Page last updated: 2024-12-08

etravirine

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 CID193962
CHEMBL ID308954
CHEBI ID63589
SCHEMBL ID52691
MeSH IDM0420447

Synonyms (102)

Synonym
HY-90005
r165335-tmc125
tmc 125
benzonitrile, 4-[[6-amino-5-bromo-2-[(4-cyanophenyl)amino]-4-pyrimidinyl]oxy]-3,5-dimethyl-
intelence(tm)
diaminopyrimidine deriv
r165335 ,
etravirine ,
tmc125
tmc-125
dapy deriv
tmc-125/r-165335
4-[6-amino-5-bromo-2-(4-cyanoanilino)pyrimidin-4-yl]oxy-3,5-dimethyl-benzonitrile
intelence
r-165335
intelence (tn)
etravirine (jan/usan/inn)
D04112
269055-15-4
4-({6-amino-5-bromo-2-[(4-cyanophenyl)amino]pyrimidin-4-yl}oxy)-3,5-dimethylbenzonitrile
65b ,
chebi:63589 ,
CHEMBL308954 ,
4-(6-amino-5-bromo-2-(4-cyanophenylamino)pyrimidin-4-ylamino)-3,5-dimethylbenzonitrile
bdbm50103642
4-(6-amino-5-bromo-2-(4-cyanophenylamino)pyrimidin-4-yloxy)-3,5-dimethylbenzonitrile
4-[6-amino-5-bromo-2-(4-cyano-phenylamino)-pyrimidin-4-yloxy]-3,5-dimethyl-benzonitrile
FT-0658058
4-[6-amino-5-bromo-2-(4-cyanoanilino)pyrimidin-4-yl]oxy-3,5-dimethylbenzonitrile
4-((6-amino-5-bromo-2-((4-cyanophenyl)amino)-4-pyrimidinyl)oxy)-3,5-dimethyl-benzonitrile
benzonitrile, 4-((6-amino-5-bromo-2-((4-cyanophenyl)amino)-4-pyrimidinyl)oxy)-3,5-dimethyl-
tmc125 cpd
4-[6-azanyl-5-bromanyl-2-[(4-cyanophenyl)amino]pyrimidin-4-yl]oxy-3,5-dimethyl-benzenecarbonitrile
4-[[6-amino-5-bromo-2-(4-cyanoanilino)-4-pyrimidinyl]oxy]-3,5-dimethylbenzonitrile
A818671
BCP9000006
0c50hw4fo1 ,
etravirine [usan:inn:ban:jan]
4-(6-amino-5-bromo-2-(4-cyanoanilino)pyrimidin-4-yloxy)-3,5-dimethylbenzonitrile
unii-0c50hw4fo1
r 165335
BCP0726000193
PB32778
cyanophenyl)amino]pyrimidin-4-yl}oxy)-3,5-
4-({6-amino-5-bromo-2-[(4-
etravirine [vandf]
4-[[6-amino-5-bromo-2-[(4-cyanophenyl)amino]-4-pyrimidinyl]oxy]-3,5-dimethyl-benzonitrile
etravirine [orange book]
etravirine [who-dd]
etravirine [mi]
etravirine [mart.]
etravirine [jan]
etravirine [ema epar]
etravirine [usan]
etravirine [inn]
CS-0435
AKOS015896355
AM20080899
S3080
DB06414
4-[[6-amino-5-bromo-2-[(4-cyanophenyl)amino]-4-pyrimidinyl]oxy]-3,5-dimethylbenzonitrile
SCHEMBL52691
3M8P
HE-0088
mfcd09837879
PYGWGZALEOIKDF-UHFFFAOYSA-N
4-[[4-amino-5-bromo-6-(4-cyano-2,6-dimethylphenyloxy)-2-pyrimidinyl]-amino]benzonitrile
4-[[4-amino 5-bromo-6-(4-cyano-2,6-dimethylphenyloxy)-2-pyrimidinyl]amino]benzonitrile
4-[[4-amino-5-bromo-6-(4-cyano-2,6-dimethylphenyloxy)-2-pyrimidinyl]amino]benzonitrile
4-((6-amino-5-bromo-2-((4-cyanophenyl) amino) pyrimidin-4-yl) oxy)-3, 5-dimethylbenzonitrile
4-((6-amino-5-bromo-2-((4-cyanophenyl)amino)pyrimidin-4-yl)oxy)-3,5-dimethylbenzonitrile
etravirine (tmc125)
AB01566873_01
DTXSID30181412 ,
J-513179
AC-8503
sr-01000944895
SR-01000944895-1
HMS3651P20
SW219570-1
etravine; etravirine
FT-0668442
BCP03562
Q414762
gtpl12675
FT-0668443
benzonitrile,4-[[6-amino-5-bromo-2-[(4-cyanophenyl)amino]-4-pyrimidinyl]oxy]-3,5-dimethyl-
CCG-269074
NCGC00345885-04
A26965
EX-A4282
4-[[6-amino-5-bromo-2-[(4-cyanophenyl)amino]-4-pyrimidinyl]oxy]-3,5-dimethyl -benzonitrile
NCGC00345885-02
etravirine- bio-x
BA164437
etravirinum
etravirine (mart.)
dtxcid40103903
etravirina
j05ag04
EN300-23542752
SY097013

Research Excerpts

Overview

Etravirine (ETV) is a second-generation nonnucleoside reverse transcriptase (RT) inhibitor (NNRTI) introduced recently for salvage antiretroviral treatment after the emergence of NNRTI-resistant human immunodeficiency virus type 1 (HIV-1) Etravirin is a substrate and inducer of cytochrome P450 (CYP) 3A and an inhibitor of CYP2C9 and CYPC2C19.

