED, 2014
)
2.57
"Cobicistat has been studied as a booster for elvitegravir, a second-generation integrase inhibitor, and protease inhibitors."( Cobicistat: a new boost for the treatment of human immunodeficiency virus infection.
Priano, J; Schafer, JJ; Shah, BM; Squires, KE, 2013
)
2.55
"Cobicistat has been evaluated in clinical trials and was recently approved in the USA as a fixed-dose combination with the integrase inhibitor, elvitegravir and two nucleos(t)ide analogs."( Pharmacokinetic enhancers in HIV therapeutics.
Acosta, EP; Delille, C; Larson, KB; Otofokun, I; Wang, K, 2014
)
1.12
"Cobicistat has been studied as a booster of elvitegravir, a second-generation integrase inhibitor, and of the protease inhibitors atazanavir and darunavir."( Cobicistat: Review of a Pharmacokinetic Enhancer for HIV Infection.
Gauthier, TP; Schafer, JJ; Sherman, EM; Unger, NR; Worley, MV, 2015
)
2.58

Actions

Cobicistat has a lower potential for off-target drug interactions than the standard boosting agent ritonavir. Cobicistat may increase serum creatinine levels via the inhibition of proximal renal tubular cell transporters and thus reduce estimated glomerular filtration rate.

ExcerptReferenceRelevance
"Cobicistat has a lower potential for off-target drug interactions than the standard boosting agent ritonavir, due to its more selective inhibition of CYP3A and lower likelihood for enzymatic induction, and is devoid of anti-HIV activity."( Cobicistat: a review of its use as a pharmacokinetic enhancer of atazanavir and darunavir in patients with HIV-1 infection.
Deeks, ED, 2014
)
2.57
"Cobicistat may increase serum creatinine levels via the inhibition of proximal renal tubular cell transporters and thus reduce estimated glomerular filtration rate, although it does not appear to affect actual glomerular filtration rate."( Cobicistat: Review of a Pharmacokinetic Enhancer for HIV Infection.
Gauthier, TP; Schafer, JJ; Sherman, EM; Unger, NR; Worley, MV, 2015
)
2.58

Toxicity

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 and EMERALD studies of darunavir/cobicistat. The rate of discontinuations because of NPAEs was low, but was significantly higher for dolutegravir.

ExcerptReferenceRelevance
" There was no statistically significant differences in the risk difference for serious adverse events (5."( An Indirect Comparison of Efficacy and Safety of Elvitegravir/Cobicistat/Emtricitabine/Tenofovir Disoproxil Fumarate and Abacavir/Lamivudine + Dolutegravir in Initial Therapy.
Behrens, G; Borg, P; Bouee, S; M Llibre, J; Moyle, G; Piontkowsky, D; Raffi, F; Reilly, G; Rogatto, F, 2016
)
0.67
" The regimen was well tolerated and no discontinuations related to adverse events occurred."( Safety, efficacy, and pharmacokinetics of a single-tablet regimen containing elvitegravir, cobicistat, emtricitabine, and tenofovir alafenamide in treatment-naive, HIV-infected adolescents: a single-arm, open-label trial.
Batra, J; Chakraborty, R; Gaur, AH; Kizito, H; Kosalaraksa, P; Luesomboon, W; Myers, M; Porter, D; Prasitsueubsai, W; Quirk, E; Rakhmanina, N; Rassool, M; Rhee, MS; SenGupta, D; Shao, Y; Ting, L, 2016
)
0.65
" Phase II and III randomized clinical trials evaluate the efficacy and safety of EVG/c/TAF/FTC and tenofovir disoproxil fumerate (TDF)-containing arms; renal impairment, bone mineral density, metabolic effects, and other adverse events are topics explored within this review."( A Review of the Efficacy and Safety of Genvoya® (Elvitegravir, Cobicistat, Emtricitabine, and Tenofovir Alafenamide) in the Management of HIV-1 Infection.
Angione, SA; Cherian, SM; Özdener, AE, 2018
)
0.72
" The most common adverse events were diarrhea, nausea, and headache."( A Review of the Efficacy and Safety of Genvoya® (Elvitegravir, Cobicistat, Emtricitabine, and Tenofovir Alafenamide) in the Management of HIV-1 Infection.
Angione, SA; Cherian, SM; Özdener, AE, 2018
)
0.72
" Data seem to suggest it may also be effective and safe in patients with mild to moderate renal impairment."( A Review of the Efficacy and Safety of Genvoya® (Elvitegravir, Cobicistat, Emtricitabine, and Tenofovir Alafenamide) in the Management of HIV-1 Infection.
Angione, SA; Cherian, SM; Özdener, AE, 2018
)
0.72
"5% of those treated with EVG discontinued due to adverse events (AE)."( Clinical Experience with the Integrase Inhibitors Dolutegravir and Elvitegravir in HIV-infected Patients: Efficacy, Safety and Tolerance.
Balboa-Barreiro, V; Castro-Iglesias, Á; Cid-Silva, P; Fernández-Bargiela, N; Llibre, JM; Margusino-Framiñán, L; Martín-Herranz, I; Pernas-Souto, B; Poveda, E, 2017
)
0.46
"Patients initiating EVG/COBI/FTC/TDF were enrolled in the SCOLTA project, a multicenter observational study reporting grade 3-4 Adverse Events in subjects beginning new antiretroviral drug regimens."( Safety and tolerability of Elvitegravir/Cobicistat/Emtricitabine/Tenofovir Disoproxil fumarate in a real life setting: Data from surveillance cohort long-term toxicity antiretrovirals/antivirals (SCOLTA) project.
Bandera, A; Bonfanti, P; Calza, L; Carenzi, L; Celesia, BM; Cordier, L; De Socio, GV; Di Biagio, A; Falasca, K; Gori, A; Madeddu, G; Maggi, P; Martinelli, C; Orofino, G; Pellicanò, GF; Quirino, T; Ricci, E; Rusconi, S; Squillace, N; Vichi, F, 2017
)
0.72
" 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
" The elvitegravir group showed more discontinuations because of renal adverse events (2."( Efficacy and safety of switching to dolutegravir plus emtricitabine/tenofovir disoproxil fumarate (TDF) or elvitegravir/cobicistat/emtricitabine/TDF in virologically suppressed HIV-infected patients in clinical practice: results from a multicentre, observ
Bagella, P; Baldin, G; Capetti, A; Ciccullo, A; Cossu, MV; De Luca, A; Di Giambenedetto, S; Giacomelli, A; Lagi, F; Latini, A; Madeddu, G; Rusconi, S; Sterrantino, G, 2019
)
0.72
"The aim of the study was to assess the rates of discontinuation of integrase inhibitor regimens because of any neuropsychiatric adverse event (NPAE) and the factors associated with discontinuation."( Discontinuation of dolutegravir, elvitegravir/cobicistat and raltegravir because of toxicity in a prospective cohort.
Abdulghani, N; Ambrosioni, J; Casabona, J; Curran, A; Domingo, P; Force, L; Homar, F; Llibre, JM; Miró, JM; Montoliu, A; Peraire, J; Riera, M; Tiraboschi, J, 2019
)
0.77
" Every discontinuation because of adverse events (AEs) was double-checked directly with treating physicians."( Discontinuation of dolutegravir, elvitegravir/cobicistat and raltegravir because of toxicity in a prospective cohort.
Abdulghani, N; Ambrosioni, J; Casabona, J; Curran, A; Domingo, P; Force, L; Homar, F; Llibre, JM; Miró, JM; Montoliu, A; Peraire, J; Riera, M; Tiraboschi, J, 2019
)
0.77
" The rate of discontinuations because of NPAEs was low, but was significantly higher for dolutegravir than for elvitegravir/cobicistat, with significant differences among centres, suggesting that greater predisposition to believe that a given adverse event is caused by a given drug of some treating physicians might play a role in the discordance seen between cohorts."( Discontinuation of dolutegravir, elvitegravir/cobicistat and raltegravir because of toxicity in a prospective cohort.
Abdulghani, N; Ambrosioni, J; Casabona, J; Curran, A; Domingo, P; Force, L; Homar, F; Llibre, JM; Miró, JM; Montoliu, A; Peraire, J; Riera, M; Tiraboschi, J, 2019
)
0.98
" 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
"6%) patients because of adverse effects."( Efficacy and Safety of Elvitegravir/Cobicistat/Emtricitabine/Tenofovir Alafenamide as Maintenance Treatment in HIV/HBV-Coinfected Patients.
Cheng, CY; Cheng, SH; Ho, MW; Huang, SH; Huang, YS; Hung, CC; Lee, CH; Lee, YT; Lin, SP; Liou, BH; Liu, CE; Lu, PL; Sun, HY; Tang, HJ; Tsai, HC; Yang, CJ, 2021
)
0.9
" The current work investigated the mechanisms of adverse drug-drug interactions associated with COBI that lead to liver damage."( Targeting Xenobiotic Nuclear Receptors PXR and CAR to Prevent Cobicistat Hepatotoxicity.
Gonzalez, FJ; Li, J; Lu, J; Ma, X; McMahon, D; Shehu, AI; Xie, W; Zhu, J, 2021
)
0.86
" Adverse events were observed in 15% of the patients, and the regimen was discontinued in only six patients."( Effectiveness and safety of elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil fumarate single-tablet combination among HIV-infected patients in Turkey: results from a real world setting.
Aydin, OA; Bilge, B; Bolukcu, S; Dokmetas, I; Gumuser, F; Gunduz, A; Karaosmanoglu, HK; Mete, B; Tabak, F; Yildiz, DS, 2021
)
0.89
"Real world data on the effectiveness and safety of E/C/F/TDF is comparable with the phase 3 trial results Adverse events are uncommon and manageable."( Effectiveness and safety of elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil fumarate single-tablet combination among HIV-infected patients in Turkey: results from a real world setting.
Aydin, OA; Bilge, B; Bolukcu, S; Dokmetas, I; Gumuser, F; Gunduz, A; Karaosmanoglu, HK; Mete, B; Tabak, F; Yildiz, DS, 2021
)
0.89
" 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,
)
0.53
" We assessed discontinuations and adverse events (AEs) that were considered related to the initial INSTI in the first year following initiation among treatment-naïve people living with HIV (PLWH)."( Treatment-related early discontinuations and adverse events among newly diagnosed people living with HIV initiating integrase inhibitors in a real-world setting.
Castano, J; DeJesus, E; Hinestrosa, F; Nguyen, V; Patel, K; Rolle, CP, 2023
)
0.91
" 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
)
1.14

Pharmacokinetics

Pregnant women receiving TAF 10 mg with cobicistat or TAF 25 mg without boosting had intensive pharmacokinetic assessments performed during the second and third trimester.

ExcerptReferenceRelevance
"We performed a within-subject open-labeled pharmacokinetic and pharmacodynamic study in 17 HIV-seronegative subjects stabilized on at least 2 weeks of buprenorphine/naloxone therapy."( The pharmacokinetic and pharmacodynamic interactions between buprenorphine/naloxone and elvitegravir/cobicistat in subjects receiving chronic buprenorphine/naloxone treatment.
Bruce, RD; Custodio, JM; Friedland, GH; Kearney, BP; Ramanathan, S; Rhee, MS; Wei, LX; Winkle, P, 2013
)
0.61
"6 hr*ng/mL) and mean Cmax (8."( The pharmacokinetic and pharmacodynamic interactions between buprenorphine/naloxone and elvitegravir/cobicistat in subjects receiving chronic buprenorphine/naloxone treatment.
Bruce, RD; Custodio, JM; Friedland, GH; Kearney, BP; Ramanathan, S; Rhee, MS; Wei, LX; Winkle, P, 2013
)
0.61
" Lack of PK alteration was defined as 90% confidence intervals about the geometric least squares means ratio (coadministration:alone) being within 70%-143% for elvitegravir Cmax (maximum concentration), Ctau (trough), and AUCtau (area under plasma concentration-time curve; 0-24 hours); cobicistat PK were explored."( Pharmacokinetics of once-daily boosted elvitegravir when administered in combination with acid-reducing agents.
Cheng, A; Kearney, BP; Koziara, J; Mathias, A; Ramanathan, S; Shen, G; Wei, X, 2013
)
0.57
"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
)
3.29
" The pharmacokinetic interaction between cobicistat-boosted elvitegravir (EVG/co) and rosuvastatin was evaluated."( Pharmacokinetics of cobicistat boosted-elvitegravir administered in combination with rosuvastatin.
Andrews, J; Cheng, A; Custodio, JM; Hao, J; Kearney, BP; Lepist, EI; Ling, KH; Ramanathan, S; Ray, AS; Wang, H, 2014
)
0.99
" In this open-label, randomized, three-way crossover study, the pharmacokinetic profiles of elvitegravir, cobicistat, emtricitabine, and tenofovir were evaluated when administered with a standard breakfast, under fasting conditions, or with a nutritional protein-rich drink."( Effects of a protein-rich drink or a standard meal on the pharmacokinetics of elvitegravir, cobicistat, emtricitabine and tenofovir in healthy Japanese male subjects: a randomized, three-way crossover study.
Ikeda, A; Irie, S; Ishikawa, T; Kimura, M; Matsuki, S; Nishino, N; Shiomi, M, 2014
)
0.84
" Darunavir AUC24h following administration of the FDCs (G003: 74,780 ng ∙ h/mL and G004: 76,490 ng ∙ h/mL) was comparable to that following darunavir/ritonavir (78,410 ng ∙ h/mL), as was Cmax (6,666 and 6,917 ng/mL versus 6,973 ng/mL, respectively)."( Pharmacokinetics of darunavir in fixed-dose combination with cobicistat compared with coadministration of darunavir and ritonavir as single agents in healthy volunteers.
Hillewaert, V; Hoetelmans, RM; Iterbeke, K; Kakuda, TN; Opsomer, M; Petrovic, R; Timmers, M; Van De Casteele, T, 2014
)
0.64
" This is the basis of pharmacokinetic enhancement."( Pharmacokinetic enhancers in HIV therapeutics.
Acosta, EP; Delille, C; Larson, KB; Otofokun, I; Wang, K, 2014
)
0.4
"Inhibition of the cytochrome p450 3A4 enzyme system leads to increases in plasma concentrations of coadministered antiretroviral agents - a concept known as pharmacokinetic boosting."( Pharmacokinetic enhancement in HIV antiretroviral therapy: a comparison of ritonavir and cobicistat.
Bow, D; Ng, J; Norton, M; Renjifo, B; Salem, AH; van Wyk, J,
)
0.35
"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
)
2.05
"Elvitegravir and cobicistat pharmacokinetic variability appears to be mainly explained by drug-drug interactions that may be encountered in routine clinical practice."( Population pharmacokinetic analysis of elvitegravir and cobicistat in HIV-1-infected individuals.
Aouri, M; Barceló, C; Buclin, T; Cavassini, M; Csajka, C; Decosterd, LA; Gaspar, F; Guidi, M; Panchaud, A; Rotger, M, 2016
)
1.02
" The pharmacokinetic parameters (Cmax , AUCtau , and Ctau ) of elvitegravir were investigated at Day 10 after each treatment, together with safety and tolerability."( Pharmacokinetic and bioequivalence evaluation of single-tablet and separate-tablet regimens for once-daily cobicistat-boosted elvitegravir in healthy Japanese male subjects: A randomized, two-way crossover study.
Ikeda, A; Irie, S; Ishikawa, T; Kimura, M; Kumagai, Y; Matsuki, S; Nishino, N; Shiomi, M,
)
0.34
"Between May 22, 2013, and July 22, 2014, we enrolled 50 participants, and all 50 received the assigned treatment; 24 participated in the intensive pharmacokinetic assessment."( Safety, efficacy, and pharmacokinetics of a single-tablet regimen containing elvitegravir, cobicistat, emtricitabine, and tenofovir alafenamide in treatment-naive, HIV-infected adolescents: a single-arm, open-label trial.
Batra, J; Chakraborty, R; Gaur, AH; Kizito, H; Kosalaraksa, P; Luesomboon, W; Myers, M; Porter, D; Prasitsueubsai, W; Quirk, E; Rakhmanina, N; Rassool, M; Rhee, MS; SenGupta, D; Shao, Y; Ting, L, 2016
)
0.65
"The elvitegravir, cobicistat, emtricitabine, and tenofovir alafenamide regimen was well tolerated and achieved component plasma pharmacokinetic exposures similar to those in adults."( Safety, efficacy, and pharmacokinetics of a single-tablet regimen containing elvitegravir, cobicistat, emtricitabine, and tenofovir alafenamide in treatment-naive, HIV-infected adolescents: a single-arm, open-label trial.
Batra, J; Chakraborty, R; Gaur, AH; Kizito, H; Kosalaraksa, P; Luesomboon, W; Myers, M; Porter, D; Prasitsueubsai, W; Quirk, E; Rakhmanina, N; Rassool, M; Rhee, MS; SenGupta, D; Shao, Y; Ting, L, 2016
)
0.99
"EVG is currently available as a single tablet regimen and requires cobisistat, a pharmacokinetic booster and CYP3A inhibitor to allow QD dosing."( How recent findings on the pharmacokinetics and pharmacodynamics of integrase inhibitors can inform clinical use.
Boffito, M; Chirwa, M; Elliot, E, 2017
)
0.46
"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
)
0.98
"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
)
2.11
" 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
)
1.9
" The antiretroviral pharmacokinetic enhancers, ritonavir and cobicistat, inhibit both these transporters."( Differential Influence of the Antiretroviral Pharmacokinetic Enhancers Ritonavir and Cobicistat on Intestinal P-Glycoprotein Transport and the Pharmacokinetic/Pharmacodynamic Disposition of Dabigatran.
Alfaro, RM; Brooks, KM; George, JM; Gordon, LA; Hadigan, C; Kellogg, A; Kumar, P; Lozier, J; McManus, M; Nghiem, K; Penzak, SR, 2017
)
0.92
" 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
)
0.77
"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
)
1.04
"Intensive steady-state 24-h pharmacokinetic profiles after 150 mg of elvitegravir and 150 mg of cobicistat given orally in fixed dose combination once-daily were performed during the second trimester, third trimester, and postpartum."( Elvitegravir/cobicistat pharmacokinetics in pregnant and postpartum women with HIV.
Acosta, EP; Badell, ML; Barr, E; Best, BM; Burchett, S; Capparelli, EV; Chakhtoura, N; Mirochnick, M; Momper, JD; Park, K; Purswani, M; Shapiro, DE; Smith, E; Stek, A; Wang, J, 2018
)
1.07
"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
)
0.76
"The potential for clinically significant drug interactions (CSDIs) for patients taking ritonavir and cobicistat is high because of their powerful pharmacokinetic effect on the cytochrome P450 (CYP) enzyme system, most notably their inhibitory effect on CYP3A4."( Pharmacokinetic enhancers (cobicistat/ritonavir) and the potential for drug-drug interactions.
de Barra, E; Hollywood, P; Lorigan, D; MacCann, R; McConkey, S, 2020
)
1.07
"Two hundred individuals attending a regional specialist human immunodeficiency virus (HIV) clinic between June and September 2014 who were receiving the pharmacokinetic enhancers ritonavir or cobicistat were interviewed to determine a medication history including medications prescribed by their general practitioner (GP), over-the-counter (OTC) medicines, herbal remedies and recreational drugs."( Pharmacokinetic enhancers (cobicistat/ritonavir) and the potential for drug-drug interactions.
de Barra, E; Hollywood, P; Lorigan, D; MacCann, R; McConkey, S, 2020
)
1.04
" 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
)
0.86
"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
)
0.86
" 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
)
0.86
" The use of boosted regimens in pregnant women living with HIV has been studied for a variety of ARVs; however, a recent recommendation by the US Food and Drug Administration advised against cobicistat-boosted regimens in pregnancy due to substantially lower drug exposures observed in clinical pharmacokinetic studies."( Pharmacokinetic Enhancement of HIV Antiretroviral Therapy During Pregnancy.
Best, BM; Eke, AC; Mirochnick, M; Momper, JD; Salama, E, 2020
)
0.75
"Pregnant women receiving TAF 10 mg with cobicistat or TAF 25 mg without boosting as part of clinical care had intensive pharmacokinetic assessments performed during the second and third trimesters, and 6-12 weeks postpartum."( Pharmacokinetics of tenofovir alafenamide with and without cobicistat in pregnant and postpartum women living with HIV.
Barr, E; Best, BM; Brooks, KM; Capparelli, EV; Chakhtoura, N; Cielo, M; Denson, K; Deville, JG; Espina, R; Febo, IL; George, K; Haubrich, R; Mirochnick, M; Momper, JD; Pinilla, M; Rooney, JF; Rungruengthanakit, K; Shapiro, DE; Smith, E; Stek, AM; Weinberg, A, 2021
)
1.13
" We used physiologically-based pharmacokinetic (PBPK) modeling to simulate DDI magnitudes of various scenarios to guide the clinical DDI management of bictegravir."( Physiologically-Based Pharmacokinetic Modeling to Support the Clinical Management of Drug-Drug Interactions With Bictegravir.
Battegay, M; Marzolini, C; Stader, F, 2021
)
0.62
"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
)
1.11
" 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
)
0.84
"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
)
0.84
" 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
)
0.84
"Intensive steady-state 24-hour pharmacokinetic profiles were performed after administration of 300 mg of atazanavir and 150 mg of cobicistat orally in fixed-dose combination once daily during the second trimester, third trimester, and postpartum."( Pharmacokinetics of Atazanavir Boosted With Cobicistat in Pregnant and Postpartum Women With HIV.
Badell, ML; Best, BM; Browning, R; Capparelli, EV; Chakhtoura, N; Denson, K; George, K; Mirochnick, M; Momper, JD; Paul, ME; Powis, KM; Rungruengthanakit, K; Shapiro, DE; Stek, A; Wang, J, 2022
)
1.19
" We hypothesized that the oral bioavailability of ibrutinib is limited by CYP3A isoform-mediated metabolism, and that this pathway can be inhibited to improve the pharmacokinetic properties of ibrutinib."( Intentional Modulation of Ibrutinib Pharmacokinetics through CYP3A Inhibition.
Baker, SD; Byrd, JC; Eisenmann, ED; Fu, Q; Garrison, DA; Jin, Y; Muhowski, EM; Sparreboom, A; Uddin, ME; Weber, RH; Woyach, J, 2021
)
0.62
"Two multidose unboosted cohorts (n = 9) (SJ733, 300 mg and 600 mg daily for 3 days) followed by three single-dose boosted cohorts combining SJ733 (n = 18) (75-, 300-, or 600-mg single dose) with cobicistat (150-mg single dose) as a pharmacokinetic booster were evaluated in healthy volunteers (ClinicalTrials."( Combining SJ733, an oral ATP4 inhibitor of Plasmodium falciparum, with the pharmacokinetic enhancer cobicistat: An innovative approach in antimalarial drug development.
Bebrevska, L; Branum, KC; Chalon, S; Dallas, RH; Flynn, PM; Freeman, BB; Gaur, AH; Gusovsky, F; Guy, RK; Hammill, JT; Heine, RN; John, E; Maki, N; McCarthy, JS; Ost, S; Panetta, JC; Patel, ND; Richardson, JL; Smith, AM; Stewart, TB; Tang, L; Yanagi, T, 2022
)
1.13

Compound-Compound Interactions

Ritonavir and cobicistat are used as pharmacokinetic enhancers in combination with some antiretrovirals (ARVs) for the treatment of HIV. They are potent inhibitors of the CYP3A4 isoenzyme.