ExcerptReferenceRelevance
"Etravirine (ETR) is a second-generation nonnucleoside reverse transcriptase (RT) inhibitor (NNRTI) active against common human immunodeficiency virus type 1 (HIV-1) drug-resistant strains. "( Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
Bar-Magen, T; Brenner, BG; Oliveira, M; Quan, Y; Schader, SM; Wainberg, MA; Xu, H, 2009
)
1.99
"Etravirine (ETV) is a second-generation nonnucleoside reverse transcriptase (RT) inhibitor (NNRTI) introduced recently for salvage antiretroviral treatment after the emergence of NNRTI-resistant human immunodeficiency virus type 1 (HIV-1). "( Combination of V106I and V179D polymorphic mutations in human immunodeficiency virus type 1 reverse transcriptase confers resistance to efavirenz and nevirapine but not etravirine.
Gatanaga, H; Hachiya, A; Hayashida, T; Ode, H; Oka, S; Sato, H, 2010
)
2
"Etravirine is a novel nonnucleoside reverse transcriptase inhibitor (NNRTI) for the treatment of HIV-1 infections. "( Interaction potential of etravirine with drug transporters assessed in vitro.
Haefeli, WE; Weiss, J; Zembruski, NC, 2011
)
2.12
"Etravirine is a second-generation non-nucleoside reverse transcriptase inhibitor (NNRTI) for the treatment of human immunodeficiency virus type 1 infection. "( Clinical Pharmacokinetics and Pharmacodynamics of Etravirine: An Updated Review.
Fletcher, CV; Havens, JP; Podany, AT; Scarsi, KK, 2020
)
2.25
"Etravirine is a substrate and inducer of cytochrome P450 (CYP) 3A and a substrate and inhibitor of CYP2C9 and CYPC2C19. "( Pharmacokinetic interaction between etravirine or darunavir/ritonavir and artemether/lumefantrine in healthy volunteers: a two-panel, two-way, two-period, randomized trial.
DeMasi, R; Kakuda, TN; Mohammed, P; van Delft, Y, 2013
)
2.11
"Etravirine (TMC125) is an orally administered second-generation non-nucleoside reverse transcriptase inhibitor (NNRTI) that is approved in treatment-experienced patients as addition to an optimized background therapy (OBT)."( Etravirine for the treatment of HIV/AIDS.
Schrijvers, R, 2013
)
3.28
"Etravirine is a well-tolerated NNRTI with a good safety profile and a higher genetic barrier for resistance compared to first-generation NNRTIs. "( Etravirine for the treatment of HIV/AIDS.
Schrijvers, R, 2013
)
3.28
"Etravirine is a second-generation non-nucleoside reverse transcriptase inhibitor (NNRTI) with reduced cross-resistance to first-generation NNRTIs. "( Prevalence and risk factors associated with resistance-associated mutations to etravirine in a cohort of perinatally HIV-infected children.
Bell, CS; Contreras, GA; Del Bianco, GP; Heresi, GP; Kleinosky, MT; Murphy, JR; Pérez, N, 2013
)
2.06
"Etravirine is an NNRTI option for treatment-experienced paediatric patients."( Etravirine in treatment-experienced, HIV-1-infected children and adolescents: 48-week safety, efficacy and resistance analysis of the phase II PIANO study.
Cahn, P; Chokephaibulkit, K; Dincq, S; Fourie, J; Kakuda, TN; Karatzios, C; Nijs, S; Opsomer, M; Tambuyzer, L; Tomaka, FL; Tudor-Williams, G, 2014
)
2.57
"Etravirine is a non-nucleoside reverse transcriptase inhibitor used in combination with other antiretrovirals for the treatment of HIV infection. "( Etravirine plasma exposure is associated with virological efficacy in treatment-experienced HIV-positive patients.
Aldieri, C; Bonora, S; Calcagno, A; Carbone, A; Castagna, A; D'Avolio, A; Di Perri, G; Ghisetti, V; Marinaro, L; Nozza, S; Trentalange, A, 2014
)
3.29
"Etravirine (TMC125) is a next-generation non-nucleoside reverse transcriptase inhibitor (NNRTI) that is being developed for the treatment of HIV-1 infections. "( Etravirine for the treatment of HIV infection.
Castagna, A; Lazzarin, A; Seminari, E, 2008
)
3.23
"Etravirine is a next-generation non-nucleoside reverse transcriptase inhibitor (NNRTI) that demonstrates potent in vitro activity against wild-type strains of HIV type 1 (HIV-1), as well as against numerous strains resistant to available NNRTIs. "( Etravirine.
Deeks, ED; Keating, GM, 2008
)
3.23
"Etravirine is an NNRTI that is able to adapt its binding orientation and overcome common NNRTI resistance associated mutations (RAMs) such as K103N. "( Etravirine: a second-generation nonnucleoside reverse transcriptase inhibitor (NNRTI) active against NNRTI-resistant strains of HIV.
Schiller, DS; Youssef-Bessler, M, 2009
)
3.24
"Etravirine is an NNRTI that was reported to be effective when used as part of an optimized, highly active antiretroviral therapy regimen in NNRTI treatment-experienced adult patients with HIV."( Etravirine: a second-generation nonnucleoside reverse transcriptase inhibitor (NNRTI) active against NNRTI-resistant strains of HIV.
Schiller, DS; Youssef-Bessler, M, 2009
)
3.24
"Etravirine is a next-generation non-nucleoside reverse transcriptase inhibitor (NNRTI) developed for the treatment of HIV-1 infection. "( Clinical pharmacokinetics and pharmacodynamics of etravirine.
De Smedt, G; Hoetelmans, RM; Kakuda, TN; Raoof, A; Schöller-Gyüre, M, 2009
)
2.05
"Etravirine (ETV) is a novel nonnucleoside reverse transcriptase inhibitor (NNRTI) with reduced cross-resistance to first-generation NNRTIs, which has been primarily studied in randomized clinical trials and not in routine clinical settings."( Prevalence of etravirine mutations and impact on response to treatment in routine clinical care: the Swiss HIV Cohort Study (SHCS).
Böni, J; Bucher, HC; Bürgisser, P; Günthard, HF; Hasse, B; Klimkait, T; Ledergerber, B; Scherrer, AU; von Wyl, V; Yerly, S, 2009
)
2.16
"Etravirine (ETR) is a second-generation nonnucleoside reverse transcriptase inhibitor (NNRTI) specifically designed for treatment-experienced patients infected with HIV-1. "( Etravirine for HIV-I: addressing the limitations of the nonnucleoside reverse transcriptase inhibitor class.
Grennan, T; Walmsley, S,
)
3.02
"Etravirine is a novel NNRTI, active against HIV-1 strains harboring multiple NNRTI mutations."( Validation of an electrospray ionization LC-MS/MS method for quantitative analysis of raltegravir, etravirine, and 9 other antiretroviral agents in human plasma samples.
Giocanti, M; Lacarelle, B; Paccou, A; Quaranta, S; Solas, C; Woloch, C, 2009
)
1.29
"Etravirine (ETR) is a diarylpyrimidine derivative with a polycyclic molecule composed of 3 aromatic rings with single bonds between the rings (C(20)H(15)BrN(60)). "( [Chemical characteristics, mechanism of action and antiviral activity of etravirine].
Estévez, MA; García, FG; Suay, VG, 2009
)
2.03
"Etravirine (ETR) is a non-nucleoside reverse transcriptase inhibitor (NNRTI) with a potent and broad in vitro spectrum of activity against HIV-1 and viruses with NNRTI resistances, allowing sequential use of drugs of this family. "( [Etravirine in first-line therapy].
Garcés, PA; Tena, EV, 2009
)
2.71
"Etravirine (ETR) is a new antiretroviral drug of the non-nucleoside reverse transcriptase inhibitor (NNRTI) family that has recently been approved by the regulatory agencies for the treatment of patients with prior experience with antiretrovirals, evidence of active viral replication, and who harbor multidrug resistant HIV-1 strains. "( [Role of etravirine in combination antiretroviral therapy].
Domingo, P, 2009
)
2.21
"Etravirine is a recently approved nonnucleoside reverse transcriptase inhibitor. "( Etravirine limits the emergence of darunavir and other protease inhibitor resistance-associated mutations in the DUET trials.
Azijn, H; De Meyer, S; Hill, A; Peeters, M; Picchio, G; Tambuyzer, L; Vingerhoets, J, 2010
)
3.25
"Etravirine is a non-nucleoside reverse transcriptase inhibitor (NNRTI) with activity against both wild-type HIV and viruses harboring NNRTI resistance. "( Effects of hepatic impairment on the steady-state pharmacokinetics of etravirine 200 mg BID: an open-label, multiple-dose, controlled Phase I study in adults.
Berckmans, C; De Smedt, G; Hoetelmans, RM; Kakuda, TN; Peeters, M; Schöller-Gyüre, M; Woodfall, B, 2010
)
2.04
"Etravirine is an effective and well-tolerated recently approved non-nucleoside reverse transcriptase inhibitor (NNRTI) for HIV type-1-infected patients with previous antiretroviral treatment experience. "( Clinical perspective on antiretroviral drug-drug interactions with the non-nucleoside reverse transcriptase inhibitor etravirine.
Hoetelmans, RM; Kakuda, TN; Schöller-Gyüre, M, 2010
)
2.01
"Etravirine is an enzyme inducer and could lower the concentration of combined drugs. "( Pharmacokinetics of etravirine, raltegravir and darunavir/ritonavir in treatment experienced patients.
Barrail-Tran, A; Bollens, D; Chêne, G; Colin, C; Descamps, D; Fagard, C; Goldwirt, L; Katlama, C; Molina, JM; Piketty, C; Taburet, AM; Yazdanpanah, Y, 2010
)
2.13
"Etravirine is a non-nucleoside reverse transcriptase inhibitor (NNRTI) that is active against NNRT-resistant HIV-1. "( LC-MS/MS determination of etravirine in rat plasma and its application in pharmacokinetic studies.
Abobo, CV; Bates, TR; John, J; Joseph, MK; Liang, D; Wu, L, 2010
)
2.1
"Etravirine (ETR) is a second-generation non-nucleoside reverse transcriptase inhibitor prescribed for the treatment of HIV-1. "( Biotransformation of the antiretroviral drug etravirine: metabolite identification, reaction phenotyping, and characterization of autoinduction of cytochrome P450-dependent metabolism.
Bumpus, NN; Yanakakis, LJ, 2012
)
2.08
"Etravirine (ETR) is a non-nucleoside analogue reverse transcriptase inhibitor (NNRTI) with a high genetic barrier to the development of resistance and with potential activity against Human immunodeficiency virus type 1 (HIV-1) strains resistant to first-generation NNRTIs. "( Prevalence of etravirine resistance associated mutations in HIV-1 strains isolated from infected individuals failing efavirenz: comparison between subtype B and non-B genetic variants.
Duque, V; Meliço-Silvestre, A; Morais, C; Mota, V; Pereira, B; Pereira-Vaz, J; Saraiva-da-Cunha, J, 2012
)
2.18
"Etravirine (Intelence®) is an orally administered next-generation non-nucleoside reverse transcriptase inhibitor (NNRTI). "( Etravirine: a review of its use in the management of treatment-experienced patients with HIV-1 infection.
Croxtall, JD, 2012
)
3.26
"Etravirine (ETR) is an expanded-spectrum nonnucleoside reverse transcriptase inhibitor (NNRTI) approved for use as an antiretroviral agent in treatment-experienced patients. "( Molecular mechanism of antagonism between the Y181C and E138K mutations in HIV-1 reverse transcriptase.
Asahchop, EL; Han, Y; McCallum, M; Oliveira, M; Quan, Y; Quashie, PK; Wainberg, MA; Xu, HT, 2012
)
1.82
"Etravirine [TMC 125] is a next-generation non-nucleoside reverse transcriptase inhibitor (NNRTI) that is being developed by Tibotec (Tibotec-Virco Group; now Johnson & Johnson) for the treatment of HIV-1 infections. "( Etravirine: R165335, TMC 125, TMC-125, TMC125.
, 2006
)
3.22
"Etravirine is a next generation nonnucleoside reverse transcriptase inhibitor with activity against nonnucleoside reverse transcriptase inhibitor resistant HIV-1 virus. "( Is there a role for etravirine in patients with Nonnucleoside reverse transcriptase inhibitor resistance?
Grover, D; Nelson, M; Scott, C, 2008
)
2.11
"Etravirine is a next-generation non-nucleoside reverse transcriptase inhibitor (NNRTI) with activity against wild-type and NNRTI-resistant HIV. "( A pharmacokinetic study of etravirine (TMC125) co-administered with ranitidine and omeprazole in HIV-negative volunteers.
Bouche, MP; De Smedt, G; Hoetelmans, RM; Kakuda, TN; Peeters, M; Schöller-Gyüre, M; Vanaken, H; Woodfall, B, 2008
)
2.09