ExcerptReferenceRelevance
" Healthy subjects (N = 12) received a single dose of rosuvastatin 10 mg alone and in combination with EVG/co."( Pharmacokinetics of cobicistat boosted-elvitegravir administered in combination with rosuvastatin.
Andrews, J; Cheng, A; Custodio, JM; Hao, J; Kearney, BP; Lepist, EI; Ling, KH; Ramanathan, S; Ray, AS; Wang, H, 2014
)
0.73
"International, randomized double-blind active-controlled trial to evaluate the efficacy and safety of COBI vs ritonavir (RTV) as a pharmacoenhancer of atazanavir in combination with emtricitabine/tenofovir disoproxil fumarate in HIV treatment-naive patients followed through week 144."( Brief Report: Cobicistat Compared With Ritonavir as a Pharmacoenhancer for Atazanavir in Combination With Emtricitabine/Tenofovir Disoproxil Fumarate: Week 144 Results.
Abram, ME; Andrade-Villanueva, JF; Antunes, F; Arastéh, K; Cao, H; Chetchotisakd, P; DeJesus, E; Fehr, J; Gallant, JE; Koenig, E; Liu, HC; Rizzardini, G; Szwarcberg, J, 2015
)
0.78
" While studies have demonstrated that boosting with either cobicistat or ritonavir results in comparable plasma exposure of the target antiretroviral agent, a better understanding of drug-drug interactions between cobicistat- and ritonavir-boosted antiretrovirals and other medications will inform treatment decisions in HIV-infected patients."( Drug Interactions with Cobicistat- or Ritonavir-Boosted Elvitegravir.
Custodio, JM; McNicholl, I; Nguyen, T; Piontkowsky, D; Szwarcberg, J,
)
0.69
"Ritonavir and cobicistat, used as pharmacokinetic enhancers in combination with some antiretrovirals (ARVs) for the treatment of HIV, are potent inhibitors of the CYP3A4 isoenzyme."( Iatrogenic Cushing's syndrome due to drug interaction between glucocorticoids and the ritonavir or cobicistat containing HIV therapies.
Baldeweg, SE; Boffito, M; Elliot, ER; Jain, LR; Marshall, NJ; Theodoraki, A; Waters, LJ, 2016
)
1.01
"We investigated whether a fixed-dose combination tablet of elvitegravir, cobicistat, emtricitabine, and tenofovirDF (Stribild) can be crushed and combined with enteral nutrition without influencing pharmacokinetics."( Brief Report: Pharmacokinetics of Crushed Elvitegravir Combination Tablet Given With or Without Enteral Nutrition.
Abbink, E; Burger, D; Colbers, A; Duisenberg-van Essenberg, M; Jongbloed-de Hoon, M; Kruijssen, M; van Crevel, R; Velthoven-Graafland, K, 2017
)
0.69
"We report a diagnosis of exogenous steroid-induced hypoadrenalism in a person living with HIV caused by a drug-drug interaction (DDI) between intrabursal triamcinolone and the pharmacokinetic booster cobicistat."( Exogenous steroid-induced hypoadrenalism in a person living with HIV caused by a drug-drug interaction between cobicistat and intrabursal triamcinolone.
Macpherson, G; Makaram, N; Roberts, SB; Russell, CD; Stevens, J, 2018
)
0.88
" We report a potential drug-drug interaction in 2 female patients both receiving treatment for HIV and cerebral toxoplasmosis: one case between E/C/F/TAF with calcium carbonate and a second case involving leucovorin as calcium salt."( HIV Viral Rebound Due to a Possible Drug-Drug Interaction between Elvitegravir/Cobicistat/Emtricitabine/Tenofovir Alafenamide and Calcium-Containing Products: Report of 2 Cases.
El-Sayed, D; Kang-Birken, SL; Prichard, J,
)
0.36
"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
)
0.76
" Drug-drug interaction (DDI) studies were only conducted for strong inhibitors and inducers, leading to some uncertainty whether moderate perpetrators or multiple drug associations can be safely coadministered with bictegravir."( Physiologically-Based Pharmacokinetic Modeling to Support the Clinical Management of Drug-Drug Interactions With Bictegravir.
Battegay, M; Marzolini, C; Stader, F, 2021
)
0.62
"Neither pharmacokinetic nor pharmacodynamic drug-drug interactions between cobicistat and GHB were identified in this study."( Absence of drug-drug interactions between γ-hydroxybutyric acid (GHB) and cobicistat.
Bailón, L; Farré, M; Martín, S; Miranda, C; Moltó, J; Mothe, B; Papaseit, E; Pérez-Mañá, C, 2021
)
1.08
" The symptoms were presumably caused by a drug-drug interaction between an antiretroviral drug combination and atorvastatin."( [Rhabdomyolysis due to drug-drug interaction of atorvastatin and cobicistat].
Hennersdorf, F; Intert, E; Knop, K; Krause, M; Rosenkranz, T, 2022
)
0.96

Bioavailability

This study compared the bioavailability of two candidate fixed-dose combinations. The findings suggest that elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil fumarate should be administered with food.

ExcerptReferenceRelevance
" These findings suggest that elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil fumarate should be administered with food, and that the bioavailability of elvitegravir and tenofovir is not affected by the type of meal ingested."( Effects of a protein-rich drink or a standard meal on the pharmacokinetics of elvitegravir, cobicistat, emtricitabine and tenofovir in healthy Japanese male subjects: a randomized, three-way crossover study.
Ikeda, A; Irie, S; Ishikawa, T; Kimura, M; Matsuki, S; Nishino, N; Shiomi, M, 2014
)
0.89
"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
)
0.85
" Protease inhibitors (PIs), an important component of the antiretroviral armada, were historically associated with poor oral bioavailability and high pill burden."( Pharmacokinetic enhancers in HIV therapeutics.
Acosta, EP; Delille, C; Larson, KB; Otofokun, I; Wang, K, 2014
)
0.4
"This randomized, two-way crossover study evaluated the bioavailability of elvitegravir administered as the new individual tablet containing 150 mg and a cobicistat 150 mg tablet, concomitantly with a fixed-dose combination tablet containing 200 mg of emtricitabine and 300 mg of tenofovir disoproxil fumarate (EVG + COBI + FTC/TDF), in comparison with a single-tablet regimen containing the same dose of each component (EVG/COBI/FTC/TDF)."( Pharmacokinetic and bioequivalence evaluation of single-tablet and separate-tablet regimens for once-daily cobicistat-boosted elvitegravir in healthy Japanese male subjects: A randomized, two-way crossover study.
Ikeda, A; Irie, S; Ishikawa, T; Kimura, M; Kumagai, Y; Matsuki, S; Nishino, N; Shiomi, M,
)
0.54
" There was a delayed onset of symptoms in many cases, most likely due to the relatively lower systemic bioavailability of intranasal fluticasone."( Prescribing intranasal steroids in HIV-positive patients: systematic review of the literature.
McGilligan, JA; Mohammed, H; Richardson, D; Robinson, M; Seymour, N; Williams, D, 2021
)
0.62
" However, ibrutinib has very low oral bioavailability that contributes to significant variability in systemic exposure between patients, and this has the potential to affect both efficacy and toxicity."( Intentional Modulation of Ibrutinib Pharmacokinetics through CYP3A Inhibition.
Baker, SD; Byrd, JC; Eisenmann, ED; Fu, Q; Garrison, DA; Jin, Y; Muhowski, EM; Sparreboom, A; Uddin, ME; Weber, RH; Woyach, J, 2021
)
0.62

Dosage Studied

Phenomenal boosting of antiretroviral (ARV) therapies with either ritonavir or cobicistat is used to achieve target drug exposure, lower pill burden, and provide simplified dosing schedules. Standard darunavir/cobicist at dosing during pregnancy results in significantly lower exposure which may increase the risk of virologic failure and perinatal transmission.

ExcerptRelevanceReference
" Single-dose escalation from 50 to 400 mg resulted in a 164-fold increase in GS-9350 exposure, whereas multiple-dose escalation in the dosage range of 50-300 mg resulted in a 47-fold increase in exposure."( Pharmacokinetics and pharmacodynamics of GS-9350: a novel pharmacokinetic enhancer without anti-HIV activity.
German, P; Hui, J; Jain, A; Kearney, BP; Mathias, AA; Murray, BP; Warren, D; Wei, L; West, S, 2010
)
0.36
" Decreases in estimated glomerular filtration rate occurred within the first few weeks of dosing in participants receiving EVG/COBI/FTC/TDF, remained within the normal range and did not progress at week 24 or 48; no participant experienced a clinical adverse event or discontinued study drug due to changes in serum creatinine or renal function."( Randomized, phase 2 evaluation of two single-tablet regimens elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil fumarate versus efavirenz/emtricitabine/tenofovir disoproxil fumarate for the initial treatment of HIV infection.
Chuck, SL; Cohen, C; DeJesus, E; Elion, R; Kearney, BP; Liu, HC; Ramanathan, S; Rashbaum, B; Ruane, P; Shamblaw, D; Warren, DR; Yale, K, 2011
)
0.6
" This therapeutic principle called 'boosting' helped to overcome the obstacles of this class of drugs concerning unfavorable pharmacokinetics, resulting in either a high frequency of dosing or subtherapeutic plasma concentrations."( Safety of pharmacoenhancers for HIV therapy.
Haberl, A; von Hentig, N, 2012
)
0.38
"EVG exposures were 40%-50% lower upon simultaneous dosing of EVG/r and antacids, probably due to local complexation with cations in gastrointestinal tract, and were unaffected with ≥2 hours staggered dosing."( Pharmacokinetics of once-daily boosted elvitegravir when administered in combination with acid-reducing agents.
Cheng, A; Kearney, BP; Koziara, J; Mathias, A; Ramanathan, S; Shen, G; Wei, X, 2013
)
0.39
" As a fixed-dose combination tablet given once daily, EFV/FTC/TDF was the first available STR combining efficacy, tolerability and convenience, with the simplest dosing schedule and smallest numbers of pills of any ART combination therapy."( Single-tablet regimens in HIV: does it really make a difference?
Aldir, I; Horta, A; Serrado, M, 2014
)
0.4
" Compared to baseline values, the R-methadone mean area under the concentration-time curve to the end of the dosing period (AUCtau) (5,550 versus 6,210 h · ng/ml) and mean maximum concentration of drug in serum (Cmax) (316 versus 337 ng/ml) did not significantly increase in the presence of EVG/COBI."( Investigation of the interactions between methadone and elvitegravir-cobicistat in subjects receiving chronic methadone maintenance.
Bruce, RD; Custodio, JM; Friedland, GH; Kearney, BP; Ramanathan, S; Rhee, MS; Wei, X; Winkle, P, 2013
)
0.62
"Pill burden, dosing frequency, and concerns about safety and tolerability are important obstacles to maintaining adequate medication adherence."( Switching from twice-daily raltegravir plus tenofovir disoproxil fumarate/emtricitabine to once-daily elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil fumarate in virologically suppressed, HIV-1-infected subjects: 48 weeks data.
Brinson, C; Cao, H; Cheng, A; Crofoot, G; Ebrahimi, R; Garner, W; Kulkarni, R; Mills, A; Ortiz, R; Rashbaum, B; Szwarcberg, J; Towner, W; Ward, D,
)
0.34
"Patients with HIV on antiretroviral therapy might benefit from regimen simplification to reduce pill burden and dosing frequency."( Simplification to coformulated elvitegravir, cobicistat, emtricitabine, and tenofovir versus continuation of ritonavir-boosted protease inhibitor with emtricitabine and tenofovir in adults with virologically suppressed HIV (STRATEGY-PI): 48 week results o
Arribas, JR; Di Perri, G; Ebrahimi, R; Gathe, J; Nguyen, T; Pialoux, G; Piontkowsky, D; Reynes, J; Tebas, P; White, K, 2014
)
0.66
" However, because the PIs are metabolized by cytochrome P450 (CYP) 3A enzymes, intentional inhibition of these enzymes leads to higher drug exposure, lower pill burden, and therefore simplified dosing schedules with this class of drug."( Pharmacokinetic enhancers in HIV therapeutics.
Acosta, EP; Delille, C; Larson, KB; Otofokun, I; Wang, K, 2014
)
0.4
"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
)
2.05
"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
)
2.04
"A new simple, rapid stability indicating assay method has been developed and validated for the determination of emtricitabine, tenofovir disoproxil fumarate, elvitegravir and cobicistat using reverse-phase high-performance liquid chromatography in their pharmaceutical dosage form."( A Validated Stability Indicating RP-HPLC Method for the Determination of Emtricitabine, Tenofovir Disoproxil Fumarate, Elvitegravir and Cobicistat in Pharmaceutical Dosage Form.
Avanapu, SR; Kumar, PR; Runja, C,
)
0.53
" 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
)
1.88
" Simulations based on the final model served to compare expected drug concentrations under standard and alternative dosage regimens."( Population pharmacokinetic analysis of elvitegravir and cobicistat in HIV-1-infected individuals.
Aouri, M; Barceló, C; Buclin, T; Cavassini, M; Csajka, C; Decosterd, LA; Gaspar, F; Guidi, M; Panchaud, A; Rotger, M, 2016
)
0.68
"Cobicistat and ritonavir are structurally distinct compounds that both potently inhibit cytochrome P450 (CYP) 3A, the metabolizing enzyme primarily responsible for the elimination of several antiretroviral medications, and, as such, are pharmacokinetic boosters for antiretroviral agents that require longer dosing intervals."( Drug Interactions with Cobicistat- or Ritonavir-Boosted Elvitegravir.
Custodio, JM; McNicholl, I; Nguyen, T; Piontkowsky, D; Szwarcberg, J,
)
1.88
" When combined with the CYP3A4 substrate daclatasvir, the daclatasvir dosage should be reduced from 60 to 30 mg once daily."( Daclatasvir 30 mg/day is the correct dose for patients taking atazanavir/cobicistat.
Burger, DM; Colbers, EP; de Kanter, CT; Drenth, JP; Smolders, EJ; Velthoven-Graafland, K, 2017
)
0.69
" 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
)
1.08
"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
" Darunavir concentrations at the end of the dosing interval were quantified by LC-MS/MS."( Darunavir concentrations in CSF of HIV-infected individuals when boosted with cobicistat versus ritonavir.
Bartels, H; Battegay, M; Decosterd, L; Marzolini, C, 2017
)
0.68
" Therefore, it is likely safe to coadminister ritonavir with DE, while there is a potential need for reduced dosing and prudent clinical monitoring with the coadministration of cobicistat due to the greater net inhibition of intestinal P-gp transport and increased bioavailability."( Differential Influence of the Antiretroviral Pharmacokinetic Enhancers Ritonavir and Cobicistat on Intestinal P-Glycoprotein Transport and the Pharmacokinetic/Pharmacodynamic Disposition of Dabigatran.
Alfaro, RM; Brooks, KM; George, JM; Gordon, LA; Hadigan, C; Kellogg, A; Kumar, P; Lozier, J; McManus, M; Nghiem, K; Penzak, SR, 2017
)
0.87
"Standard elvitegravir and cobicistat dosing during pregnancy results in significantly lower exposure which may increase the risk of virologic failure and mother-to-child transmission."( Elvitegravir/cobicistat pharmacokinetics in pregnant and postpartum women with HIV.
Acosta, EP; Badell, ML; Barr, E; Best, BM; Burchett, S; Capparelli, EV; Chakhtoura, N; Mirochnick, M; Momper, JD; Park, K; Purswani, M; Shapiro, DE; Smith, E; Stek, A; Wang, J, 2018
)
1.15
" At baseline (pre-switch) and at 12 weeks post-switch, we measured HIV-1 RNA in seminal plasma (SP) and blood plasma (BP), tenofovir (TFV) in SP and BP, and TFV-diphosphate (dp) in peripheral blood mononuclear cells (PBMCs) and seminal mononuclear cells (SMCs) at the end of the dosing interval (C24h)."( Seminal Tenofovir Concentrations, Viral Suppression, and Semen Quality With Tenofovir Alafenamide, Compared With Tenofovir Disoproxil Fumarate (Spanish HIV/AIDS Research Network, PreEC/RIS 40).
Cottrell, ML; Garcia, B; Imaz, A; Kashuba, ADM; Morenilla, S; Niubó, J; Perez, E; Podzamczer, D; Tiraboschi, JM, 2019
)
0.51
"High urine FTC and TFV concentrations could provide an indication of adherence to daily oral dosing with TDF or TAF-based regimens used for treatment and prevention."( Brief Report: Urine Emtricitabine and Tenofovir Concentrations Provide Markers of Recent Antiretroviral Drug Exposure Among HIV-Negative Men Who Have Sex With Men.
Conway-Washington, C; Dinh, C; Fountain, J; Haaland, RE; Hall, L; Holder, A; Kelley, CF; Livermont, T; Lupo, LD; Martin, A, 2019
)
0.51
" 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
)
1.12
"Pharmacokinetic boosting of antiretroviral (ARV) therapies with either ritonavir or cobicistat is used to achieve target drug exposure, lower pill burden, and provide simplified dosing schedules."( Pharmacokinetic Enhancement of HIV Antiretroviral Therapy During Pregnancy.
Best, BM; Eke, AC; Mirochnick, M; Momper, JD; Salama, E, 2020
)
0.78
" 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
)
0.83
" Reduced maternal concentrations may have a clinically important impact on the efficacy of anti-infectives for mother, fetus, and neonate, with potential dosing implications."( Anti-Infective Dosing in Special Populations: Pregnancy.
Busuulwa, P; Hazenberg, P; Navaratnam, K; Waitt, C, 2021
)
0.62
" Glans swab emtricitabine and darunavir concentrations peaked 24 h after dosing (emtricitabine 14 ng/swab, ( Antiretroviral drug exposure in urethral and glans surface sampling of the penis.
Conway-Washington, C; Dinh, C; Fountain, J; Garcia-Lerma, JG; Haaland, RE; Hall, L; Heneine, W; Kelley, CF; Lupo, LD; Martin, A, 2021
)
0.62
"We document ARV dosing in the urethra and on the glans surface with high drug concentrations noted for emtricitabine and darunavir and lower tenofovir and elvitegravir concentrations."( Antiretroviral drug exposure in urethral and glans surface sampling of the penis.
Conway-Washington, C; Dinh, C; Fountain, J; Garcia-Lerma, JG; Haaland, RE; Hall, L; Heneine, W; Kelley, CF; Lupo, LD; Martin, A, 2021
)
0.62
"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
)
1.23
"Standard atazanavir/cobicistat dosing during pregnancy results in lower exposure which may increase the risk of virologic failure and perinatal transmission."( Pharmacokinetics of Atazanavir Boosted With Cobicistat in Pregnant and Postpartum Women With HIV.
Badell, ML; Best, BM; Browning, R; Capparelli, EV; Chakhtoura, N; Denson, K; George, K; Mirochnick, M; Momper, JD; Paul, ME; Powis, KM; Rungruengthanakit, K; Shapiro, DE; Stek, A; Wang, J, 2022
)
1.31
"Determining the appropriate dosing regimens of antiretroviral (ARV) drugs for pregnant individuals living with HIV-1 infection is critical to maximize maternal health and prevent perinatal HIV transmission."( Pharmacokinetics of Antiretroviral Agents in Pregnant Individuals Living With HIV: Current Status and Considerations for Study Design and Interpretation.
Belew, Y; Choi, SY; Reynolds, K; Struble, K; Yang, X, 2023
)
0.91
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Roles (1)

RoleDescription
P450 inhibitorAn enzyme inhibitor that interferes with the activity of cytochrome P450 involved in catalysis of organic substances.
[role information is derived from Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Drug Classes (5)

ClassDescription
1,3-thiazoles
morpholinesAny compound containing morpholine as part of its structure.
ureas
carbamate esterAny ester of carbamic acid or its N-substituted derivatives.
monocarboxylic acid amideA carboxamide derived from a monocarboxylic acid.
[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 (7)

Inhibition Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Cytochrome P450 1A2Homo sapiens (human)IC50 (µMol)25.00000.00011.774010.0000AID1069067
Cytochrome P450 3A4Homo sapiens (human)IC50 (µMol)0.15000.00011.753610.0000AID1069062
Cytochrome P450 2D6Homo sapiens (human)IC50 (µMol)9.20000.00002.015110.0000AID1069066
Cytochrome P450 2C9 Homo sapiens (human)IC50 (µMol)25.00000.00002.800510.0000AID1069069
Cytochrome P450 2C19Homo sapiens (human)IC50 (µMol)25.00000.00002.398310.0000AID1069064
Broad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)IC50 (µMol)59.00000.00401.966610.0000AID1873211
Protease Human immunodeficiency virus 1IC50 (µMol)30.00000.00000.81769.8500AID1069063
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (56)

Processvia Protein(s)Taxonomy
steroid catabolic processCytochrome P450 1A2Homo sapiens (human)
porphyrin-containing compound metabolic processCytochrome P450 1A2Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 1A2Homo sapiens (human)
cholesterol metabolic processCytochrome P450 1A2Homo sapiens (human)
estrogen metabolic processCytochrome P450 1A2Homo sapiens (human)
toxin biosynthetic processCytochrome P450 1A2Homo sapiens (human)
post-embryonic developmentCytochrome P450 1A2Homo sapiens (human)
alkaloid metabolic processCytochrome P450 1A2Homo sapiens (human)
regulation of gene expressionCytochrome P450 1A2Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 1A2Homo sapiens (human)
dibenzo-p-dioxin metabolic processCytochrome P450 1A2Homo sapiens (human)
epoxygenase P450 pathwayCytochrome P450 1A2Homo sapiens (human)
lung developmentCytochrome P450 1A2Homo sapiens (human)
methylationCytochrome P450 1A2Homo sapiens (human)
monocarboxylic acid metabolic processCytochrome P450 1A2Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 1A2Homo sapiens (human)
retinol metabolic processCytochrome P450 1A2Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 1A2Homo sapiens (human)
cellular respirationCytochrome P450 1A2Homo sapiens (human)
aflatoxin metabolic processCytochrome P450 1A2Homo sapiens (human)
hydrogen peroxide biosynthetic processCytochrome P450 1A2Homo sapiens (human)
oxidative demethylationCytochrome P450 1A2Homo sapiens (human)
cellular response to cadmium ionCytochrome P450 1A2Homo sapiens (human)
omega-hydroxylase P450 pathwayCytochrome P450 1A2Homo sapiens (human)
lipid hydroxylationCytochrome P450 3A4Homo sapiens (human)
lipid metabolic processCytochrome P450 3A4Homo sapiens (human)
steroid catabolic processCytochrome P450 3A4Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 3A4Homo sapiens (human)
steroid metabolic processCytochrome P450 3A4Homo sapiens (human)
cholesterol metabolic processCytochrome P450 3A4Homo sapiens (human)
androgen metabolic processCytochrome P450 3A4Homo sapiens (human)
estrogen metabolic processCytochrome P450 3A4Homo sapiens (human)
alkaloid catabolic processCytochrome P450 3A4Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 3A4Homo sapiens (human)
calcitriol biosynthetic process from calciolCytochrome P450 3A4Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 3A4Homo sapiens (human)
vitamin D metabolic processCytochrome P450 3A4Homo sapiens (human)
vitamin D catabolic processCytochrome P450 3A4Homo sapiens (human)
retinol metabolic processCytochrome P450 3A4Homo sapiens (human)
retinoic acid metabolic processCytochrome P450 3A4Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 3A4Homo sapiens (human)
aflatoxin metabolic processCytochrome P450 3A4Homo sapiens (human)
oxidative demethylationCytochrome P450 3A4Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 2D6Homo sapiens (human)
steroid metabolic processCytochrome P450 2D6Homo sapiens (human)
cholesterol metabolic processCytochrome P450 2D6Homo sapiens (human)
estrogen metabolic processCytochrome P450 2D6Homo sapiens (human)
coumarin metabolic processCytochrome P450 2D6Homo sapiens (human)
alkaloid metabolic processCytochrome P450 2D6Homo sapiens (human)
alkaloid catabolic processCytochrome P450 2D6Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 2D6Homo sapiens (human)
isoquinoline alkaloid metabolic processCytochrome P450 2D6Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 2D6Homo sapiens (human)
retinol metabolic processCytochrome P450 2D6Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 2D6Homo sapiens (human)
negative regulation of bindingCytochrome P450 2D6Homo sapiens (human)
oxidative demethylationCytochrome P450 2D6Homo sapiens (human)
negative regulation of cellular organofluorine metabolic processCytochrome P450 2D6Homo sapiens (human)
arachidonic acid metabolic processCytochrome P450 2D6Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 2C9 Homo sapiens (human)
steroid metabolic processCytochrome P450 2C9 Homo sapiens (human)
cholesterol metabolic processCytochrome P450 2C9 Homo sapiens (human)
estrogen metabolic processCytochrome P450 2C9 Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 2C9 Homo sapiens (human)
epoxygenase P450 pathwayCytochrome P450 2C9 Homo sapiens (human)
urea metabolic processCytochrome P450 2C9 Homo sapiens (human)
monocarboxylic acid metabolic processCytochrome P450 2C9 Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 2C9 Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 2C9 Homo sapiens (human)
amide metabolic processCytochrome P450 2C9 Homo sapiens (human)
icosanoid biosynthetic processCytochrome P450 2C9 Homo sapiens (human)
oxidative demethylationCytochrome P450 2C9 Homo sapiens (human)
omega-hydroxylase P450 pathwayCytochrome P450 2C9 Homo sapiens (human)
long-chain fatty acid metabolic processCytochrome P450 2C19Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 2C19Homo sapiens (human)
steroid metabolic processCytochrome P450 2C19Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 2C19Homo sapiens (human)
epoxygenase P450 pathwayCytochrome P450 2C19Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 2C19Homo sapiens (human)
omega-hydroxylase P450 pathwayCytochrome P450 2C19Homo sapiens (human)
lipid transportBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
organic anion transportBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
urate transportBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
biotin transportBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
sphingolipid biosynthetic processBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
riboflavin transportBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
urate metabolic processBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
transmembrane transportBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
transepithelial transportBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
renal urate salt excretionBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
export across plasma membraneBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
transport across blood-brain barrierBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
cellular detoxificationBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
xenobiotic transport across blood-brain barrierBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (47)

Processvia Protein(s)Taxonomy
monooxygenase activityCytochrome P450 1A2Homo sapiens (human)
iron ion bindingCytochrome P450 1A2Homo sapiens (human)
protein bindingCytochrome P450 1A2Homo sapiens (human)
electron transfer activityCytochrome P450 1A2Homo sapiens (human)
oxidoreductase activityCytochrome P450 1A2Homo sapiens (human)
oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen, reduced flavin or flavoprotein as one donor, and incorporation of one atom of oxygenCytochrome P450 1A2Homo sapiens (human)
enzyme bindingCytochrome P450 1A2Homo sapiens (human)
heme bindingCytochrome P450 1A2Homo sapiens (human)
demethylase activityCytochrome P450 1A2Homo sapiens (human)
caffeine oxidase activityCytochrome P450 1A2Homo sapiens (human)
aromatase activityCytochrome P450 1A2Homo sapiens (human)
estrogen 16-alpha-hydroxylase activityCytochrome P450 1A2Homo sapiens (human)
estrogen 2-hydroxylase activityCytochrome P450 1A2Homo sapiens (human)
hydroperoxy icosatetraenoate dehydratase activityCytochrome P450 1A2Homo sapiens (human)
monooxygenase activityCytochrome P450 3A4Homo sapiens (human)
steroid bindingCytochrome P450 3A4Homo sapiens (human)
iron ion bindingCytochrome P450 3A4Homo sapiens (human)
protein bindingCytochrome P450 3A4Homo sapiens (human)
steroid hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
retinoic acid 4-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
oxidoreductase activityCytochrome P450 3A4Homo sapiens (human)
oxygen bindingCytochrome P450 3A4Homo sapiens (human)
enzyme bindingCytochrome P450 3A4Homo sapiens (human)
heme bindingCytochrome P450 3A4Homo sapiens (human)
vitamin D3 25-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
caffeine oxidase activityCytochrome P450 3A4Homo sapiens (human)
quinine 3-monooxygenase activityCytochrome P450 3A4Homo sapiens (human)
testosterone 6-beta-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
1-alpha,25-dihydroxyvitamin D3 23-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
anandamide 8,9 epoxidase activityCytochrome P450 3A4Homo sapiens (human)
anandamide 11,12 epoxidase activityCytochrome P450 3A4Homo sapiens (human)
anandamide 14,15 epoxidase activityCytochrome P450 3A4Homo sapiens (human)
aromatase activityCytochrome P450 3A4Homo sapiens (human)
vitamin D 24-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
estrogen 16-alpha-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
estrogen 2-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
1,8-cineole 2-exo-monooxygenase activityCytochrome P450 3A4Homo sapiens (human)
monooxygenase activityCytochrome P450 2D6Homo sapiens (human)
iron ion bindingCytochrome P450 2D6Homo sapiens (human)
oxidoreductase activityCytochrome P450 2D6Homo sapiens (human)
oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen, reduced flavin or flavoprotein as one donor, and incorporation of one atom of oxygenCytochrome P450 2D6Homo sapiens (human)
heme bindingCytochrome P450 2D6Homo sapiens (human)
anandamide 8,9 epoxidase activityCytochrome P450 2D6Homo sapiens (human)
anandamide 11,12 epoxidase activityCytochrome P450 2D6Homo sapiens (human)
anandamide 14,15 epoxidase activityCytochrome P450 2D6Homo sapiens (human)
monooxygenase activityCytochrome P450 2C9 Homo sapiens (human)
iron ion bindingCytochrome P450 2C9 Homo sapiens (human)
arachidonic acid epoxygenase activityCytochrome P450 2C9 Homo sapiens (human)
steroid hydroxylase activityCytochrome P450 2C9 Homo sapiens (human)
arachidonic acid 14,15-epoxygenase activityCytochrome P450 2C9 Homo sapiens (human)
arachidonic acid 11,12-epoxygenase activityCytochrome P450 2C9 Homo sapiens (human)
oxidoreductase activityCytochrome P450 2C9 Homo sapiens (human)
(S)-limonene 6-monooxygenase activityCytochrome P450 2C9 Homo sapiens (human)
(S)-limonene 7-monooxygenase activityCytochrome P450 2C9 Homo sapiens (human)
caffeine oxidase activityCytochrome P450 2C9 Homo sapiens (human)
(R)-limonene 6-monooxygenase activityCytochrome P450 2C9 Homo sapiens (human)
aromatase activityCytochrome P450 2C9 Homo sapiens (human)
heme bindingCytochrome P450 2C9 Homo sapiens (human)
oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen, reduced flavin or flavoprotein as one donor, and incorporation of one atom of oxygenCytochrome P450 2C9 Homo sapiens (human)
monooxygenase activityCytochrome P450 2C19Homo sapiens (human)
iron ion bindingCytochrome P450 2C19Homo sapiens (human)
steroid hydroxylase activityCytochrome P450 2C19Homo sapiens (human)
oxidoreductase activityCytochrome P450 2C19Homo sapiens (human)
(S)-limonene 6-monooxygenase activityCytochrome P450 2C19Homo sapiens (human)
(S)-limonene 7-monooxygenase activityCytochrome P450 2C19Homo sapiens (human)
oxygen bindingCytochrome P450 2C19Homo sapiens (human)
enzyme bindingCytochrome P450 2C19Homo sapiens (human)
heme bindingCytochrome P450 2C19Homo sapiens (human)
(R)-limonene 6-monooxygenase activityCytochrome P450 2C19Homo sapiens (human)
aromatase activityCytochrome P450 2C19Homo sapiens (human)
long-chain fatty acid omega-1 hydroxylase activityCytochrome P450 2C19Homo sapiens (human)
arachidonic acid epoxygenase activityCytochrome P450 2C19Homo sapiens (human)
oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen, reduced flavin or flavoprotein as one donor, and incorporation of one atom of oxygenCytochrome P450 2C19Homo sapiens (human)
protein bindingBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
ATP bindingBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
organic anion transmembrane transporter activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
ABC-type xenobiotic transporter activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
urate transmembrane transporter activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
biotin transmembrane transporter activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
efflux transmembrane transporter activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
ATP hydrolysis activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
riboflavin transmembrane transporter activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
ATPase-coupled transmembrane transporter activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
identical protein bindingBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
protein homodimerization activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
xenobiotic transmembrane transporter activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
sphingolipid transporter activityBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (12)

Processvia Protein(s)Taxonomy
endoplasmic reticulum membraneCytochrome P450 1A2Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 1A2Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 1A2Homo sapiens (human)
cytoplasmCytochrome P450 3A4Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 3A4Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 3A4Homo sapiens (human)
mitochondrionCytochrome P450 2D6Homo sapiens (human)
endoplasmic reticulumCytochrome P450 2D6Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 2D6Homo sapiens (human)
cytoplasmCytochrome P450 2D6Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2D6Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 2C9 Homo sapiens (human)
plasma membraneCytochrome P450 2C9 Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2C9 Homo sapiens (human)
cytoplasmCytochrome P450 2C9 Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2C9 Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 2C19Homo sapiens (human)
plasma membraneCytochrome P450 2C19Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2C19Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2C19Homo sapiens (human)
cytoplasmCytochrome P450 2C19Homo sapiens (human)
nucleoplasmBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
plasma membraneBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
apical plasma membraneBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
brush border membraneBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
mitochondrial membraneBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
membrane raftBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
external side of apical plasma membraneBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
plasma membraneBroad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (11)

Assay IDTitleYearJournalArticle
AID1069065Antiviral activity against HIV1 by cell based assay2014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Structure-activity relationships of diamine inhibitors of cytochrome P450 (CYP) 3A as novel pharmacoenhancers. Part II: P2/P3 region and discovery of cobicistat (GS-9350).
AID1069067Inhibition of human CYP1A22014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Structure-activity relationships of diamine inhibitors of cytochrome P450 (CYP) 3A as novel pharmacoenhancers. Part II: P2/P3 region and discovery of cobicistat (GS-9350).
AID1069069Inhibition of human CYP2C92014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Structure-activity relationships of diamine inhibitors of cytochrome P450 (CYP) 3A as novel pharmacoenhancers. Part II: P2/P3 region and discovery of cobicistat (GS-9350).
AID1069068Activation of human PXR by cell based luciferase reporter gene assay2014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Structure-activity relationships of diamine inhibitors of cytochrome P450 (CYP) 3A as novel pharmacoenhancers. Part II: P2/P3 region and discovery of cobicistat (GS-9350).
AID1069063Inhibition of HIV-1 protease2014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Structure-activity relationships of diamine inhibitors of cytochrome P450 (CYP) 3A as novel pharmacoenhancers. Part II: P2/P3 region and discovery of cobicistat (GS-9350).
AID1873211Inhibition of ABCG2 (unknown origin) expressed in dog MDCK-II-BCRP cells assessed as Hoechst 33342 accumulation using Hoechst 33342 as substrate by microplate reader analysis2022European journal of medicinal chemistry, Jul-05, Volume: 237Targeting breast cancer resistance protein (BCRP/ABCG2): Functional inhibitors and expression modulators.
AID1069062Inhibition of human CYP3A4-mdiated midazolam 1'-hydroxylase activity2014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Structure-activity relationships of diamine inhibitors of cytochrome P450 (CYP) 3A as novel pharmacoenhancers. Part II: P2/P3 region and discovery of cobicistat (GS-9350).
AID1069064Inhibition of human CYP2C192014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Structure-activity relationships of diamine inhibitors of cytochrome P450 (CYP) 3A as novel pharmacoenhancers. Part II: P2/P3 region and discovery of cobicistat (GS-9350).
AID1069066Inhibition of human CYP2D62014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Structure-activity relationships of diamine inhibitors of cytochrome P450 (CYP) 3A as novel pharmacoenhancers. Part II: P2/P3 region and discovery of cobicistat (GS-9350).
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.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (283)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's0 (0.00)18.2507
2000's0 (0.00)29.6817
2010's177 (62.54)24.3611
2020's106 (37.46)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 58.30