Effects

Etravirine and darunavir have been used in HAART since 2010 in South Korea. It has the potential for interactions by inducing CYP3A and inhibiting CYP2C9 and 2C19. It is a mild inhibitor of P-glycoprotein but not a substrate.

ExcerptReferenceRelevance
"Etravirine has a satisfactory safety profile and, in this case, was a durable alternative therapy for HIV meningoencephalitis."( Efficacy of etravirine for treatment of acute HIV meningoencephalitis.
Couzigou, C; Escaut, L; Morand-Joubert, L; Roque-Afonso, AM; Seang, S; Vittecoq, D, 2009
)
1.45
"Etravirine (ETR) has demonstrated good tolerability in clinical trials and may be an option for patients who switch therapy due to adverse events."( Switching to an etravirine-containing regimen due to drug intolerance in clinical practice.
Martín-Carbonero, L; Moreno, V; Ramírez-Olivenza, G; Valencia, E,
)
1.2
"Etravirine and darunavir have been used in HAART since 2010 in South Korea. "( Drug susceptibility to etravirine and darunavir among Human Immunodeficiency Virus Type 1-derived pseudoviruses in treatment-experienced patients with HIV/AIDS in South Korea.
Choi, JY; Kee, MK; Kim, SS; Kwon, OK; Oh, HR; Park, M; Rhee, JE, 2015
)
2.17
"Etravirine has unique activity against most HIV isolates that are resistant to other NNRTIs. "( Etravirine: the renaissance of non-nucleoside reverse transcriptase inhibitors.
Castro, J; Espinoza, L; Jayaweera, DT, 2008
)
3.23
"Etravirine has a satisfactory safety profile and, in this case, was a durable alternative therapy for HIV meningoencephalitis."( Efficacy of etravirine for treatment of acute HIV meningoencephalitis.
Couzigou, C; Escaut, L; Morand-Joubert, L; Roque-Afonso, AM; Seang, S; Vittecoq, D, 2009
)
1.45
"Etravirine has the potential for interactions by inducing CYP3A and inhibiting CYP2C9 and 2C19; it is a mild inhibitor of P-glycoprotein but not a substrate."( Clinical pharmacokinetics and pharmacodynamics of etravirine.
De Smedt, G; Hoetelmans, RM; Kakuda, TN; Raoof, A; Schöller-Gyüre, M, 2009
)
1.33
"Etravirine (ETR) has demonstrated efficacy in patients with multiple prior treatments with prior virological failure and resistance mutations to various families of antiretroviral drugs. "( [Etravirine in highly treatment-experienced patients].
Corbera, EF; Palter, DP; Tiraboschi, JM, 2009
)
2.71
"Etravirine has compromised activity in approximately half of the patients failing nevirapine-based first-line treatment in this cohort, which supports guidelines that caution against using it with NRTIs alone in such patients."( Suboptimal etravirine activity is common during failure of nevirapine-based combination antiretroviral therapy in a cohort infected with non-B subtype HIV-1.
Adewole, I; Akanmu, S; Chaplin, B; Gashau, W; Idoko, J; Kanki, P; Meloni, S; Murphy, R; Penugonda, S; Taiwo, B, 2010
)
2.19
"Etravirine (ETR) has previously shown potent in vitro activity against different primary HIV-1 isolates and demonstrated durable efficacy in treatment-experienced, HIV-1-infected patients in the Phase III DUET studies. "( Short communication: activity of etravirine on different HIV type 1 subtypes: in vitro susceptibility in treatment-naive patients and week 48 pooled DUET study data.
Azijn, H; de Béthune, MP; De Meyer, S; De Smedt, G; Dierynck, I; Nijs, S; Picchio, G; Rimsky, L; Tambuyzer, L; Vingerhoets, J, 2010
)
2.08
"Etravirine has demonstrated a favorable safety and tolerability profile; the incidence of treatment-emergent adverse events was comparable with placebo in the DUET trials, with the exception of rash."( Etravirine in the treatment of HIV-1: a clinical overview for healthcare professionals.
De Smedt, G; Kakuda, TN; Mills, A; Peeters, M; Vingerhoets, J; Woodfall, B; Yeni, P, 2010
)
2.52
"Etravirine has been approved in several countries for use as part of highly active antiretroviral therapy in treatment-experienced patients."( Pharmacokinetic interactions between etravirine and non-antiretroviral drugs.
Hoetelmans, RM; Kakuda, TN; Schöller-Gyüre, M, 2011
)
1.36
"Etravirine has high affinity for plasma drug-binding proteins, such as albumin and α1-acid glycoprotein, which limits the amount of unbound etravirine available to enter the CNS. "( Etravirine in CSF is highly protein bound.
Best, BM; Capparelli, E; Clifford, D; Collier, AC; Croteau, D; Ellis, RJ; Gelman, B; Grant, I; Letendre, S; Marra, C; McArthur, J; McCutchan, JA; Morgello, S; Nguyen, A; Rossi, S; Simpson, D, 2013
)
3.28
"Etravirine (ETR) has shown a good lipid profile in previous studies. "( Lipid-lowering effect and efficacy after switching to etravirine in HIV-infected patients with intolerance to suppressive HAART.
Casado, JL; de Los Santos, I; Del Palacio, M; García-Fraile, L; Moreno, S; Pérez-Elías, MJ; Sanz, J,
)
1.82
"Etravirine has superseded dapivirine as Tibotec's lead NNRTI in clinical development worldwide."( Etravirine: R165335, TMC 125, TMC-125, TMC125.
, 2006
)
2.5

Actions

ExcerptReferenceRelevance
"Etravirine did not inhibit P-gp/ABCB1 and was not transported by the tested ABC transporters but was a potent inhibitor of BCRP/ABCG2."( Interaction potential of etravirine with drug transporters assessed in vitro.
Haefeli, WE; Weiss, J; Zembruski, NC, 2011
)
1.39

Toxicity

The aim of the study was to investigate the frequency and severity of adverse events (AEs) and laboratory abnormalities of interest over 96 weeks of treatment. Rash was the only adverse event to occur at a significantly higher incidence in the etravirine group.

ExcerptReferenceRelevance
" There were few hepatic adverse events, and rashes were predominantly early onset and mild to moderate in severity."( Efficacy and safety of etravirine (TMC125) in patients with highly resistant HIV-1: primary 24-week analysis.
Berger, DS; Blick, G; Cimoch, PJ; Cohen, CJ; Greenberg, RN; Hicks, CB; Hoetelmans, RM; Iveson, KJ; Jayaweera, DS; Mills, AM; Nadler, JP; Peeters, MP; Ruane, PJ; Schrader, SR; Shalit, P; Smith, SM; Steinhart, CR; Thompson, M; Vingerhoets, JH; Voorspoels, E; Ward, D; Woodfall, B, 2007
)
0.65
" Most adverse events were mild or moderate in severity."( Efficacy and safety of TMC125 (etravirine) in treatment-experienced HIV-1-infected patients in DUET-1: 24-week results from a randomised, double-blind, placebo-controlled trial.
Cahn, P; de Smedt, G; Grinsztejn, B; Haubrich, R; Lalezari, J; Leopold, L; Madruga, JV; Mills, A; Peeters, M; Pialoux, G; Trefiglio, R; Vingerhoets, J; Wilkin, T; Woodfall, B, 2007
)
0.63
" The type and frequency of adverse events were much the same in the two groups."( Efficacy and safety of TMC125 (etravirine) in treatment-experienced HIV-1-infected patients in DUET-2: 24-week results from a randomised, double-blind, placebo-controlled trial.
Baeten, B; Beets, G; Campbell, T; Clotet, B; de Smedt, G; Johnson, M; Katlama, C; Lazzarin, A; Moll, A; Peeters, M; Sinha, R; Towner, W; Trottier, B; Vingerhoets, J; Woodfall, B, 2007
)
0.63
"Neuropsychiatric adverse events (AEs) of interest occurred in 46."( Safety, tolerability, and preliminary efficacy of 48 weeks of etravirine therapy in a phase IIb dose-ranging study involving treatment-experienced patients with HIV-1 infection.
Clumeck, NN; Fätkenheuer, G; Gatell, J; Hay, P; Montaner, J; Peeters, MP; Schöller-Gyüre, M; Seminari, E; Simonts, M; Woodfall, B; Yeni, P, 2008
)
0.59
"ETR was generally safe and well tolerated during long-term administration in treatment-experienced, HIV-1-infected patients, and it had a safety profile comparable to that of placebo."( Safety, tolerability, and preliminary efficacy of 48 weeks of etravirine therapy in a phase IIb dose-ranging study involving treatment-experienced patients with HIV-1 infection.
Clumeck, NN; Fätkenheuer, G; Gatell, J; Hay, P; Montaner, J; Peeters, MP; Schöller-Gyüre, M; Seminari, E; Simonts, M; Woodfall, B; Yeni, P, 2008
)
0.59
" There is a complex, combined effect of HIV infection plus antiretroviral treatment on the incidence of gastrointestinal symptoms, and, for some trials, the majority of gastrointestinal adverse events may not be related to antiretroviral treatment."( Risk factors for gastrointestinal adverse events in HIV treated and untreated patients.
Balkin, A; Hill, A,
)
0.13
" Rash was the only adverse event to occur at a significantly higher incidence in the etravirine group (19 vs."( Efficacy and safety of etravirine in treatment-experienced, HIV-1 patients: pooled 48 week analysis of two randomized, controlled trials.
De Smedt, G; Haubrich, R; Katlama, C; Lalezari, J; Lazzarin, A; Madruga, JV; Molina, JM; Peeters, M; Picchio, G; Schechter, M; Vingerhoets, J; Woodfall, B, 2009
)
0.89
" The most frequent adverse effect is rash (affecting 19% of patients), which is usually mild (grades 1 or 2) and does not lead to drug withdrawal."( [Safety and tolerability of etravirine].
Portilla, J, 2009
)
0.65
" Furthermore, etravirine was shown to be safe and well-tolerated over this period."( Etravirine in combination with darunavir/ritonavir and optimized background regimen results in suppression of HIV replication in treatment-experienced patients. Evaluation of Katlama C, Haubrich R, Lalezari J, et al. Efficacy and safety of etravirine in t
Hull, MW; Montaner, JS, 2010
)
2.16
" The nature and magnitude of adverse events observed during treatment suggest that etravirine may offer improved tolerability over existing antiretrovirals, including NNRTIs."( A review of the safety and tolerability profile of the next-generation NNRTI etravirine.
De Smedt, G; Di Perri, G; Grinsztejn, B; Peeters, M; Towner, W; Woodfall, B, 2010
)
0.81
" Adverse events (AEs) and laboratory parameters were analysed."( Safety of etravirine in HIV-1/hepatitis B and/or C virus co-infected patients: pooled 96 week results from the Phase III DUET trials.
Clotet, B; Clumeck, N; Katlama, C; Nijs, S; Witek, J, 2010
)
0.76
"A randomized, double-blind trial in patients with viral suppression but ongoing CNS adverse events after more than 12 weeks EFV."( A phase IV, double-blind, multicentre, randomized, placebo-controlled, pilot study to assess the feasibility of switching individuals receiving efavirenz with continuing central nervous system adverse events to etravirine.
Fisher, M; Garvey, L; Hadley, W; Higgs, C; Mandalia, S; Nelson, M; Nicola, M; Perry, N; Waters, L; Winston, A, 2011
)
0.56
" Baseline CNS adverse events were similar."( A phase IV, double-blind, multicentre, randomized, placebo-controlled, pilot study to assess the feasibility of switching individuals receiving efavirenz with continuing central nervous system adverse events to etravirine.
Fisher, M; Garvey, L; Hadley, W; Higgs, C; Mandalia, S; Nelson, M; Nicola, M; Perry, N; Waters, L; Winston, A, 2011
)
0.56
"Switching EFV to ETR led to a significant reduction in overall G2-4 CNS adverse events, including insomnia, abnormal dreams and nervousness as individual adverse event."( A phase IV, double-blind, multicentre, randomized, placebo-controlled, pilot study to assess the feasibility of switching individuals receiving efavirenz with continuing central nervous system adverse events to etravirine.
Fisher, M; Garvey, L; Hadley, W; Higgs, C; Mandalia, S; Nelson, M; Nicola, M; Perry, N; Waters, L; Winston, A, 2011
)
0.56
"Although efavirenz is a universally recommended treatment for naive HIV-infected individuals, neuropsychiatric adverse events are common."( A comparison of neuropsychiatric adverse events during 12 weeks of treatment with etravirine and efavirenz in a treatment-naive, HIV-1-infected population.
Banhegyi, D; Gazzard, B; Hill, A; Marks, S; Nelson, M; Podzamczer, D; Stark, T; Stellbrink, HJ; van Delft, Y; Vingerhoets, J, 2011
)
0.59
" The primary end point was the percentage of patients with grade 1-4 drug-related treatment-emergent neuropsychiatric adverse events up to week 12."( A comparison of neuropsychiatric adverse events during 12 weeks of treatment with etravirine and efavirenz in a treatment-naive, HIV-1-infected population.
Banhegyi, D; Gazzard, B; Hill, A; Marks, S; Nelson, M; Podzamczer, D; Stark, T; Stellbrink, HJ; van Delft, Y; Vingerhoets, J, 2011
)
0.59
"After 12 weeks, first-line treatment with etravirine 400 mg once daily with two NRTIs was associated with significantly fewer neuropsychiatric adverse events when compared with efavirenz with two NRTIs."( A comparison of neuropsychiatric adverse events during 12 weeks of treatment with etravirine and efavirenz in a treatment-naive, HIV-1-infected population.
Banhegyi, D; Gazzard, B; Hill, A; Marks, S; Nelson, M; Podzamczer, D; Stark, T; Stellbrink, HJ; van Delft, Y; Vingerhoets, J, 2011
)
0.86
"0%) men discontinued ETR due to adverse events."( Efficacy and safety outcomes among treatment-experienced women and men treated with etravirine in gender, race and clinical experience.
Hodder, S; Jayaweera, D; Mrus, J; Ryan, R; Witek, J, 2012
)
0.6
"The aim of the study was to investigate the frequency and severity of adverse events (AEs) and laboratory abnormalities of interest over 96 weeks of treatment with etravirine or placebo in the pooled TMC125 DUET (Demonstrate Undetectable viral load in patients Experienced with ARV Therapy) trials."( Pooled week 96 results of the phase III DUET-1 and DUET-2 trials of etravirine: further analysis of adverse events and laboratory abnormalities of special interest.
Campbell, TB; Girard, PM; Grinsztejn, B; Hartikainen, J; Nijs, S; Rachline, A; Witek, J, 2012
)
0.81
" All treatments and combinations were well tolerated, with no grade 3 or 4 adverse events observed during treatment."( Pharmacokinetics and short-term safety of etravirine in combination with fluconazole or voriconazole in HIV-negative volunteers.
Aharchi, F; Hoetelmans, RM; Kakuda, TN; Nijs, S; Smedt, GD; Van Solingen-Ristea, R; Vyncke, V; Witek, J, 2013
)
0.65
" However, data on the long-term exposure to these therapeutic options are needed, and a handful of case reports are emerging, reporting rare but potentially life-threatening adverse hepatic events in patients with hepatitis co-infection or taking other hepatotoxic drugs."( Hepatoxicity of new antiretrovirals: a systematic review.
Lacombe, K; Surgers, L, 2013
)
0.39
" The pathogenic mechanisms involved are related to hypersensitivity, as described with nevirapine, impaired metabolism and therefore increased drug levels, and direct toxic effects with production of toxic metabolites."( Liver toxicity in HIV-infected patients receiving novel second-generation nonnucleoside reverse transcriptase inhibitors etravirine and rilpivirine.
Casado, JL,
)
0.34
" At week 48, the most common treatment-related grade ≥ 2 adverse event (AE) was rash (13%); 12% experienced grade 3 AEs."( Etravirine in treatment-experienced, HIV-1-infected children and adolescents: 48-week safety, efficacy and resistance analysis of the phase II PIANO study.
Cahn, P; Chokephaibulkit, K; Dincq, S; Fourie, J; Kakuda, TN; Karatzios, C; Nijs, S; Opsomer, M; Tambuyzer, L; Tomaka, FL; Tudor-Williams, G, 2014
)
1.85
" Lersivirine and etravirine were generally safe and well-tolerated."( Efficacy and safety of lersivirine versus etravirine for the treatment of HIV-1 infection in patients with prior non-nucleoside reverse transcriptase inhibitor (NNRTI) use and evidence of NNRTI resistance: a randomized phase 2B trial.
Craig, C; DeJesus, E; Fätkenheuer, G; Heera, J; Jones, J; Orrell, C; Tawadrous, M; Wang, C,
)
0.74
"3%) adverse events (AEs) were of mild or moderate severity with few grade 3/4 events, discontinuations, or temporary discontinuations/dose reductions due to AEs or serious AEs."( The maraviroc expanded access program - safety and efficacy data from an open-label study.
Craig, C; Heera, J; Lazzarin, A; Molina, JM; Mukwaya, G; Reynes, J; Sierra-Madero, JG; Valluri, S; van der Ryst, E,
)
0.13
"Our data suggest that etravirine is a safe option for HIV/HCV-coinfected patients, including those with significant liver fibrosis."( Liver toxicity and risk of discontinuation in HIV/hepatitis C virus-coinfected patients receiving an etravirine-containing antiretroviral regimen: influence of liver fibrosis.
Bañón, S; Casado, JL; Castro, A; Mena, A; Moreno, A; Moreno, S; Pedreira, J; Quereda, C, 2016
)
0.96
"8% of patients for adverse events in 23."( Long-term efficacy and safety of etravirine-containing regimens in a real-life cohort of treatment-experienced HIV-1-infected patients.
Allavena, C; Cabie, A; Cheret, A; Cotte, L; Delobel, P; Duvivier, C; Hoen, B; Katlama, C; Lahoulou, R; Poizot-Martin, I; Pugliese, P; Raffi, F; Roger, PM, 2016
)
0.72

Pharmacokinetics

The long terminal elimination half-life of etravirine should support once-daily dosing. The study showed highly effective virological and immunological responses over 24 weeks.