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 Index58.30 (24.57)
Research Supply Index5.93 (2.92)
Research Growth Index6.91 (4.65)
Search Engine Demand Index93.06 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (58.30)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials75 (25.00%)5.53%
Reviews45 (15.00%)6.00%
Case Studies41 (13.67%)4.05%
Observational19 (6.33%)0.25%
Other120 (40.00%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (79)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
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
IntenSify: An Open-label Phase I Trial of the CYP3A Inhibitor Cobicistat and the Cytostatics Gemcitabine and Nab-Paclitaxel in Patients With Advanced Stage or Metastatic Pancreatic Ductal Adenocarcinoma to Evaluate the Combination's Pharmacokinetics, Safe [NCT05494866]Phase 118 participants (Anticipated)Interventional2022-12-07Recruiting
A Phase 1, Open-label Drug Interaction Study of PBI-200 With Ritonavir or Cobicistat in Healthy Volunteers [NCT05692570]Phase 121 participants (Actual)Interventional2022-09-09Completed
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
A Phase IV, Open-label Three-arm Study Investigating the Impact of a Combination of Tenofovir Disoproxil Fumarate/Emtricitabine With Raltegravir or Dolutegravir or Elvitegravir/Cobicistat on Renal Tubular Function and Renal Transporters in HIV-1 Antiretro [NCT02351908]Phase 460 participants (Actual)Interventional2015-03-31Completed
Assessment of the Taste Properties of Atazanavir and Cobicistat and Pediatric Oral Test Formulations Containing Both Atazanavir and Cobicistat in Healthy Adults [NCT02307656]Phase 16 participants (Actual)Interventional2014-12-12Completed
Bioequivalence of Crushed and Whole Genvoya Tablets (Elvitegravir/Cobicistat/Emtricitabine/Tenofovir Alafenamide Fumarate) [NCT03717129]Phase 412 participants (Actual)Interventional2019-04-15Completed
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 3, Randomized, Double-Blind Study to Evaluate the Safety and Efficacy of GS-9350-boosted Atazanavir Versus Ritonavir-boosted Atazanavir Each Administered With Emtricitabine/Tenofovir Disoproxil Fumarate in HIV-1 Infected, Antiretroviral Treatment- [NCT01108510]Phase 3698 participants (Actual)Interventional2010-04-30Completed
Changes in Insulin Resistance in Healthy Volunteers on STRIBILD® Medication - A Controlled, Mono Center, Three-arm, Randomized Phase I Study. [NCT02203461]Phase 130 participants (Actual)Interventional2014-07-31Completed
Is Dual Therapy as Effective as Triple Therapy Regarding CD4+/CD8+ Ratio Recovery and Improvement of Immune Activation and Inflammation in HIV-infected Patients With Consistent Plasma Viral Load Suppression (Tridual) [NCT03447873]Phase 4153 participants (Actual)Interventional2017-06-01Completed
Evaluation of Elvitegravir/Cobicistat/Emtricitabine/Tenofovir Alafenamide (E/C/F/TAF) Switch Followed by Sofosbuvir/Velpatasvir (SOF/VEL) Antiviral HCV Therapy Followed by Bictegravir/Emtricitabine/Tenofovir Alafenamide (B/F/TAF) Simplification in HIV-HCV [NCT03549312]Phase 425 participants (Anticipated)Interventional2018-02-01Recruiting
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
Change in Sleep Architecture and Neuropsychological Performance Following Switch From Atripla to Stribild. [NCT02283060]Phase 430 participants (Anticipated)Interventional2014-09-30Recruiting
Pharmacokinetic Properties of Antiretroviral and Related Drugs During Pregnancy and Postpartum [NCT00042289]1,578 participants (Actual)Observational2003-06-09Completed
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)
Clinical Trial to Evaluate Pharmacological Interactions Between γ-hydroxybutyrate (GHB) and Cobicistat [NCT04322214]Phase 112 participants (Actual)Interventional2020-01-30Completed
Efficacy and Safety of Darunavir and Cobicistat for Treatment of COVID-19 [NCT04252274]Phase 330 participants (Anticipated)Interventional2020-01-30Recruiting
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
An Open-label, Randomized Crossover Study to Obtain a Preliminary Estimate of the Bioavailability of Atazanavir and Cobicistat When Administered in an Age-appropriate Fixed-Dose Combination Formulation Compared With Coadministration of the Age-appropriate [NCT04263350]Phase 134 participants (Actual)Interventional2020-02-26Completed
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
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
A Phase 2/3, Multicenter, Open-label, Multicohort Study Evaluating Pharmacokinetics (PK), Safety, and Efficacy of Cobicistat-boosted Atazanavir (ATV/co) or Cobicistat-boosted Darunavir (DRV/co) and Emtricitabine/Tenofovir Alafenamide (F/TAF) in HIV-1 Infe [NCT02016924]Phase 2/Phase 3130 participants (Anticipated)Interventional2014-01-16Recruiting
Prophylaxis for HIV-1: Tenofovir/Emtricitabine (Truvada ®) + Lopinavir/Ritonavir (Kaletra ®) vs Tenofovir/Emtricitabine/Cobicistat/Elvitegravir (Stribild ®). Prospective, Randomized, Open. [NCT02198443]Phase 4160 participants (Actual)Interventional2015-06-06Completed
A Phase 2 Master Protocol to Assess the Efficacy and Safety of FORE8394, an Inhibitor of BRAF Class 1 and Class 2 Alterations, in Participants With Cancer Harboring BRAF Alterations [NCT05503797]Phase 2135 participants (Anticipated)Interventional2023-02-21Recruiting
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
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
A Single Arm Phase II Trial to Assess Cobicistat Boosted Venetoclax in Combination With Azacitidine (sc) in Adult Patients With Newly Diagnosed Acute Myeloid Leukaemia (AML) Who Are Not Considered Candidates for Intensive Treatment Regimens [NCT06014489]Phase 2142 participants (Anticipated)Interventional2023-12-31Not yet recruiting
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 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
Phase 1a, First-In-Human, Dose-Escalation Study of (+)-SJ000557733 (SJ733), an Oral, Novel Inhibitor of Plasmodium Falciparum Plasma Membrane Protein PfATP4 [NCT02661373]Phase 144 participants (Actual)Interventional2016-03-01Completed
A Drug-drug Interaction Study Between the Novel Anti-HCV Agent Daclatasvir and the Antiretroviral Agents Atazanavir/Ritonavir or Atazanavir/Cobicistat in Healthy Volunteers [NCT02565888]Phase 116 participants (Actual)Interventional2015-11-30Completed
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 2, Randomized, Double-Blinded Study of the Safety and Efficacy of GS-9350-boosted Atazanavir (ATV/GS-9350) Compared to Ritonavir-boosted Atazanavir (ATV/r) in Combination With Emtricitabine/Tenofovir Disoproxil Fumarate (FTC/TDF) in HIV-1 Infected [NCT00892437]Phase 285 participants (Actual)Interventional2009-05-31Completed
The Influence of Cobicistat or Ritonavir on Dabigatran Pharmacokinetics and Pharmacodynamics in Healthy Volunteers [NCT01896622]Phase 140 participants (Actual)Interventional2013-06-18Completed
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
Pharmacokinetics of DOLUTEGRAVIR Once Daily and ELVITEGRAVIR/COBICISTAT Once Daily Over 10 Days Following Drug Intake Cessation in Healthy Volunteers [NCT02219217]Phase 117 participants (Actual)Interventional2014-10-31Completed
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
The Effect of Atazanavir/Cobicistat on the Pharmacokinetics of an Oral Contraceptive Containing Ethinylestradiol and Levonorgestrel (Microgynon 30®) in Healthy Women [NCT02697851]Phase 113 participants (Actual)Interventional2016-07-31Terminated(stopped due to "Insufficient enrolment and business reasons")
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)
Improving Osimertinib Exposure and Cost-effectiveness Using Pharmacokinetic Boosting With Cobicistat [NCT05748093]Phase 460 participants (Anticipated)Interventional2023-06-01Not yet recruiting
Body Compartment Pharmacokinetics of Anti-retroviral Agents That May be Considered for Future On-demand Peri-exposure HIV Prophylaxis Regimens [NCT03976752]Phase 141 participants (Actual)Interventional2019-03-13Completed
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
Pharmacokinetic Boosting of Osimertinib in Patients With Non-small Cell Lung Cancer. [NCT03858491]Early Phase 111 participants (Actual)Interventional2020-11-01Completed
An Open-Label, Randomized, Single-Dose, 3-Treatment, 3-Period, 3-Sequence, Crossover Design, Relative Bioavailability Study Comparing the Pharmacokinetics of Atazanavir and Cobicistat Between the Coadministration of Age-Appropriate Mini-Tablet Formulation [NCT05236738]Phase 142 participants (Actual)Interventional2022-05-13Completed
Contribution of the Integrase Inhibitor Dolutegravir to Obesity and Cardiovascular Disease in Persons Living With HIV [NCT04340388]Phase 410 participants (Actual)Interventional2020-09-17Completed
A Phase 1/2, Open-label, Dose-Escalation and Dose-Expansion Cohort Study of SNDX-5613 in Patients With Relapsed/Refractory Leukemias, Including Those Harboring an MLL/KMT2A Gene Rearrangement or Nucleophosmin 1 (NPM1) Mutation [NCT04065399]Phase 1/Phase 2440 participants (Anticipated)Interventional2019-11-05Recruiting
Cost-effectiveness of Different Antiretroviral Treatment in Patients HIV Naive. Randomized Clinical, Not Masked, Trial Comparing DRVr3TC, ABC3TC (Kivexa) RPV, or EVG COBI FTC TDF (Stribild) for 48 Weeks [NCT02470650]Phase 4150 participants (Anticipated)Interventional2015-06-30Recruiting
A 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
Advanced Neuroimaging Evaluation of the Central Nervous System Biological Changes Associated With Efavirenz Therapy and Switch to an Elvitegravir-based Regimen [NCT01929759]10 participants (Actual)Interventional2014-01-31Completed
Pharmacokinetic Properties of Antiretroviral and Anti-Tuberculosis Drugs During Pregnancy and Postpartum [NCT04518228]325 participants (Anticipated)Observational2021-06-08Recruiting
Elvitegravir (EVG) Cerebrospinal Fluid (CSF) Pharmacokinetics in HIV-Infected Individuals [NCT02251236]14 participants (Actual)Interventional2016-01-31Completed
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
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 III Multicenter, Open-Label, Randomized Study to Evaluate a Switch to MK-1439A in HIV-1-Infected Subjects Virologically Suppressed on a Regimen of a Ritonavir-boosted Protease Inhibitor and Two Nucleoside Reverse Transcriptase Inhibitors (NRTIs) [NCT02397096]Phase 3673 participants (Actual)Interventional2015-06-09Completed
A Phase 2a, Multicenter, Randomized, Adaptive, Open-label, Dose Ranging Study to Evaluate the Antiviral Effect, Safety, Tolerability and Pharmacokinetics of Cobicistat-boosted GSK2838232 Monotherapy Over 10 Days in HIV-1 Infected Treatment-naive Adults [NCT03045861]Phase 233 participants (Actual)Interventional2017-03-17Completed
An Open-label, Two-part Study to Determine the Absolute Bioavailability (BA) of OZ439 Using Simultaneous Intravenous [14C]-OZ439 Microdose/800mg Oral Dosing and to Investigate the Pharmacokinetics (PK) of OZ439 Granules Administered as Single Doses Suspen [NCT04069221]Phase 126 participants (Actual)Interventional2017-02-28Completed
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
Pharmacokinetic Boosting of Olaparib to Improve Exposure, Tolerance and Cost-effectiveness [NCT05078671]Phase 4160 participants (Anticipated)Interventional2021-12-15Recruiting
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
Efficacy and Safety of Elvitegravir/Cobicistat/Emtricitabine/Tenofovir Alafenamide (Genvoya) as Maintenance Treatment of HIV-1/Hepatitis B Virus (HBV)-Coinfected Patients: an Observational Study [NCT03425994]275 participants (Actual)Observational [Patient Registry]2018-02-06Active, not recruiting
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
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
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
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))
Clinical ECG Study to Evaluate Electrophysiological Effects of Potential QT Prolonging Drugs With Novel ECG Biomarkers With Exposure-Response Analysis [NCT05716854]Phase 144 participants (Actual)Interventional2023-03-21Completed
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
An Open-Label, Randomized Comparison of Elvitegravir-Cobicistat-Tenofovir Alafenamide-Emtricitabine Versus Efavirenz-Tenofovir Disoproxil Fumarate-Lamivudine in Patients Starting Antiretroviral Therapy on the Day of HIV Diagnosis [NCT03405194]0 participants (Actual)Interventional2018-05-01Withdrawn(stopped due to Haiti gained access to dolutegravir as first-line ART, so we cancelled the study before any patient was enrolled (we didn't want SOC group to receive EFV))
A Phase 1, Open-label, Multicohort Study to Evaluate the Impact of Inhibitors and Inducers of Cytochrome P450 Enzyme (CYP)3A, P-glycoprotein (P-gp), and Breast Cancer Resistant Protein (BCRP) on the Pharmacokinetics (PK) of Vesatolimod (VES) in Virologica [NCT05458102]Phase 118 participants (Actual)Interventional2022-08-19Terminated(stopped due to Sponsor's decision to change the clinical development plan of this molecule. This decision is not based on efficacy or safety concerns.)
An Open Label Trial of STRIBILD™ (Elvitegravir, Cobicistat, Emtricitabine, Tenofovir Disoproxil Fumarate) for ARV-naïve HIV-2 Infected Adults in Dakar, Senegal [NCT02180438]Phase 430 participants (Actual)Interventional2014-09-30Completed
The Pharmacokinetics of Dolutegravir, Darunavir/Cobocistat When Co-administered in Healthy Volunteers [NCT03094507]Phase 121 participants (Actual)Interventional2017-04-19Completed
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
Effect of Antiretroviral Treatment Initiated During Acute HIV-1 Infection on Measures of HIV-1 Persistence and on HIV-1-Specific Immune Responses [NCT02859558]Phase 2195 participants (Actual)Interventional2017-01-31Active, not recruiting
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
A Randomized, 5-Period, Crossover Study in Healthy Subjects to Assess the Bioequivalence of Atazanavir When Co-Administered With Cobicistat as a Fixed Dose Combination Relative to the Single Agents Following a Light Meal, the Relative Bioavailability of A [NCT01837719]Phase 164 participants (Actual)Interventional2013-04-30Completed
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
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: 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: Area Under the Curve From 0 to 24 Hours (AUC24) With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Area Under the Curve From 0 to 24 Hours (AUC24) With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Maximum Concentration (Cmax) in mg/L With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Maximum Concentration (Cmax) in mg/L With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Maximum Concentration (Cmax) in mg/L With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Maximum Concentration (Cmax) in ng/mL With Median (95% CI) for ARVs and TB Drugs
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]PK Parameter: Maximum Concentration (Cmax) in ng/mL With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Trough Concentration (C12) With Geometric Mean (95% CI) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Trough Concentration (C12) With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: 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]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: 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]PK Parameter: Trough Concentration (C24) With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]Number of Women Who Met PK Target of Area Under the Curve (AUC) for ARVs
NCT00042289 (26) [back to overview]Number of Women Who Met PK Target of Area Under the Curve (AUC) for ARVs
NCT00042289 (26) [back to overview]Pharmacokinetic (PK) Parameter: Infant Plasma Washout Concentration of ARVs and TB Drugs
NCT00855335 (15) [back to overview]Predose (Trough) Plasma Concentration (C0h)
NCT00855335 (15) [back to overview]Time to Reach the Maximum Plasma Concentration (Tmax)
NCT00855335 (15) [back to overview]Number of Infants With Human Immunodeficiency Virus (HIV) Positive Test Result
NCT00855335 (15) [back to overview]Number of Participants With Resistance at Virological Failure
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]Area Under the Plasma Concentration-Time Curve From Time of Administration to 24 Hours Post-dose (AUC0-24h)
NCT00855335 (15) [back to overview]Maximum Plasma Concentration (Cmax)
NCT00855335 (15) [back to overview]Mean Change From Baseline in CD4+ Cell Count
NCT00855335 (15) [back to overview]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]Mean Change From Baseline in Log10 Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA) Viral Load Value
NCT00855335 (15) [back to overview]Minimum Plasma Concentration (Cmin)
NCT00855335 (15) [back to overview]Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)
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]Plasma Concentration of Drug in the Cord Plasma and Maternal Plasma Samples Collected at the Time of Delivery
NCT00892437 (6) [back to overview]Change From Baseline in CD4 Cell Count at Week 24
NCT00892437 (6) [back to overview]Change From Baseline in CD4 Cell Count at Week 48
NCT00892437 (6) [back to overview]Change From Baseline in HIV-1 RNA at Week 24
NCT00892437 (6) [back to overview]Change From Baseline in HIV-1 RNA at Week 48
NCT00892437 (6) [back to overview]Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 24
NCT00892437 (6) [back to overview]Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 48
NCT01108510 (8) [back to overview]Change From Baseline in CD4 Cell Count at Week 96
NCT01108510 (8) [back to overview]Change From Baseline in CD4 Cell Count at Week 144
NCT01108510 (8) [back to overview]Change From Baseline in CD4 Cell Count at Week 192
NCT01108510 (8) [back to overview]Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 144
NCT01108510 (8) [back to overview]Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 192
NCT01108510 (8) [back to overview]Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 48
NCT01108510 (8) [back to overview]Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 96
NCT01108510 (8) [back to overview]Change From Baseline in CD4 Cell Count at Week 48
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]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 (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 Achieving HIV-1 RNA < 50 Copies/mL at Week 48 (Snapshot Analysis)
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]Change From Baseline in CD4+ Cell Count at Week 48
NCT01440569 (7) [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 24
NCT01565850 (6) [back to overview]Change From Baseline in CD4+ Cell Count 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 HIV-1 RNA at Week 24
NCT01565850 (6) [back to overview]Change From Baseline in HIV-1 RNA at Week 48
NCT01565850 (6) [back to overview]Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 48
NCT01837719 (12) [back to overview]Maximum Observed Plasma Concentration (Cmax) of Cobicistat
NCT01837719 (12) [back to overview]Area Under the Concentration Curve From Time 0 to Time of Last Quantifiable Concentration (AUC[0-T]) and Area Under the Concentration Curve From Time 0 to Infinity (AUC[INF]) of Cobicistat
NCT01837719 (12) [back to overview]Time of Maximum Observed Concentration (Tmax) of Atazanavir
NCT01837719 (12) [back to overview]Number of Participants With Out-of-range Intervals on Electrocardiogram (ECG) Findings
NCT01837719 (12) [back to overview]T-HALF of Cobicistat
NCT01837719 (12) [back to overview]Observed Concentration at 24 Hours (C24) of Atazanavir
NCT01837719 (12) [back to overview]Number of Participants With Marked Abnormalities in Results of Clinical Laboratory Tests
NCT01837719 (12) [back to overview]Maximum Observed Plasma Concentration (Cmax) of Atazanavir
NCT01837719 (12) [back to overview]Time of Maximum Observed Concentration (Tmax) of Cobicistat
NCT01837719 (12) [back to overview]Apparent Terminal Half-life (T-HALF) of Atazanavir
NCT01837719 (12) [back to overview]Number of Participants Who Died and With Serious Adverse Events (SAEs)
NCT01837719 (12) [back to overview]Area Under the Plasma Concentration-time Curve (AUC) From Time 0 to Time of Last Quantifiable Concentration (AUC[0-T]) and From Time 0 to Infinity (AUC[INF]) for Atazanavir
NCT01929759 (8) [back to overview]Change in Neurometabolites Based on Magnetic Resonance Spectroscopy (MRS)
NCT01929759 (8) [back to overview]Sleep Quality
NCT01929759 (8) [back to overview]Neurocognitive Changes
NCT01929759 (8) [back to overview]Neural Activation Networks Using Functional Magnetic Resonance Imaging (fMRI)
NCT01929759 (8) [back to overview]Markers of Immune Activation
NCT01929759 (8) [back to overview]Fasting Lipid Profile
NCT01929759 (8) [back to overview]Effect of EFV and Its Metabolites
NCT01929759 (8) [back to overview]Change in Other Neurometabolite Measured by MRS Between Week 0 and Week 8
NCT02180438 (12) [back to overview]Grade 3 or 4 Adverse Events
NCT02180438 (12) [back to overview]Virologic Failure, FDA Snapshot (HIV-2 Plasma Viral Load >50 and >400 Copies/ml)
NCT02180438 (12) [back to overview]< 50 CD4 T-cell Increase at 48 Weeks From Baseline
NCT02180438 (12) [back to overview]CD4 T-cell Count at 48 Weeks < Baseline
NCT02180438 (12) [back to overview]Death
NCT02180438 (12) [back to overview]Development of Drug Resistance Mutations to Elvitegravir or Emtricitabine or Tenofovir DF
NCT02180438 (12) [back to overview]Interim 24 Weeks Analysis of Death
NCT02180438 (12) [back to overview]Interim Analysis at 24 Weeks of Grade 3 and 4 Adverse Events
NCT02180438 (12) [back to overview]Interim Analysis at 24 Weeks of HIV-2 Virologic Failure
NCT02180438 (12) [back to overview]Interim Analysis at 24 Weeks of New WHO Stage 3 or 4 Event
NCT02180438 (12) [back to overview]New WHO Stage 3 or 4 Event
NCT02180438 (12) [back to overview]Switching Off Stribild Prior to 48 Weeks
NCT02251236 (4) [back to overview]Concentration of Tenofovir in Cerebrospinal Fluid at Week 24
NCT02251236 (4) [back to overview]Concentration of Elvitegravir in Cerebrospinal Fluid at Week 24
NCT02251236 (4) [back to overview]Concentration of Tenofovir in Cerebrospinal Fluid at Baseline
NCT02251236 (4) [back to overview]Concentration of Elvitegravir in Cerebrospinal Fluid at Baseline
NCT02397096 (12) [back to overview]Mean Change From Baseline in Cluster of Differentiation (CD4) Cell Counts
NCT02397096 (12) [back to overview]Percentage of Participants Maintaining HIV-1 RNA <40 Copies/mL
NCT02397096 (12) [back to overview]Mean Change From Baseline in Fasting Low-density Lipoprotein Cholesterol (LDL-C)
NCT02397096 (12) [back to overview]Mean Change From Baseline in Fasting Non-high-density Lipoprotein Cholesterol (Non-HDL-C)
NCT02397096 (12) [back to overview]Percentage of Participants Maintaining HIV-1 RNA <50 Copies/mL
NCT02397096 (12) [back to overview]Percentage of Participants Maintaining Human Immunodeficiency Virus-1 Ribonucleic Acid (HIV-1 RNA) <50 Copies/mL
NCT02397096 (12) [back to overview]Percentage of Participants With HIV-1 RNA >=50 Copies/mL
NCT02397096 (12) [back to overview]Mean Change From Baseline in Cluster of Differentiation (CD4) Cell Counts
NCT02397096 (12) [back to overview]Percentage of Participants Discontinuing From Study Medication Due to an AE(s)
NCT02397096 (12) [back to overview]Percentage of Participants Experiencing ≥1 Adverse Event (AE)
NCT02397096 (12) [back to overview]Percentage of Participants Experiencing ≥1 Serious Adverse Event (SAE)
NCT02397096 (12) [back to overview]Percentage of Participants Maintaining HIV-1 RNA <40 Copies/mL
NCT02431247 (64) [back to overview]Percentage of Participants With >95% Treatment Adherence Assessed by Drug Accountability
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]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 ALP Levels
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]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]Percent Change From Baseline in Urine Fractional Excretion of Phosphate (FEPO4) at Week 48
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]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]Plasma Concentrations 2 Hours After Dosing (C0-2h) of Tenofovir Alafenamide
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
NCT02431247 (64) [back to overview]Percentage of Participants With >95% Treatment Adherence Assessed by Drug Accountability
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 Alkaline Phosphatase (ALP) Levels at Weeks 24 and 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 Baseline in Urine Protein to Creatinine Ratio (UPCR) 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 Albumin to Creatinine Ratio (UACR) at Week 48
NCT02431247 (64) [back to overview]Change From Baseline in Serum Creatinine 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 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 Estimated Glomerular Filtration Rate Based on Serum Creatinine by (Cockcroft-Gault Formula) 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 Cluster of Differentiation-4 (CD4+) Cell Count at Week 48
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 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]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]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 Baseline in Hip and Spine Bone Mineral Density (BMD)
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 Grade 3 and 4 AEs, SAEs, and Premature Discontinuations Due to Adverse Events Post-Week 96 to End of Extension
NCT02625207 (24) [back to overview]Part 1: Average Plasma Concentration (Cavg) 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 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 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 1: Time to Reach Maximum Observed Plasma Concentration (Tmax) of Maraviroc
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: Average Plasma Concentration (Cavg) of Maraviroc
NCT02625207 (24) [back to overview]Part 2: Maximum Observed Plasma Concentration (Cmax) of Maraviroc
NCT02625207 (24) [back to overview]Part 2: Number of Participants With 12-Lead Electrocardiogram (ECG) 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 Laboratory Abnormalities
NCT02625207 (24) [back to overview]Part 2: Plasma Concentration of Maraviroc at 24 Hours Post-dose
NCT02625207 (24) [back to overview]Part 2: Time to Reach Maximum Observed Plasma Concentration (Tmax) 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 1: Average Plasma Concentration (Cav) of Metabolites of Maraviroc
NCT02625207 (24) [back to overview]Part 1: Maximum Observed Plasma Concentration (Cmax) of Metabolites of Maraviroc
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: Number of Participants With Treatment Emergent Adverse Events (AEs) and Serious Adverse Events (SAEs)
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: Time to Reach Maximum Observed Plasma Concentration (Tmax) of Metabolites of Maraviroc
NCT02625207 (24) [back to overview]Part 2: Number of Participants With Treatment Emergent Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT02859558 (4) [back to overview]Proportion of Participants With Undetectable Cell-associated HIV-1 DNA (CAHD) Prior to ART Initiation
NCT02859558 (4) [back to overview]HIV-1-specific CD8+ and T-cell Responses to Nef, Gag, Pol and Env by Flow Cytometry
NCT02859558 (4) [back to overview]HIV-1-specific CD4+ and T-cell Responses to Nef, Gag, Pol and Env by Flow Cytometry
NCT02859558 (4) [back to overview]Proportion of Participants With Undetectable Cell-associated HIV-1 DNA (CAHD)
NCT03045861 (33) [back to overview]Concentration at End of Dosing Interval (Ctau) for GSK2838232 on Day 10
NCT03045861 (33) [back to overview]Number of Participants With Abnormal Urine Parameters
NCT03045861 (33) [back to overview]Number of Participants Who Were Administered Concomitant Medications
NCT03045861 (33) [back to overview]Maximum Observed Concentration (Cmax) for GSK2838232 on Day 1
NCT03045861 (33) [back to overview]Maximum Decline From Baseline in Plasma HIV-1 Ribonucleic Acid (RNA)
NCT03045861 (33) [back to overview]Concentration of GSK2838232 at 24 Hours Post-dose on Day 1
NCT03045861 (33) [back to overview]Change From Baseline to Day 11 in CD4+ Count Relative to Day 10 AUC (0 to Tau)
NCT03045861 (33) [back to overview]Change From Baseline to Day 11 in CD4+ Count Relative to Day 10 Cmax
NCT03045861 (33) [back to overview]Change From Baseline to Day 11 in CD4+ Count Relative to Day 10 Ctau
NCT03045861 (33) [back to overview]Number of Participants With Liver Function Laboratory Abnormalities of Potential Clinical Importance
NCT03045861 (33) [back to overview]Change From Baseline to Day 11 in log10 Plasma HIV-1 RNA Relative to Day 10 AUC (0 to Tau)
NCT03045861 (33) [back to overview]Change From Baseline to Day 11 in log10 Plasma HIV-1 RNA Relative to Day 10 Cmax
NCT03045861 (33) [back to overview]Change From Baseline to Day 11 in log10 Plasma HIV-1 RNA Relative to Day 10 Ctau
NCT03045861 (33) [back to overview]Cmax for GSK2838232 on Day 10
NCT03045861 (33) [back to overview]Change From Baseline in Cluster of Differentiation 4+ (CD4+) Cell Count to Day 11
NCT03045861 (33) [back to overview]Absorption Lag Time (Tlag) for GSK2838232 on Day 1
NCT03045861 (33) [back to overview]Time to Maximum Observed Concentration (Tmax) for GSK2838232 on Day 1
NCT03045861 (33) [back to overview]Terminal Elimination Half-life (T1/2) of GSK2838232 Following Administration on Day 10
NCT03045861 (33) [back to overview]Steady State Assessment of Plasma Pre-dose Concentrations by Treatment
NCT03045861 (33) [back to overview]Apparent Oral Clearance of GSK2838232 Following Administration on Day 10
NCT03045861 (33) [back to overview]Area Under the Concentration-time Curve Over the Dosing Interval (AUC [0 to Tau]) for GSK2838232 on Day 10
NCT03045861 (33) [back to overview]Area Under the Plasma Concentration Time Curve From Zero (Pre-dose) to 24 Hours (AUC[0 to 24]) for GSK2838232 on Day 1
NCT03045861 (33) [back to overview]Pre-morning Dose Concentrations (C0) on Day 2 Through 11
NCT03045861 (33) [back to overview]Number of Participants With Vital Signs Data Outside Clinical Concern Range
NCT03045861 (33) [back to overview]Pre-dose Concentration (C0) of GSK2838232 on Day 10
NCT03045861 (33) [back to overview]Number of Participants With Hematology Parameter Abnormalities of Potential Clinical Importance
NCT03045861 (33) [back to overview]Number of Participants With Emergent Drug Resistance Mutations
NCT03045861 (33) [back to overview]Number of Participants With Clinical Chemistry Abnormalities of Potential Clinical Importance
NCT03045861 (33) [back to overview]Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT03045861 (33) [back to overview]Number of Participants With Abnormal Electrocardiogram (ECG) Findings
NCT03045861 (33) [back to overview]Dose Proportionality of GSK2838232
NCT03045861 (33) [back to overview]Accumulation Ratio for GSK2838232
NCT03045861 (33) [back to overview]Tmax for GSK2838232 Following Administration on Day 10
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]Concentration at Last Quantifiable Time Point (Clast)
NCT03123848 (10) [back to overview]Maximum Observed Plasma Concentration (Cmax)
NCT03123848 (10) [back to overview]Number of Participants With Adverse Events (AEs)
NCT03123848 (10) [back to overview]Terminal Elimination Half-Life (t1/2)
NCT03123848 (10) [back to overview]Time to Reach the Maximum Plasma Concentration (Tmax)
NCT03123848 (10) [back to overview]Elimination Rate Constant (Lambda[z])
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]Apparent Total Body Clearance of Drug at the Terminal Phase After Extravascular Administration (CL/F)
NCT03123848 (10) [back to overview]Apparent Volume of Distribution (Vz/F)
NCT03717129 (9) [back to overview]AUC0-∞ for Tenofovir (TFV)
NCT03717129 (9) [back to overview]EVG Cmax
NCT03717129 (9) [back to overview]FTC Half-life
NCT03717129 (9) [back to overview]EVG Half-life
NCT03717129 (9) [back to overview]Area Under the Curve From 0 to Infinity (AUC0-∞) for EVG
NCT03717129 (9) [back to overview]FTC Cmax
NCT03717129 (9) [back to overview]TFV Cmax
NCT03717129 (9) [back to overview]TFV Half-life
NCT03717129 (9) [back to overview]AUC0-∞ for FTC
NCT03864406 (8) [back to overview]Apparent Volume of Distribution (V/F) for Rivaroxaban
NCT03864406 (8) [back to overview]Apparent Oral Clearance (CL/F) for Rivaroxaban
NCT03864406 (8) [back to overview]Area Under the Concentration Versus Time Curve (AUC0-∞) for Rivaroxaban
NCT03864406 (8) [back to overview]Area Under the Concentration Versus Time Curve (AUC0-24hr) 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
NCT03864406 (8) [back to overview]Terminal Elimination Half-life (t½) for Rivaroxaban
NCT03864406 (8) [back to overview]Time to Maximum Plasma Concentration (Tmax) for Rivaroxaban
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Emtricitabine-triphosphate (FTC-TP)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Emtricitabine-triphosphate (FTC-TP)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Emtricitabine-triphosphate (FTC-TP)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Emtricitabine (FTC)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Emtricitabine (FTC)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Emtricitabine (FTC)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Elvitegravir (EVG)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Elvitegravir (EVG)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Elvitegravir (EVG)
NCT03976752 (40) [back to overview]Peripheral Blood Mononuclear Cell (PBMC) Concentration of Emtricitabine-triphosphate (FTC-TP)
NCT03976752 (40) [back to overview]Peripheral Blood Mononuclear Cell (PBMC) Concentration of Emtricitabine-triphosphate (FTC-TP)
NCT03976752 (40) [back to overview]Peripheral Blood Mononuclear Cell (PBMC) Concentration of Emtricitabine-triphosphate (FTC-TP)
NCT03976752 (40) [back to overview]Peripheral Blood Mononuclear Cell (PBMC) Concentration of Tenofovir-diphosphate (TFV-DP)
NCT03976752 (40) [back to overview]Peripheral Blood Mononuclear Cell (PBMC) Concentration of Tenofovir-diphosphate (TFV-DP)
NCT03976752 (40) [back to overview]Peripheral Blood Mononuclear Cell (PBMC) Concentration of Tenofovir-diphosphate (TFV-DP)
NCT03976752 (40) [back to overview]Peripheral Blood Mononuclear Cell (PBMC) Concentration of Tenofovir-diphosphate (TFV-DP)
NCT03976752 (40) [back to overview]Plasma Concentration of Elvitegravir (EVG)
NCT03976752 (40) [back to overview]Plasma Concentration of Elvitegravir (EVG)
NCT03976752 (40) [back to overview]Plasma Concentration of Elvitegravir (EVG)
NCT03976752 (40) [back to overview]Plasma Concentration of Elvitegravir (EVG)
NCT03976752 (40) [back to overview]Plasma Concentration of Emtricitabine (FTC)
NCT03976752 (40) [back to overview]Plasma Concentration of Emtricitabine (FTC)
NCT03976752 (40) [back to overview]Plasma Concentration of Emtricitabine (FTC)
NCT03976752 (40) [back to overview]Plasma Concentration of Emtricitabine (FTC)
NCT03976752 (40) [back to overview]Plasma Concentration of Tenofovir (TFV)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Tenofovir-diphosphate (TFV-DP)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Emtricitabine (FTC)
NCT03976752 (40) [back to overview]Plasma Concentration of Tenofovir (TFV)
NCT03976752 (40) [back to overview]Plasma Concentration of Tenofovir (TFV)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Tenofovir-diphosphate (TFV-DP)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Elvitegravir (EVG)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Tenofovir-diphosphate (TFV-DP)
NCT03976752 (40) [back to overview]Plasma Concentration of Tenofovir (TFV)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Tenofovir-diphosphate (TFV-DP)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Tenofovir (TFN)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Tenofovir (TFN)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Tenofovir (TFN)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Tenofovir (TFN)
NCT03976752 (40) [back to overview]Rectal Tissue Concentration of Emtricitabine-triphosphate (FTC-TP)
NCT03976752 (40) [back to overview]Peripheral Blood Mononuclear Cell (PBMC) Concentration of Emtricitabine-triphosphate (FTC-TP)
NCT04069221 (5) [back to overview]OZ439 Fpo
NCT04069221 (5) [back to overview]OZ439 AUC0-168h
NCT04069221 (5) [back to overview]OZ439 AUC0-inf
NCT04069221 (5) [back to overview]OZ439 C168h
NCT04069221 (5) [back to overview]OZ439 Cmax