ExcerptReferenceRelevance
"To assess the pharmacokinetic profile, safety and virological response of two novel investigational antiretroviral agents when used in combination in HIV-1-infected subjects with multidrug-resistant virus."( Pharmacokinetics and antiretroviral response to darunavir/ritonavir and etravirine combination in patients with high-level viral resistance.
Boffito, M; Fletcher, C; Gazzard, B; Hoetelmans, R; Jackson, A; Miralles, D; Moyle, G; Nelson, M; Pozniak, A; Tolowinska, I; Winston, A, 2007
)
0.57
" Detailed pharmacokinetic assessments of darunavir and etravirine were determined on days 7 and 28."( Pharmacokinetics and antiretroviral response to darunavir/ritonavir and etravirine combination in patients with high-level viral resistance.
Boffito, M; Fletcher, C; Gazzard, B; Hoetelmans, R; Jackson, A; Miralles, D; Moyle, G; Nelson, M; Pozniak, A; Tolowinska, I; Winston, A, 2007
)
0.82
"This first study to assess the use of etravirine and darunavir in HIV-1-infected subjects with no treatment options showed highly effective virological and immunological responses over 24 weeks of therapy with no new safety concerns or unexpected pharmacokinetic interactions."( Pharmacokinetics and antiretroviral response to darunavir/ritonavir and etravirine combination in patients with high-level viral resistance.
Boffito, M; Fletcher, C; Gazzard, B; Hoetelmans, R; Jackson, A; Miralles, D; Moyle, G; Nelson, M; Pozniak, A; Tolowinska, I; Winston, A, 2007
)
0.84
" For TMC125 200 mg twice daily coadministered with DRV/r, AUC12h, Cmax and Cmin of TMC125 were 80%, 81% and 67% greater, respectively, versus TMC125 100 mg twice daily alone."( Pharmacokinetics of darunavir/ritonavir and TMC125 alone and coadministered in HIV-negative volunteers.
De Smedt, G; Hoetelmans, RM; Kakuda, TN; Lefebvre, E; Peeters, M; Schöller-Gyüre, M; Sekar, V; Woodfall, B, 2007
)
0.34
" Plasma concentrations and pharmacokinetic parameters of R- and S-methadone isomers were determined on days -1, 7, and 14 and of TMC125 on days 7 and 14."( Pharmacokinetic and pharmacodynamic study of the concomitant administration of methadone and TMC125 in HIV-negative volunteers.
De Smedt, G; Hoetelmans, RM; Kakuda, TN; Peeters, M; Schöller-Gyüre, M; Stevens, T; van den Brink, W; Vanaken, H; Vandermeulen, K; Woodfall, B, 2008
)
0.35
" Lack of pharmacokinetic alteration bounds for 90% confidence intervals (CI) about the geometric mean ratio (GMR; coadministration versus alone) were 70-143% for elvitegravir and ritonavir pharmacokinetics (maximum concentration [C(max)], concentration at the end of the dosing interval [C(tau)] and area under the plasma concentration-time curve [AUC(tau); 0-24 h] and 80-125% for etravirine pharmacokinetics (AUC(tau) 0-12 h)."( Pharmacokinetics of elvitegravir and etravirine following coadministration of ritonavir-boosted elvitegravir and etravirine.
Kakuda, TN; Kearney, BP; Mack, R; Ramanathan, S; West, S, 2008
)
0.79
" Elvitegravir pharmacokinetic GMR was 6-7% higher following elvitegravir/r plus etravirine dosing versus elvitegravir/r."( Pharmacokinetics of elvitegravir and etravirine following coadministration of ritonavir-boosted elvitegravir and etravirine.
Kakuda, TN; Kearney, BP; Mack, R; Ramanathan, S; West, S, 2008
)
0.85
"Two open-label, randomized, cross-over trials in healthy volunteers were conducted to investigate the pharmacokinetic interaction between etravirine and tenofovir disoproxil fumarate."( Assessment of the steady-state pharmacokinetic interaction between etravirine administered as two different formulations and tenofovir disoproxil fumarate in healthy volunteers.
Aharchi, F; Beets, G; De Smedt, G; Hoetelmans, RM; Kakuda, TN; Peeters, MP; Schöller-Gyüre, M; Vandermeulen, K; Woodfall, BJ, 2009
)
0.79
"Etravirine was administered as either 800 mg twice a day (bid) (phase II formulation in Study 1) or 200 mg bid (phase III formulation in Study 2) for 8 days followed by a 12 h pharmacokinetic evaluation."( Assessment of the steady-state pharmacokinetic interaction between etravirine administered as two different formulations and tenofovir disoproxil fumarate in healthy volunteers.
Aharchi, F; Beets, G; De Smedt, G; Hoetelmans, RM; Kakuda, TN; Peeters, MP; Schöller-Gyüre, M; Vandermeulen, K; Woodfall, BJ, 2009
)
2.03
" Etravirine 200 mg bid was coadministered on Day 1 to Day 15 of Cycle 3, with pharmacokinetic assessments of ethinylestradiol, norethindrone and etravirine on Day 15."( Effect of steady-state etravirine on the pharmacokinetics and pharmacodynamics of ethinylestradiol and norethindrone.
Aharchi, F; Hoetelmans, RM; Kakuda, TN; Peeters, M; Schöller-Gyüre, M; Vandermeulen, K; Woodfall, B, 2009
)
1.57
" Pharmacokinetic parameters were compared (before and after efavirenz 14-day intake) by determining geometric mean ratios and 90% confidence intervals."( Pharmacokinetics and safety of etravirine administered once or twice daily after 2 weeks treatment with efavirenz in healthy volunteers.
Asboe, D; Back, D; Boffito, M; Gazzard, B; Jackson, A; Lamorde, M; Pozniak, A; Taylor, J; Waters, L; Watson, V, 2009
)
0.64
"9%) to albumin and alpha(1)-acid glycoprotein and shows a relatively long elimination half-life of 30-40 hours."( Clinical pharmacokinetics and pharmacodynamics of etravirine.
De Smedt, G; Hoetelmans, RM; Kakuda, TN; Raoof, A; Schöller-Gyüre, M, 2009
)
0.61
"In this Phase I pharmacokinetic study, no clinically relevant differences were observed between patients with mild or moderate hepatic impairment and healthy matched subjects with regard to the pharmacokinetics of etravirine."( Effects of hepatic impairment on the steady-state pharmacokinetics of etravirine 200 mg BID: an open-label, multiple-dose, controlled Phase I study in adults.
Berckmans, C; De Smedt, G; Hoetelmans, RM; Kakuda, TN; Peeters, M; Schöller-Gyüre, M; Woodfall, B, 2010
)
0.78
" Drug concentrations were measured using a validated LC-MS/MS assay and pharmacokinetic analysis was performed using non-linear mixed effect modelling."( Intracellular and plasma steady-state pharmacokinetics of raltegravir, darunavir, etravirine and ritonavir in heavily pre-treated HIV-infected patients.
Beijnen, JH; Huitema, AD; Mulder, JW; Ter Heine, R; van Gorp, EC; Wagenaar, JF, 2010
)
0.59
"A pharmacokinetic trial was conducted to evaluate the potential for once-daily etravirine in antiretroviral regimens without and with darunavir/ritonavir."( Pharmacokinetics of once-daily etravirine without and with once-daily darunavir/ritonavir in antiretroviral-naive HIV type-1-infected adults.
DeJesus, E; Kakuda, TN; Lalezari, JP; Osiyemi, OO; Ruane, PJ; Ryan, R; Witek, J, 2010
)
0.87
"Of 23 enrolled patients (male 87%, Caucasian 39%), pharmacokinetic profiles for etravirine were available for 21 and 20 patients on day 14 and 28, respectively."( Pharmacokinetics of once-daily etravirine without and with once-daily darunavir/ritonavir in antiretroviral-naive HIV type-1-infected adults.
DeJesus, E; Kakuda, TN; Lalezari, JP; Osiyemi, OO; Ruane, PJ; Ryan, R; Witek, J, 2010
)
0.87
" Pharmacokinetic data were available in 577 patients randomized to receive etravirine."( Pharmacokinetics and pharmacodynamics of the non-nucleoside reverse-transcriptase inhibitor etravirine in treatment-experienced HIV-1-infected patients.
Corbett, C; De Smedt, G; Hoetelmans, RM; Kakuda, TN; Leopold, L; Nijs, S; Peeters, MP; Schöller-Gyüre, M; Snoeck, E; Vingerhoets, J; Vis, P; Wade, JR; Woodfall, BJ, 2010
)
0.81
" All pharmacokinetic parameters appeared to be highly variable regardless to the addition of etravirine."( Pharmacokinetics of etravirine, raltegravir and darunavir/ritonavir in treatment experienced patients.
Barrail-Tran, A; Bollens, D; Chêne, G; Colin, C; Descamps, D; Fagard, C; Goldwirt, L; Katlama, C; Molina, JM; Piketty, C; Taburet, AM; Yazdanpanah, Y, 2010
)
0.9
" The assay has been successfully used for pharmacokinetic evaluation of etravirine using the rat as an animal model."( LC-MS/MS determination of etravirine in rat plasma and its application in pharmacokinetic studies.
Abobo, CV; Bates, TR; John, J; Joseph, MK; Liang, D; Wu, L, 2010
)
0.89
" An extensive drug-drug interaction programme in HIV-negative subjects has been carried out to assess the potential for pharmacokinetic interactions between etravirine and a variety of non-antiretroviral drugs."( Pharmacokinetic interactions between etravirine and non-antiretroviral drugs.