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

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

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

[back to top]

PK Parameter: Area Under the Curve From 0 to 24 Hours (AUC24) With Median (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: Maximum Concentration (Cmax) in mg/L With Median (IQR) for ARVs and TB Drugs

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

PK Parameter: Maximum Concentration (Cmax) in ng/mL With Median (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]

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]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

PK Parameter: 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 (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]

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: 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]

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

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

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

[back to top]

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

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

,
InterventionParticipants (Count of Participants)
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]

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]

Predose (Trough) Plasma Concentration (C0h)

C0h is defined as the predose (trough) plasma concentration or concentration just prior to study drug administration. (NCT00855335)
Timeframe: Predose on Weeks 24-28 (Visit 4, 2nd trimester), 34-38 (visit 5, 3rd trimester) and 6-12 weeks postpartum (visit 8)

,,,,,,,
Interventionnanogram per milliliter (ng/mL) (Mean)
Postpartum (6-12 W)2nd Trimester3rd Trimester
Cobicistat 150 mg (Darunavir 800/Cobicistat 150 mg) Once Daily134NA30.1
Darunavir 600 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily360823233280
Darunavir 800 mg (Darunavir 800/Cobicistat 150 mg) Once Daily2811540824
Darunavir 800 mg (Darunavir 800/Ritonavir 100 mg) Once Daily248117931528
Etravirine 200 mg Twice Daily281439413
Rilpivirine 25 mg Once Daily12775.678.0
Ritonavir 100 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily491.4225.9236.0
Ritonavir 100 mg (Darunavir 800/Ritonavir 100 mg) Once Daily14794.274.6

[back to top]

Time to Reach the Maximum Plasma Concentration (Tmax)

The Tmax is defined as actual sampling time to reach maximum observed plasma concentration. (NCT00855335)
Timeframe: Between Predose and 24 hours postdose at Weeks 24-28 (Visit 4, 2nd trimester), 34-38 (visit 5, 3rd trimester) and 6-12 weeks postpartum (visit 8)

,,,,,,,
Interventionhour (h) (Median)
Postpartum (6-12 W)2nd Trimester3rd Trimester
Cobicistat 150 mg (Darunavir 800/Cobicistat 150 mg) Once Daily4.004.033.50
Darunavir 600 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily3.003.003.00
Darunavir 800 mg (Darunavir 800/Cobicistat 150 mg) Once Daily4.004.003.50
Darunavir 800 mg (Darunavir 800/Ritonavir 100 mg) Once Daily4.004.003.05
Etravirine 200 mg Twice Daily4.003.053.00
Rilpivirine 25 mg Once Daily4.004.004.00
Ritonavir 100 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily5.044.174.07
Ritonavir 100 mg (Darunavir 800/Ritonavir 100 mg) Once Daily4.185.926.00

[back to top]

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]

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]

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]

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

The AUC (0-24) is the area under the plasma concentration-time curve from time zero to 24 hours post dose. The selected arms were based on the dosing frequency (once daily). (NCT00855335)
Timeframe: Between Predose and 24 hours postdose at Weeks 24-28 (Visit 4, 2nd trimester), 34-38 (visit 5, 3rd trimester) and 6-12 weeks postpartum (visit 8)

,,,,
Interventionnanogram*hour per milliliter (ng*h/mL) (Mean)
Postpartum (6-12 W)2nd trimester3rd trimester
Cobicistat 150 mg (Darunavir 800/Cobicistat 150 mg) Once Daily864338624736
Darunavir 800 mg (Darunavir 800/Cobicistat 150 mg) Once Daily996134729347991
Darunavir 800 mg (Darunavir 800/Ritonavir 100 mg) Once Daily921166228961112
Rilpivirine 25 mg Once Daily271417921762
Ritonavir 100 mg (Darunavir 800/Ritonavir 100 mg) Once Daily658439353821

[back to top]

Maximum Plasma Concentration (Cmax)

The Cmax is the maximum observed plasma concentration. (NCT00855335)
Timeframe: Between Predose and 24 hours postdose at Weeks 24-28 (Visit 4, 2nd trimester), 34-38 (visit 5, 3rd trimester) and 6-12 weeks postpartum (visit 8)

,,,,,,,
Interventionnanogram per milliliter (ng/mL) (Mean)
Postpartum (6-12 W)2nd Trimester3rd Trimester
Cobicistat 150 mg (Darunavir 800/Cobicistat 150 mg) Once Daily996571759
Darunavir 600 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily665946685328
Darunavir 800 mg (Darunavir 800/Cobicistat 150 mg) Once Daily791843404910
Darunavir 800 mg (Darunavir 800/Ritonavir 100 mg) Once Daily731049645132
Etravirine 200 mg Twice Daily569774785
Rilpivirine 25 mg Once Daily167121123
Ritonavir 100 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily1110546.8536.1
Ritonavir 100 mg (Darunavir 800/Ritonavir 100 mg) Once Daily742439397

[back to top]

Mean Change From Baseline in CD4+ Cell Count

Mean Change From Baseline in CD4+ Cell Count were assessed for immunology testing. (NCT00855335)
Timeframe: Baseline, 4 weeks after baseline, 2nd and 3rd trimesters of pregnancy and postpartum (2-5 weeks and 6-12 weeks)

Intervention10^6 Cells/Liter (Mean)
Baseline2nd trimester3rd trimesterPostpartum (2-5 weeks)Postpartum (6-12 weeks)
Darunavir 800 mg/Cobicistat 150 mg Once Daily594.1713.2972.17163244.67

[back to top]

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]

Mean Change From Baseline in Log10 Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA) Viral Load Value

Mean change from baseline in log 10 HIV-1 RNA VL was assessed up to postpartum (6-12 weeks). (NCT00855335)
Timeframe: Baseline, 4 weeks after baseline, 2nd and 3rd trimesters of pregnancy and postpartum (2-5 weeks and 6-12 weeks)

InterventionLog 10 copies per milliliter (copies/mL) (Mean)
Baseline2nd trimester3rd trimesterPostpartum (2-5 weeks)Postpartum (6-12 weeks)
Darunavir 800 mg/Cobicistat 150 mg Once Daily1.770.10.210.180.23

[back to top]

Mean Change From Baseline in Log10 Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA) Viral Load Value

Mean change from baseline in log 10 HIV-1 RNA VL was assessed up to postpartum (6-12 weeks). (NCT00855335)
Timeframe: Baseline, 4 weeks after baseline, 2nd and 3rd trimesters of pregnancy and postpartum (2-5 weeks and 6-12 weeks)

,,,
InterventionLog 10 copies per milliliter (copies/mL) (Mean)
Baseline4 Weeks after Baseline2nd trimester3rd trimesterPostpartum (2-5 weeks)Postpartum (6-12 weeks)
Darunavir 600 mg /Ritonavir 100 Twice Daily2.12-0.26-0.19-0.31-0.180.09
Darunavir 800 mg /Ritonavir 100 mg Once Daily1.88-0.27-0.16-0.23-0.040.11
Etravirine 200 mg Twice Daily2.060.180.160.170.130.05
Rilpivirine 25 mg Once Daily1.840.200.160.250.200.08

[back to top]

Minimum Plasma Concentration (Cmin)

The Cmin is the minimum observed plasma concentration. (NCT00855335)
Timeframe: Between Predose and 24 hours postdose at Weeks 24-28 (Visit 4, 2nd trimester), 34-38 (visit 5, 3rd trimester) and 6-12 weeks postpartum (visit 8)

,,,,,,,
Interventionnanogram per milliliter (ng/mL) (Mean)
Postpartum (6-12 W)2nd Trimester3rd Trimester
Cobicistat 150 mg (Darunavir 800/Cobicistat 150 mg) Once Daily41.4NANA
Darunavir 600 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily285119222661
Darunavir 800 mg (Darunavir 800/Cobicistat 150 mg) Once Daily1538168184
Darunavir 800 mg (Darunavir 800/Ritonavir 100 mg) Once Daily147312481075
Etravirine 200 mg Twice Daily269383349
Rilpivirine 25 mg Once Daily84.054.352.9
Ritonavir 100 mg (Darunavir 600/Ritonavir 100 mg) Twice Daily264.7141.1148.1
Ritonavir 100 mg (Darunavir 800/Ritonavir 100 mg) Once Daily40.532.228.0

[back to top]

Number of Participants With 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]

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]

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]

Change From Baseline in CD4 Cell Count at Week 24

The change from baseline in CD4 cell count at Week 24 was analyzed. (NCT00892437)
Timeframe: Baseline to Week 24

Interventioncells/μL (Mean)
ATV+COBI+FTC/TDF200
ATV+RTV+FTC/TDF202

[back to top]

Change From Baseline in CD4 Cell Count at Week 48

The change from baseline in CD4 cell count at Week 48 was analyzed. (NCT00892437)
Timeframe: Baseline to Week 48

Interventioncells/μL (Mean)
ATV+COBI+FTC/TDF243
ATV+RTV+FTC/TDF213

[back to top]

Change From Baseline in HIV-1 RNA at Week 24

The change from baseline in log_10 HIV-1 RNA at Week 24 was analyzed. (NCT00892437)
Timeframe: Baseline to Week 24

Interventionlog_10 copies/mL (Mean)
ATV+COBI+FTC/TDF-2.80
ATV+RTV+FTC/TDF-2.97

[back to top]

Change From Baseline in HIV-1 RNA at Week 48

The change from baseline in log_10 HIV-1 RNA at Week 48 was analyzed. (NCT00892437)
Timeframe: Baseline to Week 48

Interventionlog_10 copies/mL (Mean)
ATV+COBI+FTC/TDF-2.79
ATV+RTV+FTC/TDF-2.96

[back to top]

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

The percentage of participants with HIV-1 RNA < 50 copies/mL at Week 24 was analyzed using the missing = failure method, where participants with missing data were considered to have failed to achieve the endpoint. (NCT00892437)
Timeframe: Week 24

Interventionpercentage of participants (Number)
ATV+COBI+FTC/TDF84.0
ATV+RTV+FTC/TDF89.7

[back to top]

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

The percentage of participants with HIV-1 RNA < 50 copies/mL at Week 48 was analyzed using the missing = failure method. (NCT00892437)
Timeframe: Week 48

Interventionpercentage of participants (Number)
ATV+COBI+FTC/TDF82.0
ATV+RTV+FTC/TDF89.7

[back to top]

Change From Baseline in CD4 Cell Count at Week 96

(NCT01108510)
Timeframe: Baseline to Week 96

Interventioncells/μL (Mean)
ATV+COBI+FTC/TDF277
ATV+RTV+FTC/TDF287

[back to top]

Change From Baseline in CD4 Cell Count at Week 144

(NCT01108510)
Timeframe: Baseline to Week 144

Interventioncells/μL (Mean)
ATV+COBI+FTC/TDF310
ATV+RTV+FTC/TDF332

[back to top]

Change From Baseline in CD4 Cell Count at Week 192

(NCT01108510)
Timeframe: Baseline to Week 192

Interventioncells/μL (Mean)
ATV+COBI+FTC/TDF350
ATV+RTV+FTC/TDF343

[back to top]

Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 144

The percentage of participants with HIV-1 RNA < 50 copies/mL at Week 144 was analyzed using the snapshot algorithm. (NCT01108510)
Timeframe: Week 144

Interventionpercentage of participants (Number)
ATV+COBI+FTC/TDF72.1
ATV+RTV+FTC/TDF74.1

[back to top]

Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 192

The percentage of participants with HIV-1 RNA < 50 copies/mL at Week 192 was analyzed using the snapshot algorithm. (NCT01108510)
Timeframe: Week 192

Interventionpercentage of participants (Number)
ATV+COBI+FTC/TDF71.6
ATV+RTV+FTC/TDF79.7

[back to top]

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

The percentage of participants with HIV-1 RNA < 50 copies/mL at Week 48 was analyzed using the snapshot algorithm, which defines a participant's virologic response status using only the viral load at the prespecified time point within an allowed window of time, along with study drug discontinuation status. (NCT01108510)
Timeframe: Week 48

Interventionpercentage of participants (Number)
ATV+COBI+FTC/TDF85.2
ATV+RTV+FTC/TDF87.4

[back to top]

Percentage of Participants With HIV-1 RNA < 50 Copies/mL at Week 96

The percentage of participants with HIV-1 RNA < 50 copies/mL at Week 96 was analyzed using the snapshot algorithm. (NCT01108510)
Timeframe: Week 96

Interventionpercentage of participants (Number)
ATV+COBI+FTC/TDF77.9
ATV+RTV+FTC/TDF79.3

[back to top]

Change From Baseline in CD4 Cell Count at Week 48

(NCT01108510)
Timeframe: Baseline to Week 48

Interventioncells/μL (Mean)
ATV+COBI+FTC/TDF213
ATV+RTV+FTC/TDF219

[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]

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]

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 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 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]

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]

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]

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]

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]

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 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 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]

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]

Maximum Observed Plasma Concentration (Cmax) of Cobicistat

Blood samples for testing plasma concentrations were obtained at predose and at specific timepoints up to 48 hours after dosing on Days 1, 8, 15, 22, and 29. Cmax was derived from plasma concentration versus time data. (NCT01837719)
Timeframe: Days 1, 8, 15, 22, and 29 (1,2, 2.5, 3, 4, 5, 6, 8, 12, and 16 hours postdose); Days 2, 9, 16, 23, and 30 (24 hours postdose)

Interventionng/mL (Geometric Mean)
Treatment A: Atazanavir + Cobicistat Coadministered1321
Treatment B: Atazanavir/Cobicistat FDC1348
Treatment C: Atazanavir + Cobicistat Coadministered952
Treatment D: Atazanavir/Cobicistat FDC1033
Treatment E: Atazanavir/Cobicistat FDC1060

[back to top]

Area Under the Concentration Curve From Time 0 to Time of Last Quantifiable Concentration (AUC[0-T]) and Area Under the Concentration Curve From Time 0 to Infinity (AUC[INF]) of Cobicistat

Blood samples for testing plasma concentrations were obtained at predose and at specific timepoints up to 48 hours after dosing on Days 1, 8, 15, 22, and 29. AUC(0-T) and AUC(INF) were derived from plasma concentration versus time data. (NCT01837719)
Timeframe: Days 1, 8, 15, 22, and 29 (1,2, 2.5, 3, 4, 5, 6, 8, 12, and 16 hours postdose); Days 2, 9, 16, 23, and 30 (24 hours postdose)

,,,,
Interventionng*h/mL (Geometric Mean)
AUC(0-T)AUC(INF) (n=63, 62, 63, 63, 28)
Treatment A: Atazanavir + Cobicistat Coadministered87389045
Treatment B: Atazanavir/Cobicistat FDC88669178
Treatment C: Atazanavir + Cobicistat Coadministered65417884
Treatment D: Atazanavir/Cobicistat FDC72047408
Treatment E: Atazanavir/Cobicistat FDC79168298

[back to top]

Time of Maximum Observed Concentration (Tmax) of Atazanavir

Blood samples for plasma concentrations were obtained at predose and at specific timepoints up to 48 hours after dosing on Days 1, 8, 15, 22, and 29. Tmax was derived from plasma concentration versus time data. (NCT01837719)
Timeframe: Days 1, 8, 15, 22, and 29 (predose and at 1, 2, 2.5, 3, 4, 5, 6, 8, 12, and 16 hours postdose); Days 2, 9, 16, 23, and 30 (24, 30, and 36 hours postdose); Days 3, 10, 17, 24, and 31 (48 hours postdose)

InterventionHours (Median)
Treatment A: Atazanavir + Cobicistat Coadministered3.00
Treatment B: Atazanavir/Cobicistat FDC2.50
Treatment C: Atazanavir + Cobicistat Coadministered2.00
Treatment D: Atazanavir/Cobicistat FDC2.00
Treatment E: Atazanavir/Cobicistat FDC3.54

[back to top]

Number of Participants With Out-of-range Intervals on Electrocardiogram (ECG) Findings

A 12-lead ECG was recorded at predose and 4 hours post dose at screening, Days -1, 1, 8 15, 22, 29 and study discharge. ECGs were recorded after the patient had been supine for at least 5 minutes. All ECG readings post dosing (including unscheduled) were included. (NCT01837719)
Timeframe: At screening; on Day -1; predose and 4 hours postdose on Days 1, 18, 15, 22, and 29; and at study discharge (Day 31)

,,,,
InterventionParticipants (Number)
PR interval >210 msecQRS interval >120 msecQT interval >500 msecQTcF>450 msec
Treatment A: Atazanavir + Cobicistat Coadministered0101
Treatment B: Atazanavir/Cobicistat FDC0100
Treatment C: Atazanavir + Cobicistat Coadministered0000
Treatment D: Atazanavir/Cobicistat FDC1000
Treatment E: Atazanavir/Cobicistat FDC0000

[back to top]

T-HALF of Cobicistat

Blood samples for testing plasma concentrations were obtained at predose and at specific timepoints up to 48 hours after dosing on Days 1, 8, 15, 22, and 29. T-HALF was derived from plasma concentration versus time data. (NCT01837719)
Timeframe: Days 1, 8, 15, 22, and 29 (1,2, 2.5, 3, 4, 5, 6, 8, 12, and 16 hours postdose); Days 2, 9, 16, 23, and 30 (24 hours postdose)

InterventionHours (Mean)
Treatment A: Atazanavir + Cobicistat Coadministered4.34
Treatment B: Atazanavir/Cobicistat FDC4.33
Treatment C: Atazanavir + Cobicistat Coadministered4.23
Treatment D: Atazanavir/Cobicistat FDC4.09
Treatment E: Atazanavir/Cobicistat FDC4.27

[back to top]

Observed Concentration at 24 Hours (C24) of Atazanavir

Blood samples for plasma concentrations were obtained at predose and at specific timepoints up to 48 hours after dosing on Days 1, 8, 15, 22, and 29. C24 was derived from plasma concentration versus time data. (NCT01837719)
Timeframe: Days 1, 8, 15, 22, and 29 (predose and at 1, 2, 2.5, 3, 4, 5, 6, 8, 12, and 16 hours postdose); Days 2, 9, 16, 23, and 30 (24, 30, and 36 hours postdose); Days 3, 10, 17, 24, and 31 (48 hours postdose)

Interventionng/mL (Geometric Mean)
Treatment A: Atazanavir + Cobicistat Coadministered416
Treatment B: Atazanavir/Cobicistat FDC449
Treatment C: Atazanavir + Cobicistat Coadministered295
Treatment D: Atazanavir/Cobicistat FDC337
Treatment E: Atazanavir/Cobicistat FDC398

[back to top]

Number of Participants With Marked Abnormalities in Results of Clinical Laboratory Tests

LLN=lower limit of normal; ULN=upper limit of normal; preRx=pretreatment; h=high; hpf=high power field. Abnormal criteria: Leukocytes, low (*10^3 c/uL): <0.85*preRx if preRxULN. Neutrophils, low (*10^3 c/uL): <0.85*preRx if preRx<1.5; <1.5 if preRx=missing; <1.5 if preRx ≥1.5. Bilirubin, h (mg/dL): >1.1* ULN if preRx≤ULN; >1.1*ULN if preRx=missing; >1.25*preRx if preRx>ULN. Bilirubin, h (mg/dL): >1.1* ULN if preRx≤ULN; >1.1*ULN if preRx=missing; >1.25*preRx if preRx>ULN. Blood, urine, h: ≥2*preRx if preRx≥1; ≥2 if preRx <1; ≥2 if preRx=missing. RBCs/WBCs, h (hpf): ≥2 if preRx=missing ≥2 if preRx<2 ≥4 if preRx ≥2. Creatine kinase, h (U/L): >1.5*preRx if preRx>ULN; >1.5*ULN if preRx≤ULN; >1.5*ULN if preRx=missing; AST, h (U/L): >1.25* preRx if preRx>ULN; >1.25*ULN if preRx≤ULN; >1.25*ULN if preRx=missing. Lactate dehydrogenase, h (U/L): >1.25*ULN if preRx≤ULN; >1.25*ULN if preRx=missing; >1.5*preRx if preRx>ULN. (NCT01837719)
Timeframe: At Screening and on Days -1,4, 11, 18, and 31 (study discharge)

,,,,
InterventionParticipants (Number)
Leukocytes (low)Neutrophils, absolute (low)Bilirubin, total (high)Bilirubin, directBlood, urine (high)Red blood cells (RBC), urine (high)White blood cells (WBC), urine (high)Creatine kinase (high)Aspartate aminotransferase (AST) (high)Lactate dehydrogenase (high)
Treatment A: Atazanavir + Cobicistat Coadministered3210112000
Treatment B: Atazanavir/Cobicistat FDC2400000100
Treatment C: Atazanavir + Cobicistat Coadministered3300000111
Treatment D: Atazanavir/Cobicistat FDC1231103000
Treatment E: Atazanavir/Cobicistat FDC1141112000

[back to top]

Maximum Observed Plasma Concentration (Cmax) of Atazanavir

Blood samples for plasma concentrations were obtained at predose and at specific timepoints up to 48 hours after dosing on Days 1, 8, 15, 22, and 29. Cmax was derived from plasma concentration versus time data. (NCT01837719)
Timeframe: Days 1, 8, 15, 22, and 29 (predose and at 1, 2, 2.5, 3, 4, 5, 6, 8, 12, and 16 hours postdose); Days 2, 9, 16, 23, and 30 (24, 30, and 36 hours postdose); Days 3, 10, 17, 24, and 31 (48 hours postdose)

Interventionng/mL (Geometric Mean)
Treatment A: Atazanavir + Cobicistat Coadministered3832
Treatment B: Atazanavir/Cobicistat FDC4104
Treatment C: Atazanavir + Cobicistat Coadministered2585
Treatment D: Atazanavir/Cobicistat FDC2941
Treatment E: Atazanavir/Cobicistat FDC2545

[back to top]

Time of Maximum Observed Concentration (Tmax) of Cobicistat

Blood samples for testing plasma concentrations were obtained at predose and at specific timepoints up to 48 hours after dosing on Days 1, 8, 15, 22, and 29. Tmax was derived from plasma concentration versus time data. (NCT01837719)
Timeframe: Days 1, 8, 15, 22, and 29 (1,2, 2.5, 3, 4, 5, 6, 8, 12, and 16 hours postdose); Days 2, 9, 16, 23, and 30 (24 hours postdose)

InterventionHours (Median)
Treatment A: Atazanavir + Cobicistat Coadministered2.52
Treatment B: Atazanavir/Cobicistat FDC2.52
Treatment C: Atazanavir + Cobicistat Coadministered2.00
Treatment D: Atazanavir/Cobicistat FDC2.00
Treatment E: Atazanavir/Cobicistat FDC4.00

[back to top]

Apparent Terminal Half-life (T-HALF) of Atazanavir

Blood samples for testing plasma concentrations were obtained at predose and at specific timepoints up to 48 hours after dosing on Days 1, 8, 15, 22, and 29. T-HALF was derived from plasma concentration versus time data. (NCT01837719)
Timeframe: Days 1, 8, 15, 22, and 29 (predose and at 1, 2, 2.5, 3, 4, 5, 6, 8, 12, and 16 hours postdose); Days 2, 9, 16, 23, and 30 (24, 30, and 36 hours postdose); Days 3, 10, 17, 24, and 31 (48 hours postdose)

InterventionHours (Mean)
Treatment A: Atazanavir + Cobicistat Coadministered7.54
Treatment B: Atazanavir/Cobicistat FDC7.50
Treatment C: Atazanavir + Cobicistat Coadministered7.25
Treatment D: Atazanavir/Cobicistat FDC7.21
Treatment E: Atazanavir/Cobicistat FDC7.14

[back to top]

Number of Participants Who Died and With Serious Adverse Events (SAEs)

An AE is defined as any new untoward medical occurrence or worsening of a preexisting medical condition in a clinical investigation participant who receives an investigational product and that does not necessarily have a causal relationship with this treatment. An AE can be any unfavorable and unintended sign (such as an abnormal laboratory finding), symptom, or disease temporally associated with the use of investigational product, whether or not considered related to the investigational product. An SAE is any untoward medical occurrence that at any dose results in death; is life-threatening; or requires or prolongs inpatient hospitalization. (NCT01837719)
Timeframe: On Day 24 or 31

,,,,
InterventionParticipants (Number)
DeathsSAEs
Treatment A: Atazanavir + Cobicistat Coadministered00
Treatment B: Atazanavir/Cobicistat FDC00
Treatment C: Atazanavir + Cobicistat Coadministered00
Treatment D: Atazanavir/Cobicistat FDC00
Treatment E: Atazanavir/Cobicistat FDC00

[back to top]

Area Under the Plasma Concentration-time Curve (AUC) From Time 0 to Time of Last Quantifiable Concentration (AUC[0-T]) and From Time 0 to Infinity (AUC[INF]) for Atazanavir

Blood samples for plasma concentrations were obtained at predose and at specific timepoints up to 48 hours after dosing on Days 1, 8, 15, 22, and 29. AUC(0-T) and AUC(INF) were derived from plasma concentration versus time data. (NCT01837719)
Timeframe: Days 1, 8, 15, 22, and 29 (predose and at 1, 2, 2.5, 3, 4, 5, 6, 8, 12, and 16 hours postdose); Days 2, 9, 16, 23, and 30 (24, 30, and 36 hours postdose); Days 3, 10, 17, 24, and 31 (48 hours postdose)

,,,,
Interventionng*h/mL (Geometric Mean)
AUC(0-T)AUC(0-INF)
Treatment A: Atazanavir + Cobicistat Coadministered3277533523
Treatment B: Atazanavir/Cobicistat FDC3490535673
Treatment C: Atazanavir + Cobicistat Coadministered2501725547
Treatment D: Atazanavir/Cobicistat FDC2787528378
Treatment E: Atazanavir/Cobicistat FDC2587326510

[back to top]

Change in Neurometabolites Based on Magnetic Resonance Spectroscopy (MRS)

Assess the change in levels of neuro-metabolites measured by MRS from week 0 (before switching to the efavirenz-based therapy) and then at week 8 (after completing 9 weeks of integrase-inhibitor based regimen with Stribild). Two areas of the brain: 1) posterior cingulate gyrus and 2) anterior cingulate will be assessed for the levels of brain Cr, GABA and GLU. (NCT01929759)
Timeframe: week 0 to week 8

Interventionarbitrary units (Mean)
Posterior Cingulate CreatinePosterior Cingulate GlutamatePosterior Cingulate GABAAnterior Cingulate CreatineAnterior Cingulate GlutamateAnterior Cingulate GABA
Drug Switching19.6124.546.7112.0816.163.67

[back to top]

Sleep Quality

Assess for changes in sleep pattern and quality prior to and after switching off EFV-based regimen through a self-administered Pittsburg Sleep Quality Index (PSQI). Measure consists of 19 items with each weighted on 0-3 scale and the sum produces a total score, which ranges from 0-21. The lower the score the healthier the sleep quality; minimum Score = 0 (better); maximum Score = 21 (worse). (NCT01929759)
Timeframe: week 0 and week 8

Interventionunits on a scale (Mean)
pre-switch PSQI indexpost-switch PSQI index
Drug Switching4.83.1

[back to top]

Neurocognitive Changes

"Assess for changes in cognitive and affective function prior to and after switching off EFV-based regimen. Indexes used to access neurocognitive changes included:~Wechsler Adult Intelligence Scale (WAIS-R) Digital Symbol Substitution Test: sensitive to brain damage, dementia, age and depressive changes. Range of 0-100, the higher the score the better the person's performance~Hamilton Rating Scale for Depression (HAMD): Measure of depression. Score of 0-7 is normal, score of >20 is moderate/severe depression~Depression Anxiety Stress Scale (DASS-21) the lower the score, the less severe depression, anxiety and stress. Scale range of 0-63~Frontal Systems Behavior Scale (FRSBE): Increased score indicates greater behavioral impairment associated with frontal systems, range 37.2 to 186~6. Spielberger state trait anxiety inventory (STAI): the higher the score the greater then anxiety level, range of 20 to 80." (NCT01929759)
Timeframe: week 0 and week 8

Interventionunits on a scale (Mean)
pre-switch WAISpost-switch WAISpre-switch FRSBEpost-switch FRSBEpre-switch HAMDpost-switch HAMDpre-switch DASS depressionpost-switch DASS depressionpre-swtich STAIpost-switch STAI
Drug Switching49.353.477.570.45.52.87.63.428.824.2

[back to top]

Neural Activation Networks Using Functional Magnetic Resonance Imaging (fMRI)

Assess changes in neural activation correlated with affective disturbances associated with efavirenz-based therapy using fMRI employing an Emotional Word/Go-NoGo task paradigm that probes affective symptomatologies typical with EFV use, specifically anxiety/dysphoria and affective dysregulation and their association with changes in cognitive function. Four brain regions of interests (ROIs) are specified to show the differential frontal-limbic activation patterns in the task-evoked neural responses to the 3 linear contrasts of Pre-switch / Post-switch / Pre- vs. Post-switch: [Negative Word vs. Neutral Word] x [No-Go Trial Block vs. Go Trial Block]: anterior Frontal Pole (aFP), posterior Cingulate Gyrus (pCG), dorsal anterior Cingulate Gyrus (daCG), Left Hippocampus (LHC). A linear mixed-effects model is utilized to examine the effect sizes of the key Regimen/Condition contrasts, with the Subject factor as the random-effect and Age incorporated as a co-variate of no interest. (NCT01929759)
Timeframe: week 0 and week 8

Interventionz-score (Number)
PreVsPostXNegVsNeuXNoGoVsGo: aFPPreVsPostXNegVsNeuXNoGoVsGo: pCGPreVsPostXNegVsNeuXNoGoVsGo: daCGPreVsPostXNegVsNeuXNoGoVsGo:LHCPre: NegVsNeuXNoGoVsGo: aFPPre: NegVsNeuXNoGoVsGo:pCGPre: NegVsNeuXNoGoVsGo: daCGPre: NegVsNeuXNoGoVsGo: LHCPost: NegVsNeuXNoGoVsGo: aFPPost: NegVsNeuXNoGoVsGo: pCGPost: NegVsNeuXNoGoVsGo: daCGPost: NegVsNeuXNoGoVsGo: LHC
Drug Switching3.423.90-2.76-2.963.393.61-3.15-2.27-3.17-2.663.693.47

[back to top]

Markers of Immune Activation

Change in markers of immune activation and inflammation associated with change to Stibild: sCD14, IP-10,sCD163, IL-6) (NCT01929759)
Timeframe: week 0 and week 8

Interventionpg/ML (Mean)
pre-switch sCD14post-switch sCD14pre-switch IP-10post-switch IP-10pre-switch sCD163post-switch sCD163pre-switch MCP-1post-switch MCP-1pre-switch IL-6post-switch IL-6pre-switch TNFR1post-switch TNFR1
Drug Switching33290002879000237.3224.2732000599500102.291.391.2551.301833.2878.1

[back to top]

Fasting Lipid Profile

Measure the change in fasting lipid panel prior to and after switching off EFV-based regimen. (NCT01929759)
Timeframe: 8 weeks

Interventionmg/dl (Mean)
pre-switch Total cholesterolpost-switch Total cholesterolpre-switch HDLpost-switch HDLpre-switch LDLpost-switch LDLpre-switch triglyceridepost-switch triglyceride
Drug Switching178.8168.552.652.3106.197.4101.394.5

[back to top]

Effect of EFV and Its Metabolites

Level of EFV (efavirenz) in Atripla and its two known metabolites known to cause cerebral side effects, 7-hydroxy (OH) EFV and 8-OH EFV, were measured in the plasma prior to switch off Atripla and after 8 weeks of RAL-based regimen (no EFV). (NCT01929759)
Timeframe: week 0 and week 8

Interventionparticipants (Number)
Number with detectable 7-OH and 8-OH (pre-switch)Number with detectable 7-OH and 8-OH (post-switch)
Drug Switching100

[back to top]

Change in Other Neurometabolite Measured by MRS Between Week 0 and Week 8

Use MRS to evaluate a fuller panel of known neurometabolites (in addition to the primary endpoints) between week 0 and week 8 to identify prominent and significant changes associated with EFV use. (NCT01929759)
Timeframe: week 0 to week 8

Interventionarbitrary units (Mean)
Posterior cingulate glutathionePosterior cingulate aspartateAnterior cingulate glutathioneAnterior cingulate aspartate
Drug Switching5.063.693.172.57

[back to top]

Grade 3 or 4 Adverse Events

Adverse event per NIH/DAIDS criteria (NCT02180438)
Timeframe: 48 weeks

InterventionParticipants (Count of Participants)
Lab: Grade 3 or 4Clinical: Grade 3 or 4
Open Label Prospective Single Arm Study of Stribild71

[back to top]

Virologic Failure, FDA Snapshot (HIV-2 Plasma Viral Load >50 and >400 Copies/ml)

(NCT02180438)
Timeframe: 48 weeks

InterventionParticipants (Count of Participants)
>50 copies/mL>400 copies/mL
Open Label Prospective Single Arm Study of Stribild10

[back to top]

< 50 CD4 T-cell Increase at 48 Weeks From Baseline

(NCT02180438)
Timeframe: 48 weeks

InterventionParticipants (Count of Participants)
Open Label Prospective Single Arm Study of Stribild2

[back to top]

CD4 T-cell Count at 48 Weeks < Baseline

(NCT02180438)
Timeframe: 48 weeks

InterventionParticipants (Count of Participants)
Open Label Prospective Single Arm Study of Stribild0

[back to top]

Death

Number of Participants Experiencing Death within the study period (NCT02180438)
Timeframe: 48 weeks

InterventionParticipants (Count of Participants)
Open Label Prospective Single Arm Study of Stribild0

[back to top]

Development of Drug Resistance Mutations to Elvitegravir or Emtricitabine or Tenofovir DF

(NCT02180438)
Timeframe: 48 weeks

InterventionParticipants (Count of Participants)
Open Label Prospective Single Arm Study of Stribild1

[back to top]

Interim 24 Weeks Analysis of Death

(NCT02180438)
Timeframe: 24 weeks

Interventionparticipants (Number)
Open Label Prospective Single Arm Study of Stribild0

[back to top]

Interim Analysis at 24 Weeks of Grade 3 and 4 Adverse Events

(NCT02180438)
Timeframe: 24 weeks

InterventionParticipants (Count of Participants)
Open Label Prospective Single Arm Study of Stribild0

[back to top]

Interim Analysis at 24 Weeks of HIV-2 Virologic Failure

Virologic failure, FDA Snapshot (HIV-2 plasma viral load >50 and >400 copies/ml) (NCT02180438)
Timeframe: 24 weeks

InterventionParticipants (Count of Participants)
Open Label Prospective Single Arm Study of Stribild0

[back to top]

Interim Analysis at 24 Weeks of New WHO Stage 3 or 4 Event

(NCT02180438)
Timeframe: 24 weeks

InterventionParticipants (Count of Participants)
Open Label Prospective Single Arm Study of Stribild0

[back to top]

New WHO Stage 3 or 4 Event

New AIDS defining event per WHO criteria (NCT02180438)
Timeframe: 48 weeks

InterventionParticipants (Count of Participants)
Open Label Prospective Single Arm Study of Stribild0

[back to top]

Switching Off Stribild Prior to 48 Weeks

(NCT02180438)
Timeframe: 48 Weeks

InterventionParticipants (Count of Participants)
Open Label Prospective Single Arm Study of Stribild0

[back to top]

Concentration of Tenofovir in Cerebrospinal Fluid at Week 24

(NCT02251236)
Timeframe: Week 24

Interventionng/mL (Median)
Stribild Arm0.507
Genvoya Arm0.481

[back to top]

Concentration of Elvitegravir in Cerebrospinal Fluid at Week 24

(NCT02251236)
Timeframe: Week 24

Interventionng/mL (Median)
Stribild Arm5.90
Genvoya Arm3.09

[back to top]

Concentration of Tenofovir in Cerebrospinal Fluid at Baseline

(NCT02251236)
Timeframe: Baseline

Interventionng/mL (Median)
Stribild Arm3.03
Genvoya Arm0.49

[back to top]

Concentration of Elvitegravir in Cerebrospinal Fluid at Baseline

(NCT02251236)
Timeframe: Baseline

Interventionng/mL (Median)
Stribild Arm4.3
Genvoya Arm2.72

[back to top]

Mean Change From Baseline in Cluster of Differentiation (CD4) Cell Counts

The mean change from baseline in CD4 cell counts was assessed using the Observed Failure (OF) approach. With the OF approach, baseline values were carried forward for participants who discontinued due to lack of efficacy. Cell counts were measured and expressed as cells/mm^3, and percent change was then calculated. CD4 cell counts were quantified by a central laboratory using a commercially available assay. (NCT02397096)
Timeframe: Immediate Switch to MK-1439A arm: Baseline and Week 48; Delayed Switch to MK-1439A arm: Baseline and Week 24

,
Interventioncells/mm^3 (Mean)
BaselineChange from Baseline
Delayed Switch Group (DSG)655.618.0
Immediate Switch Group (ISG)660.513.9

[back to top]

Percentage of Participants Maintaining HIV-1 RNA <40 Copies/mL

To evaluate the immunological effect of an immediate switch to MK -1439A on Study Day 1 compared with continuation of a ritonavir boosted, PI-based regimen, as measured by the proportion of subjects maintaining HIV-1 RNA below the limit of quantification (BLoQ) by the Abbott RealTime HIV-1 Assay (<40 copies/mL) in both treatment groups. (NCT02397096)
Timeframe: Immediate Switch to MK-1439A arm: Week 24; Delayed Switch to MK-1439A arm: Week 24

InterventionPercentage of Participants (Number)
Immediate Switch Group (ISG)92.8
Delayed Switch Group (DSG)93.3

[back to top]

Mean Change From Baseline in Fasting Low-density Lipoprotein Cholesterol (LDL-C)

To evaluate the effect on fasting LDL-C of an immediate switch to DOR/3TC/TDF on Study Day 1 compared with continuation of a ritonavir-boosted, PI-based regimen, as measured by mean change from baseline in each treatment group. The Last Observation Carry Forward (LOCF) approach was applied to missing data and data collected after a participant initiated lipid-modifying therapy. (NCT02397096)
Timeframe: Baseline and Week 24

,
Interventionmg/dL (Mean)
BaselineChange from Baseline
Delayed Switch (Ritonavir-boosted, PI-based) Group (DSG)109.00-1.94
Immediate Switch (Ritonavir--boosted, PI-based) Group (ISG)108.82-16.54

[back to top]

Mean Change From Baseline in Fasting Non-high-density Lipoprotein Cholesterol (Non-HDL-C)

Serum non-HDL-C was determined after an overnight fast. Change from Baseline was analyzed using ANCOVA models with terms for Baseline lipid level and treatment group. The Last Observation Carry Forward (LOCF) approach was applied for missing data or data collected after modifying lipid lowering therapy. (NCT02397096)
Timeframe: Baseline and Week 24

,
Interventionmg/dL (Mean)
BaselineChange from Baseline
Delayed Switch (Ritonavir-boosted, PI-based) Group (DSG)137.99-1.31
Immediate Switch (Ritonavir--boosted, PI-based) Group (ISG)139.14-24.74

[back to top]

Percentage of Participants Maintaining HIV-1 RNA <50 Copies/mL

"The percentage of participants in each arm achieving HIV-1 RNA levels <50 copies/mL was determined. Plasma HIV-1 RNA levels were quantified with the Abbott RealTime HIV-1 Assay. Data were handled according to the US Food and Drug Administration (FDA) snapshot approach and all missing data were considered treatment failures, regardless of the reason." (NCT02397096)
Timeframe: Week 24

InterventionPercentage of Participants (Number)
Immediate Switch Group (ISG)93.7
Delayed Switch Group (DSG)94.6

[back to top]

Percentage of Participants Maintaining Human Immunodeficiency Virus-1 Ribonucleic Acid (HIV-1 RNA) <50 Copies/mL

"The percentage of participants in each arm achieving HIV-1 RNA levels <50 copies/mL was determined. Plasma HIV-1 RNA levels were quantified with the Abbott RealTime HIV-1 Assay. Data were handled according to the US Food and Drug Administration (FDA) snapshot approach and all missing data were considered treatment failures, regardless of the reason." (NCT02397096)
Timeframe: Immediate Switch to MK-1439A arm: Week 48; Delayed Switch to MK-1439A arm: Week 24

InterventionPercentage of Participants (Number)
Immediate Switch Group (ISG)90.8
Delayed Switch Group (DSG)94.6

[back to top]

Percentage of Participants With HIV-1 RNA >=50 Copies/mL

"The percentage of participants in each arm achieving HIV-1 RNA levels >=50 copies/mL was determined. Plasma HIV-1 RNA levels were quantified with the Abbott RealTime HIV-1 Assay. Data were handled according to the US Food and Drug Administration (FDA) snapshot approach." (NCT02397096)
Timeframe: Immediate Switch to MK-1439A arm: Week 48; Delayed Switch to MK-1439A arm: Week 24

InterventionPercentage of Participants (Number)
Immediate Switch Group (ISG)1.6
Delayed Switch Group (DSG)1.8

[back to top]

Mean Change From Baseline in Cluster of Differentiation (CD4) Cell Counts

The mean change from baseline in CD4 cell counts at Week 48 was assessed using the Observed Failure (OF) approach. With the OF approach, baseline values were carried forward for participants who discontinued due to lack of efficacy. Cell counts were measured and expressed as cells/mm^3, and percent change was then calculated. CD4 cell counts were quantified by a central laboratory using a commercially available assay. (NCT02397096)
Timeframe: Baseline and Week 24

,
Interventioncells/mm^3 (Geometric Mean)
BaselineChange from Baseline
Delayed Switch Group (DSG)655.618.0
Immediate Switch Group (ISG)664.55.1

[back to top]

Percentage of Participants Discontinuing From Study Medication Due to an AE(s)

An AE is defined as any untoward medical occurrence in a participant administered a pharmaceutical product and which does not necessarily have to have a causal relationship with this treatment. (NCT02397096)
Timeframe: Up to Week 24

InterventionPercentage of Participants (Number)
Immediate Switch Group (ISG)2.5
Delayed Switch Group (DSG)0.4

[back to top]

Percentage of Participants Experiencing ≥1 Adverse Event (AE)

An AE is defined as any untoward medical occurrence in a participant administered a pharmaceutical product and which does not necessarily have to have a causal relationship with this treatment. (NCT02397096)
Timeframe: Up to week 24