Hoetelmans, RM; Kakuda, TN; Schöller-Gyüre, M, 2011
)
0.84
" Thus, a once-daily dosing regimen is supported not only by plasma etravirine pharmacokinetic profiles but also by intracellular levels."( Intracellular and plasma pharmacokinetics of 400 mg of etravirine once daily versus 200 mg of etravirine twice daily in HIV-infected patients.
Castillo-Ferrando, JR; Gutiérrez-Valencia, A; López-Cortés, LF; Martin-Peña, R; Ruiz-Valderas, R; Torres-Cornejo, A, 2012
)
0.86
"A pharmacokinetic (PK) study was performed in 12 HIV-negative men receiving 600 mg of darunavir, 100 mg of ritonavir, and 200 mg of etravirine orally, twice daily for 8 days."( Single- and multiple-dose pharmacokinetics of darunavir plus ritonavir and etravirine in semen and rectal tissue of HIV-negative men.
Asher Prince, HM; Brown, KC; Cohen, MS; Jennings, SH; Kashuba, AD; Malone, SA; Patterson, KB; Shaheen, NJ; Spacek, M, 2012
)
0.81
" To identify the potential impact of genetic and non-genetic factors involved in ETV metabolism, we carried out a two-step pharmacogenetics-based population pharmacokinetic study in HIV-1 infected individuals."( Pharmacogenetics-based population pharmacokinetic analysis of etravirine in HIV-1 infected individuals.
Arab-Alameddine, M; Bernasconi, E; Buclin, T; Calmy, A; Cavassini, M; Csajka, C; Decosterd, LA; di Iulio, J; Fayet-Mello, A; Furrer, H; Guidi, M; Günthard, HF; Lubomirov, R; Martinez, R; Marzolini, C; Rotger, M; Telenti, A, 2013
)
0.63
" A first step population pharmacokinetic model included non-genetic and known genetic factors (seven SNPs in CYP2C, one SNP in CYP3A5) as covariates."( Pharmacogenetics-based population pharmacokinetic analysis of etravirine in HIV-1 infected individuals.
Arab-Alameddine, M; Bernasconi, E; Buclin, T; Calmy, A; Cavassini, M; Csajka, C; Decosterd, LA; di Iulio, J; Fayet-Mello, A; Furrer, H; Guidi, M; Günthard, HF; Lubomirov, R; Martinez, R; Marzolini, C; Rotger, M; Telenti, A, 2013
)
0.63
" This single-center, open-label, two-period, single-sequence crossover study evaluated the effects of ETR coadministration on the pharmacokinetic profile of S/GSK1265744, an investigational integrase inhibitor in phase 2 studies."( Effects of etravirine on the pharmacokinetics of the integrase inhibitor S/GSK1265744.
Chen, S; Dumont, E; Ford, SL; Gould, E; Lou, Y; Piscitelli, S; Spreen, W, 2013
)
0.78
" Pharmacokinetic parameters were analysed using non-compartmental methods."( Effects of etravirine on the pharmacokinetics and pharmacodynamics of warfarin in rats.
Abobo, CV; Hsiao, C; John, J; John, M; Liang, D; Wu, L, 2013
)
0.78
" Pharmacokinetic interactions and safety of etravirine 200 mg twice daily coadministered with fluconazole 200 mg daily or voriconazole 200 mg twice daily, both inhibitors of CYP3A4, CYP2C9, and CYP2C19, were evaluated in an open-label, randomized, 3-period crossover trial in 18 HIV-negative volunteers."( Pharmacokinetics and short-term safety of etravirine in combination with fluconazole or voriconazole in HIV-negative volunteers.
Aharchi, F; Hoetelmans, RM; Kakuda, TN; Nijs, S; Smedt, GD; Van Solingen-Ristea, R; Vyncke, V; Witek, J, 2013
)
0.92
" The current study evaluated the pharmacokinetic interaction between etravirine and the lopinavir/ritonavir melt extrusion formulation."( Steady-state pharmacokinetics of etravirine and lopinavir/ritonavir melt extrusion formulation, alone and in combination, in healthy HIV-negative volunteers.
Akuma, SH; De Smedt, G; Hoetelmans, RM; Kakuda, TN; Schöller-Gyüre, M; Spittaels, K; Vyncke, V; Witek, J, 2013
)
0.91
" Steady-state pharmacokinetics were assessed for all antiretrovirals alone and coadministered; pharmacokinetic parameters were obtained by noncompartmental analysis."( Steady-state pharmacokinetics of etravirine and lopinavir/ritonavir melt extrusion formulation, alone and in combination, in healthy HIV-negative volunteers.
Akuma, SH; De Smedt, G; Hoetelmans, RM; Kakuda, TN; Schöller-Gyüre, M; Spittaels, K; Vyncke, V; Witek, J, 2013
)
0.67
" The objective was to investigate pharmacokinetic interactions between darunavir/ritonavir or etravirine and arthemether/lumefrantrine."( Pharmacokinetic interaction between etravirine or darunavir/ritonavir and artemether/lumefantrine in healthy volunteers: a two-panel, two-way, two-period, randomized trial.
DeMasi, R; Kakuda, TN; Mohammed, P; van Delft, Y, 2013
)
0.88
" The long terminal elimination half-life of etravirine should support once-daily dosing."( Pharmacokinetics and pharmacodynamics of etravirine 400 mg once daily in treatment-naïve patients.
Bickel, M; Di Perri, G; Faetkenheuer, G; Green, B; Hill, A; Kakuda, T; Kurowski, M; Morrish, G; van Delft, Y,
)
0.66
" Area under the curve over the dosing interval (AUC24h) and trough concentration (C0h) were estimated using a population pharmacokinetic model and compared with previous results using the 200-mg twice-daily dosage."( Pharmacokinetics and pharmacodynamics of etravirine 400 mg once daily in treatment-naïve patients.
Bickel, M; Di Perri, G; Faetkenheuer, G; Green, B; Hill, A; Kakuda, T; Kurowski, M; Morrish, G; van Delft, Y,
)
0.4
" Pharmacokinetic parameters were determined using non-compartmental analysis and least square means (LSM) ratios and 90% confidence intervals (CI) were calculated."( Single-dose pharmacokinetics of pediatric and adult formulations of etravirine and swallowability of the 200-mg tablet: results from three Phase 1 studies.
Berckmans, C; De Smedt, G; Hoetelmans, RM; Kakuda, TN; Leemans, R; Leopold, L; Nijs, S; Peeters, M; van Solingen-Ristea, R; Vyncke, V, 2013
)
0.63
" Full pharmacokinetic profiles of each drug and safety/tolerability were assessed."( Pharmacokinetic evaluation of the interaction between etravirine and rifabutin or clarithromycin in HIV-negative, healthy volunteers: results from two Phase 1 studies.
Aharchi, F; De Marez, T; Hoetelmans, RM; Kakuda, TN; Peeters, M; Vandermeulen, K; Woodfall, B, 2014
)
0.65
" Pharmacokinetic assessments were conducted for each drug at steady-state when given alone and when coadministered; statistical analyses were least-square means with 90% confidence intervals."( Pharmacokinetic interaction between etravirine or rilpivirine and telaprevir in healthy volunteers: A randomized, two-way crossover trial.
Bertelsen, KM; Crauwels, HM; Hoetelmans, RM; Kakuda, TN; Leopold, L; Nijs, S; Stevens, M; Tomaka, F; van Delft, Y; Vandevoorde, A; Witek, J, 2014
)
0.68
"Population pharmacokinetic (PopPK) analyses often rely on steady state and full adherence to prescribed dosage regimen assumptions from data gathered during therapeutic drug monitoring (TDM)."( Comparison of Population Pharmacokinetics Based on Steady-State Assumption Versus Electronically Monitored Adherence to Lopinavir, Atazanavir, Efavirenz, and Etravirine: A Retrospective Study.
Buclin, T; Bugnon, O; Cavassini, M; Csajka, C; Fuchs, A; Rotzinger, A; Schneider, MP, 2016
)
0.63
" At each time point, plasma concentrations were measured over 12 hours (12-hour time point was obtained before the second daily dose of etravirine); pharmacokinetic parameters were derived using noncompartmental analysis and were compared between pregnancy and postpartum using general linear models."( Pharmacokinetics of Total and Unbound Etravirine in HIV-1-Infected Pregnant Women.
Baugh, B; Brown, K; Crauwels, HM; Hillewaert, V; Osiyemi, O; Ramgopal, M; Ryan, R; Zorrilla, C, 2016
)
0.91
"Etravirine pharmacokinetic profiles were available for 13 of 15 enrolled women."( Pharmacokinetics of Total and Unbound Etravirine in HIV-1-Infected Pregnant Women.
Baugh, B; Brown, K; Crauwels, HM; Hillewaert, V; Osiyemi, O; Ramgopal, M; Ryan, R; Zorrilla, C, 2016
)
2.15
" 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
)
0.74
"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
)
2.18
" The pharmacokinetic profile of etravirine and clinical data support twice-daily dosing, although once-daily dosing has been investigated in treatment-naïve and treatment-experienced persons."( Clinical Pharmacokinetics and Pharmacodynamics of Etravirine: An Updated Review.
Fletcher, CV; Havens, JP; Podany, AT; Scarsi, KK, 2020
)
1.09