InterventionPercentage of Participants (Number)
Immediate Switch Group (ISG)68.9
Delayed Switch Group (DSG)52.5

[back to top]

Percentage of Participants Experiencing ≥1 Serious Adverse Event (SAE)

A serious adverse event is an AE that results in death, is life threatening, results in persistent or significant disability or incapacity, results in or prolongs a hospitalization, is a congenital anomaly or birth defect, is a cancer, is associated with an overdose, or is another important medical event. The percentage of participants with any SAE was assessed. (NCT02397096)
Timeframe: Up to 24 weeks

InterventionPercentage of Participants (Number)
Immediate Switch Group (ISG)2.9
Delayed Switch Group (DSG)3.6

[back to top]

Percentage of Participants Maintaining HIV-1 RNA <40 Copies/mL

"The percentage of participants in each arm achieving HIV-1 RNA levels <40 copies/mL was determined. Plasma HIV-1 RNA levels were quantified with the Abbott RealTime HIV-1 Assay. Data were handled according to the US Food and Drug Administration (FDA) snapshot approach and all missing data were considered treatment failures, regardless of the reason." (NCT02397096)
Timeframe: Immediate Switch to MK-1439A arm: Week 48; Delayed Switch to MK-1439A arm: Week 24

InterventionPercentage of Participants (Number)
Immediate Switch Group (ISG)89.7
Delayed Switch Group (DSG)93.3

[back to top]

Percentage of Participants 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]

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]

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 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 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]

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]

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]

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]

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]

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]

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]

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]

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 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 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 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 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 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 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]

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 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 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]

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 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]

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 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]

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]

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 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]

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 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]

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: 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: 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 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 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 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: 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 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: 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 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 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: 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 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 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 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: 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 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: 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: 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: 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 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]

Proportion of Participants With Undetectable Cell-associated HIV-1 DNA (CAHD) Prior to ART Initiation

Proportion of participants with 0 copies of CAHD per million CD4+ blood-derived CD4+ T-cells (assayed by quantitative PCR [qPCR]), assessed separately and jointly by integrase and gag assays. In order for a participant to be considered as having 0 copies of CAHD for joint assays, the participant must be found to have an undetectable CAHD result from both integrase and gag assays. If all participants in both arms had undetectable CAHD then the statistical test was not performed. (NCT02859558)
Timeframe: At week 0

,,
InterventionProportion of participants (Number)
Joint Assay (Integrase + Gag)Integrase AssayGag Assay
Arm 1: Fiebig I/II0.000.040.00
Arm 2: Fiebig III/IV0.010.030.01
Arm 3: Fiebig V0.000.070.00

[back to top]

HIV-1-specific CD8+ and T-cell Responses to Nef, Gag, Pol and Env by Flow Cytometry

"Percent of HIV-1-specific CD8+ T-cells expressing any cytokine/marker (CD40L, CD107a, IFNg, MIP1B, TNFa) to each of the 4 protein stimulants (nef, gag, pol and env) by flow cytometry while HIV-1 RNA is suppressed on ART. The results for a specific participant are calculated by subtracting the corresponding background control value (media control). If the result would be less than zero after background subtraction, the result was set to zero.~Cytokine/Marker Names: CD40 ligand (CD40L); Cluster of Differentiation 107a (CD107a); Interferon gamma (IFNg); Macrophage Inflammatory Protein beta (MIP1B); Tumor Necrosis Factor alpha (TNFa)" (NCT02859558)
Timeframe: At 48 weeks

,,
InterventionPercentage of CD8+ T-cells (Median)
CD8+ T-cell Response to EnvCD8+ T-cell Response to GagCD8+ T-cell Response to NefCD8+ T-cell Response to Pol
Arm 1: Fiebig I/II0.000.150.030.00
Arm 2: Fiebig III/IV0.000.330.150.05
Arm 3: Fiebig V0.000.280.330.08

[back to top]

HIV-1-specific CD4+ and T-cell Responses to Nef, Gag, Pol and Env by Flow Cytometry

"Percent of HIV-1-specific CD4+ T-cells expressing any cytokine/marker (CD40L, CD107a, IFNg, MIP1B, TNFa) to each of the 4 protein stimulants (nef, gag, pol and env) by flow cytometry while HIV-1 RNA is suppressed on ART. The results for a specific participant are calculated by subtracting the corresponding background control value (media control). If the result would be less than zero after background subtraction, the result was set to zero.~Cytokine/Marker Names: CD40 ligand (CD40L); Cluster of Differentiation 107a (CD107a); Interferon gamma (IFNg); Macrophage Inflammatory Protein beta (MIP1B); Tumor Necrosis Factor alpha (TNFa)" (NCT02859558)
Timeframe: At week 48

,,
InterventionPercentage of CD4+ T-cells (Median)
CD4+ T-cell Response to EnvCD4+ T-cell Response to GagCD4+ T-cell Response to NefCD4+ T-cell Response to Pol
Arm 1: Fiebig I/II0.000.060.040.00
Arm 2: Fiebig III/IV0.000.190.100.04
Arm 3: Fiebig V0.080.140.100.04

[back to top]

Proportion of Participants With Undetectable Cell-associated HIV-1 DNA (CAHD)

Proportion of participants with 0 copies of CAHD per 5 million CD4+ blood-derived CD4+ T-cells (assayed by quantitative polymerase chain reaction [qPCR]), assessed separately and jointly by integrase and gag assays. In order for a participant to be considered as having 0 copies of CAHD for joint assays, the participant must be found to have an undetectable CAHD result from both integrase and gag assays. If all participants in both arms had undetectable CAHD then the statistical test was not performed. (NCT02859558)
Timeframe: At week 48

,,
InterventionProportion of participants (Number)
Joint Assay (Integrase + Gag)Integrase AssayGag Assay
Arm 1: Fiebig I/II0.000.100.03
Arm 2: Fiebig III/IV0.000.060.02
Arm 3: Fiebig V0.000.100.00

[back to top]

Concentration at End of Dosing Interval (Ctau) for GSK2838232 on Day 10

Serial plasma samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232. (NCT03045861)
Timeframe: 24 hours post-dose on Day 10

InterventionNanograms per milliliter (Geometric Mean)
GSK2838232 20 mg19.24
GSK2838232 50 mg31.76
GSK2838232 100 mg78.98
GSK2838232 200 mg180.3

[back to top]

Number of Participants With Abnormal Urine Parameters

Urine samples were collected for the assessment of following urine parameters by dipstick method: pH, glucose, protein, blood and ketones. The number of participants with abnormal urine parameters is presented. (NCT03045861)
Timeframe: Up to Day 22

InterventionParticipants (Count of Participants)
GSK2838232 20 mg0
GSK2838232 50 mg0
GSK2838232 100 mg0
GSK2838232 200 mg0

[back to top]

Number of Participants Who Were Administered Concomitant Medications

Concomitant medications (prescription and non-prescription) were administered only as medically necessary during the study. Number of participants who received any concomitant medications is presented. (NCT03045861)
Timeframe: Up to Day 22

InterventionParticipants (Count of Participants)
GSK2838232 20 mg2
GSK2838232 50 mg3
GSK2838232 100 mg3
GSK2838232 200 mg3

[back to top]

Maximum Observed Concentration (Cmax) for GSK2838232 on Day 1

Serial plasma samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232. (NCT03045861)
Timeframe: Pre dose and 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12 and 24 hours post-dose on Day 1

InterventionNanograms per milliliter (Geometric Mean)
GSK2838232 20 mg12.65
GSK2838232 50 mg32.39
GSK2838232 100 mg65.62
GSK2838232 200 mg190.0

[back to top]

Maximum Decline From Baseline in Plasma HIV-1 Ribonucleic Acid (RNA)

Plasma samples were collected for quantitative analysis of plasma HIV-1 RNA. An HIV-1 RNA polymerase chain reaction (PCR) assay with a lower limit of detection (LLOD) of 50 copies/milliliter (ultrasensitive assay) was used for post-baseline assessments. Baseline value was the value at latest pre-dose assessment. The maximum decline was determined using change from Baseline in plasma HIV-RNA values at each time point. The analysis was performed on Intent To Treat (ITT) Population which comprised of all participants who met study criteria and were enrolled into the study with documented evidence of having received at least 1 dose of treatment and at least one post-Baseline HIV-1 RNA measurement. (NCT03045861)
Timeframe: Baseline (Day 1) to Day 21

InterventionCopies per milliliter (Mean)
GSK2838232 20 mg-42095.14
GSK2838232 50 mg-49065.88
GSK2838232 100 mg-32947.50
GSK2838232 200 mg-33149.13

[back to top]

Concentration of GSK2838232 at 24 Hours Post-dose on Day 1

Serial plasma samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232. (NCT03045861)
Timeframe: 24 hours post-dose on Day 1

InterventionNanograms per milliliter (Geometric Mean)
GSK2838232 20 mg5.890
GSK2838232 50 mg13.45
GSK2838232 100 mg30.72
GSK2838232 200 mg87.33

[back to top]

Change From Baseline to Day 11 in CD4+ Count Relative to Day 10 AUC (0 to Tau)

The relationship between pharmacokinetic parameters (AUC) and pharmacodynamic measures (change from Baseline CD4+ cell count) was explored using a frequentist linear model. The model parameters estimated included slope and intercept. The estimate (slope) along with 95% confidence interval is presented. (NCT03045861)
Timeframe: Baseline (Day 1), Days 10 and 11

Interventioncells/microliter/ng*h/mL (Number)
Participants Receiving GSK2838232-0.003

[back to top]

Change From Baseline to Day 11 in CD4+ Count Relative to Day 10 Cmax

The relationship between pharmacokinetic parameters (Cmax) and pharmacodynamic measures (change from Baseline CD4+ cell count) was explored using a frequentist linear model. The model parameters estimated included slope and intercept. The estimate (slope) along with 95% confidence interval is presented (NCT03045861)
Timeframe: Baseline (Day 1), Days 10 and 11

Interventioncells/microliter/nanograms/milliliter (Number)
Participants Receiving GSK2838232-0.067

[back to top]

Change From Baseline to Day 11 in CD4+ Count Relative to Day 10 Ctau

The relationship between pharmacokinetic parameters (Ctau) and pharmacodynamic measures (change from Baseline CD4+ cell count) was explored using a frequentist linear model. The model parameters estimated included slope and intercept. The estimate (slope) along with 95% confidence interval is presented. (NCT03045861)
Timeframe: Baseline (Day 1), Days 10 and 11

Interventioncells/microliter/nanograms/milliliter (Number)
Participants Receiving GSK2838232-0.079

[back to top]

Number of Participants With Liver Function Laboratory Abnormalities of Potential Clinical Importance

Blood samples were collected for the assessment of liver function parameters. The clinical concern range for liver function parameters were: albumin (low: <30 g/L), total protein (low: <15 and high: >15 g/L), alanine aminotransferase (high: >=2 times upper limit of normal [ULN]); aspartate aminotransferase (high: >=2 times ULN); alkaline phosphatase (high: >=2 times ULN); total bilirubin (high: >=1.5 times ULN); direct bilirubin (high: >0.3 times ULN). (NCT03045861)
Timeframe: Up to Day 22

,,,
InterventionParticipants (Count of Participants)
Albumin; <30Total Protein; ULN+15Total Protein; lower limit of normal (LLN)-15ALT; >=2 times ULNAST; >=2 times ULNALP; >=2 times ULNTotal bilirubin; >=1.5 times ULNDirect bilirubin; >=0.3 times ULN
GSK2838232 100 mg00000010
GSK2838232 20 mg00000000
GSK2838232 200 mg00000000
GSK2838232 50 mg00001000

[back to top]

Change From Baseline to Day 11 in log10 Plasma HIV-1 RNA Relative to Day 10 AUC (0 to Tau)

Plasma samples were collected for quantitative analysis of HIV-1 RNA. Change from Baseline is the value at indicated time point minus Baseline value. Statistical analysis for relationship between pharmacokinetic parameters (AUC) and pharmacodynamic measures (Change from Baseline in plasma HIV-1 RNA) was explored using a frequentist three parameter Emax non-linear model. The model parameters estimated included: maximum response (Emax), pharmacokinetic parameter value that attains 50% of the maximal effect (ED50) and residual variability (s2e). Pharmacokinetic/Pharmacodynamic Population comprised of participants who met criteria for Per-Protocol (all participants who met study criteria and are enrolled into the study with documented evidence of having received all doses and all post-baseline HIV-1 RNA measurement, with exceptions of those who have at least one major protocol deviation) and Pharmacokinetic Population analysis sets and who underwent pharmacodynamic sampling during study. (NCT03045861)
Timeframe: Baseline (Day 1), Days 10 and 11

Interventionlog10 copies per milliliter (Mean)
GSK2838232 20 mg-0.4979
GSK2838232 50 mg-1.1671
GSK2838232 100 mg-1.1745
GSK2838232 200 mg-1.5994

[back to top]

Change From Baseline to Day 11 in log10 Plasma HIV-1 RNA Relative to Day 10 Cmax

Plasma samples were collected for quantitative analysis of HIV-1 RNA. Change from Baseline is the value at indicated time point minus Baseline value. Statistical analysis for the relationship between pharmacokinetic parameters (Cmax) and pharmacodynamic measures (change from Baseline in log10 plasma HIV-1 RNA) was explored using a frequentist three parameter Emax non-linear model. The model parameters estimated included: maximum response (Emax), pharmacokinetic parameter value that attains the 50% of the maximal effect (ED50) and s2e. (NCT03045861)
Timeframe: Baseline (Day 1), Days 10 and 11

Interventionlog10 copies per milliliter (Mean)
GSK2838232 20 mg-0.4979
GSK2838232 50 mg-1.1671
GSK2838232 100 mg-1.1745
GSK2838232 200 mg-1.5994

[back to top]

Change From Baseline to Day 11 in log10 Plasma HIV-1 RNA Relative to Day 10 Ctau

Plasma samples were collected for quantitative analysis of HIV-1 RNA. Change from Baseline is the value at indicated time point minus Baseline value. Statistical analysis for the relationship between pharmacokinetic parameters (Ctau) and pharmacodynamic measures (change from Baseline in log10 plasma HIV-1 RNA) was explored using a frequentist three parameter Emax non-linear model. The model parameters estimated included: maximum response (Emax), pharmacokinetic parameter value that attains the 50% of the maximal effect (ED50) and s2e. (NCT03045861)
Timeframe: Baseline (Day 1), Days 10 and 11

Interventionlog10 copies per milliliter (Mean)
GSK2838232 20 mg-0.4979
GSK2838232 50 mg-1.1671
GSK2838232 100 mg-1.1745
GSK2838232 200 mg-1.5994

[back to top]

Cmax for GSK2838232 on Day 10

Serial plasma samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232. (NCT03045861)
Timeframe: Pre dose and 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12 and 24 hours post-dose on Day 10

InterventionNanograms per milliliter (Geometric Mean)
GSK2838232 20 mg32.65
GSK2838232 50 mg56.40
GSK2838232 100 mg157.9
GSK2838232 200 mg306.5

[back to top]

Change From Baseline in Cluster of Differentiation 4+ (CD4+) Cell Count to Day 11

CD4+ cell counts were assessed by flow cytometry. Baseline value is the latest pre-dose assessment value. Change from Baseline is calculated as the post-dose visit value minus Baseline value. (NCT03045861)
Timeframe: Baseline (Day 1) and Day 11

InterventionCells per microliter (Mean)
GSK2838232 20 mg-1.4
GSK2838232 50 mg52.0
GSK2838232 100 mg40.7
GSK2838232 200 mg11.1

[back to top]

Absorption Lag Time (Tlag) for GSK2838232 on Day 1

Serial plasma samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232. (NCT03045861)
Timeframe: Pre dose and 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12 and 24 hours post-dose on Day 1

InterventionHours (Median)
GSK2838232 20 mg1.083
GSK2838232 50 mg1.000
GSK2838232 100 mg0.500
GSK2838232 200 mg0.500

[back to top]

Time to Maximum Observed Concentration (Tmax) for GSK2838232 on Day 1

Serial plasma samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232. (NCT03045861)
Timeframe: Pre dose and 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12 and 24 hours post-dose on Day 1

InterventionHours (Median)
GSK2838232 20 mg4.033
GSK2838232 50 mg3.250
GSK2838232 100 mg6.000
GSK2838232 200 mg4.000

[back to top]

Terminal Elimination Half-life (T1/2) of GSK2838232 Following Administration on Day 10

Serial plasma samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232. (NCT03045861)
Timeframe: Pre dose and 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12 and 24 hours post-dose on Day 10

InterventionHours (Geometric Mean)
GSK2838232 20 mg19.221
GSK2838232 50 mg16.298
GSK2838232 100 mg18.113
GSK2838232 200 mg16.351

[back to top]

Steady State Assessment of Plasma Pre-dose Concentrations by Treatment

A linear mixed model using Day, treatment and Day by treatment as fixed effects and participant as a random effect on the log-transformed pre-dose values was performed to evaluate if steady state was achieved using the Helmert transformation approach. The comparison was done as Day 8 versus the average of Days 9 and 10 values. The ratio of geometric least square mean for Day 8 versus average of Days 9 and 10 values is presented along with 95% confidence interval. (NCT03045861)
Timeframe: Pre-dose on Days 8, 9 and 10

InterventionRatio (Number)
GSK2838232 20 mg0.924
GSK2838232 50 mg0.999
GSK2838232 100 mg0.958
GSK2838232 200 mg0.961

[back to top]

Apparent Oral Clearance of GSK2838232 Following Administration on Day 10

Serial plasma samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232. (NCT03045861)
Timeframe: Pre dose and 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12 and 24 hours post-dose on Day 10

InterventionLiters per hour (Geometric Mean)
GSK2838232 20 mg33.91
GSK2838232 50 mg48.84
GSK2838232 100 mg36.67
GSK2838232 200 mg35.31

[back to top]

Area Under the Concentration-time Curve Over the Dosing Interval (AUC [0 to Tau]) for GSK2838232 on Day 10

Serial plasma samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232. (NCT03045861)
Timeframe: Pre dose and 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12 and 24 hours post-dose on Day 10

InterventionHours*nanograms per milliliter (Geometric Mean)
GSK2838232 20 mg590
GSK2838232 50 mg1024
GSK2838232 100 mg2727
GSK2838232 200 mg5664

[back to top]

Area Under the Plasma Concentration Time Curve From Zero (Pre-dose) to 24 Hours (AUC[0 to 24]) for GSK2838232 on Day 1

Serial plasma samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232. The analysis was performed on Pharmacokinetic Population which comprised of participants who received GSK2838232 and underwent plasma pharmacokinetic sampling during the study. (NCT03045861)
Timeframe: Pre dose and 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12 and 24 hours post-dose on Day 1

InterventionHours*nanograms per milliliter (Geometric Mean)
GSK2838232 20 mg187
GSK2838232 50 mg453
GSK2838232 100 mg966
GSK2838232 200 mg2752

[back to top]

Pre-morning Dose Concentrations (C0) on Day 2 Through 11

Blood samples were collected for pharmacokinetic analysis of GSK2838232. The pre-morning dose concentrations for Days 2 to 11 is presented. One participant from GSK2838232 100 mg arm was dosed with GSK2838232 50 mg on Days 1 and 2; hence, pharmacokinetic parameters for that participant were summarized with GSK2838232 50 mg for Days 1 and 2 and with GSK2838232 100 mg for Days 3 to 10. (NCT03045861)
Timeframe: Pre-dose on Days 2, 3, 4, 5, 8, 9, 10 and 11

,,,
InterventionNanograms per milliliter (Mean)
Day 2; n=7, 9, 9, 8Day 3; n=7, 8, 10, 8Day 4; n=7, 7, 10, 8Day 5; n=4, 6, 10, 7Day 8; n=7, 8, 10, 8Day 9; n=7, 6, 10, 8Day 10; n=7, 7, 10, 8Day 11; n=7, 7, 10, 8
GSK2838232 100 mg34.1751.2764.4976.1486.3989.3284.5289.87
GSK2838232 20 mg6.0813.9914.6615.0319.7220.5320.7620.16
GSK2838232 200 mg96.94163.98176.28206.29187.16206.75189.05219.23
GSK2838232 50 mg16.3929.0331.8034.3543.1443.8339.1935.09

[back to top]

Number of Participants With Vital Signs Data Outside Clinical Concern Range

Vital signs were measured in a semi-supine position after 5 minutes rest and included temperature, systolic and diastolic blood pressure and pulse rate. The clinical concern range for vital signs were: systolic blood pressure (SBP) (low: <85 and high: >160 millimeters of mercury [mmHg]) and diastolic blood pressure (DBP) (low: <45 and high: >100 mmHg). Number of participants with vital signs data outside clinical concern range is presented. (NCT03045861)
Timeframe: Day 1 (pre-dose)

,,,
InterventionParticipants (Count of Participants)
SBP; >Clinical concern rangeSBP; DBP; >Clinical concern rangeDBP;
GSK2838232 100 mg1010
GSK2838232 20 mg0000
GSK2838232 200 mg0000
GSK2838232 50 mg0000

[back to top]

Pre-dose Concentration (C0) of GSK2838232 on Day 10

Serial plasma samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232. (NCT03045861)
Timeframe: Pre dose on Day 10

InterventionNanograms per milliliter (Geometric Mean)
GSK2838232 20 mg20.02
GSK2838232 50 mg36.66
GSK2838232 100 mg75.31
GSK2838232 200 mg155.5

[back to top]

Number of Participants With Hematology Parameter Abnormalities of Potential Clinical Importance

Blood samples were collected for the assessment of hematology parameters. The clinical concern range for the parameters were: hematocrit (high: >0.54 proportion of red blood cells in blood); hemoglobin (high: >180 grams per liter [g/L]), lymphocytes (low: <0.8x10^9 cells/L); neutrophil count (low: <1.5x10^9 cells/L); platelet count (low: <100x10^9 cells/L and high: >550x10^9 cells/L); white blood cells (low: <3x10^9 cells/L and high: >20x10^9cells/L). Data for any visit post-Baseline is reported. (NCT03045861)
Timeframe: Up to Day 22

,,,
InterventionParticipants (Count of Participants)
Hematocrit; >0.54Hematocrit; decline from Baseline >0.075Hemoglobin; >180Hemoglobin; decline from Baseline >25Lymphocytes; <0.8Neutrophils; <1.5Platelets; >550Platelets; <100White blood cells; >20White blood cells; <3
GSK2838232 100 mg1301030100
GSK2838232 20 mg0100010000
GSK2838232 200 mg1000010001
GSK2838232 50 mg0100000000

[back to top]

Number of Participants With Emergent Drug Resistance Mutations

Plasma samples were collected to evaluate treatment-emergent genotypic mutations in Gag, reverse transcriptase (RT) and protease (PR) and to assess phenotypic resistance to GSK2838232 and RT and PR drugs. Number of participants with treatment emergent RT/PR mutations, reduced susceptibility to nucleoside/nucleotide reverse transcriptase inhibitor (NRTI), non-nucleoside reverse transcriptase inhibitor (NNRTI), or protease inhibitor (PI), treatment emergent maturation inhibitor A364A/V and GSK2838232 phenotypic resistance is presented. (NCT03045861)
Timeframe: Up to Day 11

,,,
InterventionParticipants (Count of Participants)
RT/PR mutationsReduced susceptibility to NRTIs, NNRTIs, PIsGSK2838232 phenotypic resistanceTreatment emergent A364A/V
GSK2838232 100 mg0001
GSK2838232 20 mg0000
GSK2838232 200 mg0000
GSK2838232 50 mg0011

[back to top]

Number of Participants With Clinical Chemistry Abnormalities of Potential Clinical Importance

Blood samples were collected for the assessment of clinical chemistry parameters. The clinical concern range for the parameters were: carbon dioxide/bicarbonate (low: <18 millimoles per liter [mmol/L] and high: >32 mmol/L); urea (high: >9 mmol/L); creatinine (high: change from Baseline >44.2 micromoles per liter [µmol/L]), glucose (low: <3 and high: >9 mmol/L); potassium (low: <3 and high: >5.5 mmol/L); troponin I (high: >=0.01 micrograms per liter [µg/L]) and sodium (low: <130 mmol/L and high: >150 mmol/L). Data for any visit post-Baseline is reported. (NCT03045861)
Timeframe: Up to Day 22

,,,
InterventionParticipants (Count of Participants)
Carbon dioxide/bicarbonate; >32 mmol/LCarbon dioxide/bicarbonate; <18 mmol/LUrea; >9 mmol/LCreatinine; Change >44.2 mmol/LGlucose; >9 mmol/LGlucose; <3 mmol/LPotassium; >5.5 mmol/LPotassium; <3 mmol/LSodium; >150 mmol/LSodium; <130 mmol/LTroponin I; >=0.01 µg/L
GSK2838232 100 mg030000000010
GSK2838232 20 mg00100000007
GSK2838232 200 mg00000020008
GSK2838232 50 mg00000000008

[back to top]

Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)

An AE is any untoward medical occurrence in a participant or clinical investigation participant, temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product. SAE is defined as any untoward medical occurrence that, at any dose: results in death; is life threatening; requires hospitalization or prolongation of existing hospitalization; results in disability/incapacity; is a congenital anomaly/birth defect; other important medical events; is associated with liver injury and impaired liver function. Safety Population comprised of all participants who received at least one dose of study treatment. (NCT03045861)
Timeframe: Up to Day 22

,,,
InterventionParticipants (Count of Participants)
Any AEsAny SAEs
GSK2838232 100 mg50
GSK2838232 20 mg30
GSK2838232 200 mg50
GSK2838232 50 mg30

[back to top]

Number of Participants With Abnormal Electrocardiogram (ECG) Findings

Triplicate 12-lead ECGs were obtained using an ECG machine that automatically calculated the heart rate and measured PR, QRS, QT, and corrected QT (QTc) intervals. Number of participants with abnormal ECG findings at worst-case post Baseline is presented. (NCT03045861)
Timeframe: Up to Day 22

,,,
InterventionParticipants (Count of Participants)
Abnormal-not clinically significantAbnormal-clinically significant
GSK2838232 100 mg80
GSK2838232 20 mg50
GSK2838232 200 mg40
GSK2838232 50 mg60

[back to top]

Dose Proportionality of GSK2838232

Blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232. Dose proportionality was assessed using a fixed effects power model. Estimated slope and 90% confidence interval is presented. (NCT03045861)
Timeframe: pre dose, and 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12 and 24 hours post-dose on Days 1 and 10

Interventionslope of log dose (Number)
AUC(0-24); Day 1Cmax; Day 1C24; Day 1AUC (0 to tau); Day 10Cmax; Day 10Ctau; Day 10
Participants Receiving GSK28382321.1571.1581.1701.0121.0130.988

[back to top]

Accumulation Ratio for GSK2838232

Serial blood samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232. The accumulation ratios were calculated as R_AUC=AUC(0-tau) Day 10/AUC(0-24) Day 1; R_Cmax=Cmax Day 10/Cmax Day 1 and R_Ctau=Ctau Day 10/C24 Day 1. (NCT03045861)
Timeframe: pre dose, and 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12 and 24 hours post-dose on Days 1 and 10; pre-dose on Days 3, 4, 5, 8 and 9; Days 12 and 14

,,,
InterventionRatio (Geometric Mean)
R_AUCR_CmaxR_Ctau
GSK2838232 100 mg3.2292.7533.038
GSK2838232 20 mg3.1492.5803.267
GSK2838232 200 mg2.2111.6682.210
GSK2838232 50 mg2.0041.4982.118

[back to top]

Tmax for GSK2838232 Following Administration on Day 10

Serial plasma samples were collected at indicated time points for pharmacokinetic analysis of GSK2838232. (NCT03045861)
Timeframe: Pre dose and 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12 and 24 hours post-dose on Day 10

InterventionHours (Median)
GSK2838232 20 mg6.000
GSK2838232 50 mg4.000
GSK2838232 100 mg6.000
GSK2838232 200 mg4.083

[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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

AUC0-∞ for Tenofovir (TFV)

AUC 0 to Infinity for TFV (ng*h/mL) (NCT03717129)
Timeframe: 72 hours

Interventionng*h/mL (Mean)
Genvoya Oral Dose253
Genvoya Crushed Dose241

[back to top]

EVG Cmax

Maximum Plasma Concentration (Cmax) for Elvitegravir (ng/mL) (NCT03717129)
Timeframe: 72 Hr

Interventionng/mL (Mean)
Genvoya Oral Dose1650
Genvoya Crushed Dose1946

[back to top]

FTC Half-life

Terminal elimination half-life for FTC (hours). (NCT03717129)
Timeframe: 72 hours

Interventionhours (Mean)
Genvoya Oral Dose15.2
Genvoya Crushed Dose16.1

[back to top]

EVG Half-life

Terminal elimination half-life for EVG (hours). (NCT03717129)
Timeframe: 72 hours

Interventionhours (Mean)
Genvoya Oral Dose5
Genvoya Crushed Dose5.1

[back to top]

Area Under the Curve From 0 to Infinity (AUC0-∞) for EVG

AUC 0 to Infinity for Elvitegravir (ng*h/mL) (NCT03717129)
Timeframe: 72 hours

Interventionng*h/mL (Mean)
Genvoya Oral Dose24219
Genvoya Crushed Dose26948

[back to top]

FTC Cmax

Maximum Plasma Concentration (Cmax) for Emtricitabine (ng/mL) (NCT03717129)
Timeframe: 72 hr

Interventionng/mL (Mean)
Genvoya Oral Dose2095
Genvoya Crushed Dose1968

[back to top]

TFV Cmax

Maximum Plasma Concentration (Cmax) for Tenofovir (ng/mL) (NCT03717129)
Timeframe: 72 hr

Interventionng/mL (Mean)
Genvoya Oral Dose11
Genvoya Crushed Dose9.5

[back to top]

TFV Half-life

Terminal elimination half-life for TFV (hours). (NCT03717129)
Timeframe: 72 hours

Interventionhours (Mean)
Genvoya Oral Dose42.5
Genvoya Crushed Dose40.7

[back to top]

AUC0-∞ for FTC

AUC 0 to Infinity for Emtricitabine (ng*h/mL) (NCT03717129)
Timeframe: 72 hours

Interventionng*h/mL (Mean)
Genvoya Oral Dose11603
Genvoya Crushed Dose10969

[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]

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]

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]

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]

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]

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]

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]

Rectal Tissue Concentration of Emtricitabine-triphosphate (FTC-TP)

Median drug concentrations of FTC-TP in rectal tissue were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionfmol/mg (Median)
Baseline24 hours96 hours
Genvoya - 24 and 96 Hours Specimen Collection0104.312.5

[back to top]

Rectal Tissue Concentration of Emtricitabine-triphosphate (FTC-TP)

Median drug concentrations of FTC-TP in rectal tissue were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionfmol/mg (Median)
Baseline2 hours48 hours
Genvoya - 2 and 48 Hours Specimen Collection0187.556.3

[back to top]

Rectal Tissue Concentration of Emtricitabine-triphosphate (FTC-TP)

Median drug concentrations of FTC-TP in rectal tissue were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionfmol/mg (Median)
Baseline8 hours
Genvoya - Single Time Point Specimen Collection0116.5

[back to top]

Rectal Tissue Concentration of Emtricitabine (FTC)

Median drug concentrations in rectal tissue of the FTC component of Genvoya were determined at baseline and at each of the protocol specified time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mg (Median)
Baseline4 hours72 hours
Genvoya - 4 and 72 Hours Specimen Collection09.250

[back to top]

Rectal Tissue Concentration of Emtricitabine (FTC)

Median drug concentrations in rectal tissue of the FTC component of Genvoya were determined at baseline and at each of the protocol specified time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mg (Median)
Baseline24 hours96 hours
Genvoya - 24 and 96 Hours Specimen Collection01.230.02

[back to top]

Rectal Tissue Concentration of Emtricitabine (FTC)

Median drug concentrations in rectal tissue of the FTC component of Genvoya were determined at baseline and at each of the protocol specified time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mg (Median)
Baseline8 hours
Genvoya - Single Time Point Specimen Collection00.17

[back to top]

Rectal Tissue Concentration of Elvitegravir (EVG)

Median drug concentrations in rectal tissue of the EVG component of Genvoya were determined at baseline and at each of the protocol specified time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 72, 96 hours after taking the second dose of medication

Interventionng/mg (Median)
Baseline4 hours72 hours
Genvoya - 4 and 72 Hours Specimen Collection041.470

[back to top]

Rectal Tissue Concentration of Elvitegravir (EVG)

Median drug concentrations in rectal tissue of the EVG component of Genvoya were determined at baseline and at each of the protocol specified time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 72, 96 hours after taking the second dose of medication

Interventionng/mg (Median)
Baseline24 hours96 hours
Genvoya - 24 and 96 Hours Specimen Collection00.600.02

[back to top]

Rectal Tissue Concentration of Elvitegravir (EVG)

Median drug concentrations in rectal tissue of the EVG component of Genvoya were determined at baseline and at each of the protocol specified time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 72, 96 hours after taking the second dose of medication

Interventionng/mg (Median)
Baseline2 hours48 hours
Genvoya - 2 and 48 Hours Specimen Collection051.231.03

[back to top]

Peripheral Blood Mononuclear Cell (PBMC) Concentration of Emtricitabine-triphosphate (FTC-TP)

A peripheral blood mononuclear cell (PBMC) is any peripheral blood cell having a round nucleus: lymphocytes (T cells, B cells, NK cells) and monocytes. Median drug levels were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionfmol/10^6 cells (Median)
Baseline8 hours
Genvoya - Single Time Point Specimen Collection06855

[back to top]

Peripheral Blood Mononuclear Cell (PBMC) Concentration of Emtricitabine-triphosphate (FTC-TP)

A peripheral blood mononuclear cell (PBMC) is any peripheral blood cell having a round nucleus: lymphocytes (T cells, B cells, NK cells) and monocytes. Median drug levels were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionfmol/10^6 cells (Median)
Baseline24 hours96 hours
Genvoya - 24 and 96 Hours Specimen Collection05140714

[back to top]

Peripheral Blood Mononuclear Cell (PBMC) Concentration of Emtricitabine-triphosphate (FTC-TP)

A peripheral blood mononuclear cell (PBMC) is any peripheral blood cell having a round nucleus: lymphocytes (T cells, B cells, NK cells) and monocytes. Median drug levels were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionfmol/10^6 cells (Median)
Baseline4 hour72 hours
Genvoya - 4 and 72 Hours Specimen Collection07800955

[back to top]

Peripheral Blood Mononuclear Cell (PBMC) Concentration of Tenofovir-diphosphate (TFV-DP)

A peripheral blood mononuclear cell (PBMC) is any peripheral blood cell having a round nucleus: lymphocytes (T cells, B cells, natural killer (NK) cells) and monocytes. Median drug concentrations were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionfmol/10^6 cells (Median)
Baseline8 hours
Genvoya - Single Time Point Specimen Collection0375

[back to top]

Peripheral Blood Mononuclear Cell (PBMC) Concentration of Tenofovir-diphosphate (TFV-DP)

A peripheral blood mononuclear cell (PBMC) is any peripheral blood cell having a round nucleus: lymphocytes (T cells, B cells, natural killer (NK) cells) and monocytes. Median drug concentrations were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionfmol/10^6 cells (Median)
Baseline2 hours48 hours
Genvoya - 2 and 48 Hours Specimen Collection0569291

[back to top]

Peripheral Blood Mononuclear Cell (PBMC) Concentration of Tenofovir-diphosphate (TFV-DP)

A peripheral blood mononuclear cell (PBMC) is any peripheral blood cell having a round nucleus: lymphocytes (T cells, B cells, natural killer (NK) cells) and monocytes. Median drug concentrations were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionfmol/10^6 cells (Median)
Baseline24 hours96 hours
Genvoya - 24 and 96 Hours Specimen Collection0429117

[back to top]

Peripheral Blood Mononuclear Cell (PBMC) Concentration of Tenofovir-diphosphate (TFV-DP)

A peripheral blood mononuclear cell (PBMC) is any peripheral blood cell having a round nucleus: lymphocytes (T cells, B cells, natural killer (NK) cells) and monocytes. Median drug concentrations were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionfmol/10^6 cells (Median)
Baseline4 hours72 hours
Genvoya - 4 and 72 Hours Specimen Collection0621158

[back to top]

Plasma Concentration of Elvitegravir (EVG)

Median drug concentrations of the EVG component of Genvoya were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mL (Median)
Baseline8 hours
Genvoya - Single Time Point Specimen Collection0283

[back to top]

Plasma Concentration of Elvitegravir (EVG)

Median drug concentrations of the EVG component of Genvoya were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mL (Median)
Baseline2 hours48 hours
Genvoya - 2 and 48 Hours Specimen Collection022430

[back to top]

Plasma Concentration of Elvitegravir (EVG)

Median drug concentrations of the EVG component of Genvoya were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mL (Median)
Baseline24 hours96 hours
Genvoya - 24 and 96 Hours Specimen Collection02320

[back to top]

Plasma Concentration of Elvitegravir (EVG)

Median drug concentrations of the EVG component of Genvoya were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mL (Median)
Baseline4 hours72 hours
Genvoya - 4 and 72 Hours Specimen Collection018180

[back to top]

Plasma Concentration of Emtricitabine (FTC)

Median drug concentrations of the FTC component of Genvoya were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mL (Median)
Baseline8 hours
Genvoya - Single Time Point Specimen Collection0300

[back to top]

Plasma Concentration of Emtricitabine (FTC)

Median drug concentrations of the FTC component of Genvoya were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mL (Median)
Baseline2 hours48 hours
Genvoya - 2 and 48 Hours Specimen Collection027100

[back to top]

Plasma Concentration of Emtricitabine (FTC)

Median drug concentrations of the FTC component of Genvoya were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mL (Median)
Baseline24 hours96 hours
Genvoya - 24 and 96 Hours Specimen Collection01130

[back to top]

Plasma Concentration of Emtricitabine (FTC)

Median drug concentrations of the FTC component of Genvoya were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mL (Median)
Baseline4 hours72 hours
Genvoya - 4 and 72 Hours Specimen Collection017250

[back to top]

Plasma Concentration of Tenofovir (TFV)

Median drug concentrations of the TFV component of Genvoya were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mL (Median)
Baseline8 hours
Genvoya - Single Time Point Specimen Collection00

[back to top]

Rectal Tissue Concentration of Tenofovir-diphosphate (TFV-DP)

Median drug concentrations of TFV-DP in rectal tissue were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionfmol/mg (Median)
Baseline24 hours96 hours
Genvoya - 24 and 96 Hours Specimen Collection020.214.7

[back to top]

Rectal Tissue Concentration of Emtricitabine (FTC)

Median drug concentrations in rectal tissue of the FTC component of Genvoya were determined at baseline and at each of the protocol specified time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mg (Median)
Baseline2 hours48 hours
Genvoya - 2 and 48 Hours Specimen Collection09.540.02

[back to top]

Plasma Concentration of Tenofovir (TFV)

Median drug concentrations of the TFV component of Genvoya were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mL (Median)
Baseline24 hours96 hours
Genvoya - 24 and 96 Hours Specimen Collection000

[back to top]

Plasma Concentration of Tenofovir (TFV)

Median drug concentrations of the TFV component of Genvoya were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mL (Median)
Baseline4 hours72 hours
Genvoya - 4 and 72 Hours Specimen Collection000

[back to top]

Rectal Tissue Concentration of Tenofovir-diphosphate (TFV-DP)

Median drug concentrations of TFV-DP in rectal tissue were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionfmol/mg (Median)
Baseline4 hours72 hours
Genvoya - 4 and 72 Hours Specimen Collection045.911.9

[back to top]

Rectal Tissue Concentration of Elvitegravir (EVG)

Median drug concentrations in rectal tissue of the EVG component of Genvoya were determined at baseline and at each of the protocol specified time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 72, 96 hours after taking the second dose of medication

Interventionng/mg (Median)
Baseline8 hours
Genvoya - Single Time Point Specimen Collection00.31

[back to top]

Rectal Tissue Concentration of Tenofovir-diphosphate (TFV-DP)

Median drug concentrations of TFV-DP in rectal tissue were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionfmol/mg (Median)
Baseline2 hours48 hours
Genvoya - 2 and 48 Hours Specimen Collection039.118.2

[back to top]

Plasma Concentration of Tenofovir (TFV)

Median drug concentrations of the TFV component of Genvoya were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mL (Median)
Baseline2 hours48 hours
Genvoya - 2 and 48 Hours Specimen Collection000

[back to top]

Rectal Tissue Concentration of Tenofovir-diphosphate (TFV-DP)

Median drug concentrations of TFV-DP in rectal tissue were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionfmol/mg (Median)
Baseline8 hours
Genvoya - Single Time Point Specimen Collection023.7

[back to top]

Rectal Tissue Concentration of Tenofovir (TFN)

Median drug concentrations in rectal tissue of the TFN component of Genvoya were determined at baseline and at each of the protocol specified time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mg (Median)
Baseline4 hours72 hours
Genvoya - 4 and 72 Hours Specimen Collection00.300

[back to top]

Rectal Tissue Concentration of Tenofovir (TFN)

Median drug concentrations in rectal tissue of the TFN component of Genvoya were determined at baseline and at each of the protocol specified time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mg (Median)
Baseline24 hours96 hours
Genvoya - 24 and 96 Hours Specimen Collection00.030

[back to top]

Rectal Tissue Concentration of Tenofovir (TFN)

Median drug concentrations in rectal tissue of the TFN component of Genvoya were determined at baseline and at each of the protocol specified time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mg (Median)
Baseline2 hours48 hours
Genvoya - 2 and 48 Hours Specimen Collection000

[back to top]

Rectal Tissue Concentration of Tenofovir (TFN)

Median drug concentrations in rectal tissue of the TFN component of Genvoya were determined at baseline and at each of the protocol specified time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionng/mg (Median)
Baseline8 hours
Genvoya - Single Time Point Specimen Collection00.01

[back to top]

Rectal Tissue Concentration of Emtricitabine-triphosphate (FTC-TP)

Median drug concentrations of FTC-TP in rectal tissue were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionfmol/mg (Median)
Baseline4 hours72 hours
Genvoya - 4 and 72 Hours Specimen Collection0198.215.1

[back to top]

Peripheral Blood Mononuclear Cell (PBMC) Concentration of Emtricitabine-triphosphate (FTC-TP)

A peripheral blood mononuclear cell (PBMC) is any peripheral blood cell having a round nucleus: lymphocytes (T cells, B cells, NK cells) and monocytes. Median drug levels were determined at baseline and at each of the protocol specified follow-up time points. Concentrations below the limit of quantification (LOQ) are assigned a value of zero. (NCT03976752)
Timeframe: Baseline, and 2, 4, 8, 24, 48, 72, 96 hours after taking the second dose of medication

Interventionfmol/10^6 cells (Median)
Baseline2 hours48 hours
Genvoya - 2 and 48 Hours Specimen Collection077052120

[back to top]

OZ439 Fpo

OZ439 Absolute oral bioavailability (NCT04069221)
Timeframe: OZ439: Pre-dose, 0.25, 0.5, 0.75, 1, 2, 3, 4, 5, 6, 8, 12, 16, 24, 48, 72, 96, 168 hours post-dose. [14C]-OZ439: 10, 15, 20, 30 and 45 minutes after start of iv infusion then 3, 4, 5, 6, 8, 12, 16, 24, 48, 72, 96, 168 hours

InterventionAbsolute bioavailability in % (Geometric Mean)
Part 135

[back to top]

OZ439 AUC0-168h

Area under the OZ439 plasma concentration time curve from time zero to 168h (NCT04069221)
Timeframe: Pre-dose, 0.5, 0.75, 1, 2, 3, 4, 5, 6, 8, 12, 16, 24, 48, 72, 96, 168 hours post-dose

Interventionng.h/mL (Geometric Mean)
Part 2, Treatment B: Single Oral Dose of 800 mg OZ4399857
Part 2, Treatment C: Single Oral Dose of 400 mg OZ4396049
Part 2, Treatment D:Single Oral Dose 400 mg OZ439+Cobicistat13949

[back to top]

OZ439 AUC0-inf

Area under the OZ439 plasma concentration time curve from time zero to infinity (NCT04069221)
Timeframe: Pre-dose, 0.5, 0.75, 1, 2, 3, 4, 5, 6, 8, 12, 16, 24, 48, 72, 96, 168, 216, 264, 312 hours post-dose

Interventionng.h/mL (Geometric Mean)
Part 2, Treatment B: Single Oral Dose of 800 mg OZ43910222
Part 2, Treatment C: Single Oral Dose of 400 mg OZ4396378
Part 2, Treatment D:Single Oral Dose 400 mg OZ439+Cobicistat14984

[back to top]

OZ439 C168h

OZ439 concentration observed at 168h (NCT04069221)
Timeframe: 168 hours post-dose

Interventionng/mL (Geometric Mean)
Part 2, Treatment B: Single Oral Dose of 800 mg OZ4393.54
Part 2, Treatment C: Single Oral Dose of 400 mg OZ4392.48
Part 2, Treatment D:Single Oral Dose 400 mg OZ439+Cobicistat6.07

[back to top]

OZ439 Cmax

OZ439 maximum concentration observed (NCT04069221)
Timeframe: Pre-dose, 0.5, 0.75, 1, 2, 3, 4, 5, 6, 8, 12, 16, 24, 48, 72, 96, 168, 216, 264, 312 hours post-dose

Interventionng/mL (Geometric Mean)
Part 2, Treatment B: Single Oral Dose of 800 mg OZ439949
Part 2, Treatment C: Single Oral Dose of 400 mg OZ439649
Part 2, Treatment D:Single Oral Dose 400 mg OZ439+Cobicistat966

[back to top]