Compound-Compound Interactions

Echinacea purpurea can interact with etravirine, a nonnucleoside reverse transcriptase inhibitor of HIV. N348I decreases tenofovir susceptibility when combined with thymidine analogue mutations.

ExcerptReferenceRelevance
" Proton pump inhibitors and H(2)-antagonists are frequently used in the HIV-negative-infected population, and drug-drug interactions have been described with other antiretrovirals."( A pharmacokinetic study of etravirine (TMC125) co-administered with ranitidine and omeprazole in HIV-negative volunteers.
Bouche, MP; De Smedt, G; Hoetelmans, RM; Kakuda, TN; Peeters, M; Schöller-Gyüre, M; Vanaken, H; Woodfall, B, 2008
)
0.64
" However, N348I significantly decreases tenofovir susceptibility when combined with thymidine analogue mutations and etravirine susceptibility when combined with Y181C."( N348I in HIV-1 reverse transcriptase decreases susceptibility to tenofovir and etravirine in combination with other resistance mutations.
Moore, K; Radzio, J; Sluis-Cremer, N; Sonza, S; Tachedjian, G, 2010
)
0.8
" Interaction with fosamprenavir/ritonavir is not clinically significant, although their plasma levels vary slightly when used in combination with ETR."( [Etravirine drug interactions].
Pérez, VE; Sánchez-Parra, C; Serrano Villar, S, 2009
)
1.26
" Considering the importance of combining antiretrovirals for their optimal use in treating HIV, a number of drug-drug interactions with etravirine and other antiretrovirals have been evaluated."( Clinical perspective on antiretroviral drug-drug interactions with the non-nucleoside reverse transcriptase inhibitor etravirine.
Hoetelmans, RM; Kakuda, TN; Schöller-Gyüre, M, 2010
)
0.77
"The aim of this open-label, fixed-sequence study was to investigate the potential of the botanical supplement Echinacea purpurea to interact with etravirine, a nonnucleoside reverse transcriptase inhibitor of HIV."( Herb-drug interaction between Echinacea purpurea and etravirine in HIV-infected patients.
Cedeño, S; Clotet, B; Miranda, C; Moltó, J; Negredo, E; Valle, M, 2012
)
0.83
"We report the cases of two treatment-experienced HIV-infected patients with complex antiretroviral regimens that showed significant drug-drug interactions with etravirine."( Optimizing concentrations of concomitant antiretrovirals by reducing etravirine doses: two case reports of complex drug-drug interactions.
Cabot, JF; Denault, JS; Langlois, H; Marcotte, S; Sheehan, NL, 2019
)
0.95

Bioavailability

The antiviral drugs Raltegravir, Indinavir, Tipranavir, DolutegravIR, and Etravirine also exhibited good bioavailability and drug-likeness properties. The aim of this study was to develop a fast, effective, and material sparing screening method to design amorphous solid dispersions (ASDs) of etravirines.

ExcerptReferenceRelevance
" The Phase III formulation of etravirine significantly improves the bioavailability of etravirine over the Phase II formulation with reduced interpatient variability in etravirine pharmacokinetics."( Single- and multiple-dose pharmacokinetics of etravirine administered as two different formulations in HIV-1-infected patients.
Arasteh, K; Beets, G; De Smedt, G; Hoetelmans, RM; Kakuda, TN; Peeters, M; Pozniak, AL; Schöller-Gyüre, M; Vandermeulen, K; Woodfall, BJ; Workman, C, 2008
)
0.89
" Relative bioavailability of ritonavir was low, when compared with other ritonavir regimens."( Intracellular and plasma steady-state pharmacokinetics of raltegravir, darunavir, etravirine and ritonavir in heavily pre-treated HIV-infected patients.
Beijnen, JH; Huitema, AD; Mulder, JW; Ter Heine, R; van Gorp, EC; Wagenaar, JF, 2010
)
0.59
" While providing for a potentially increased oral bioavailability secondary to an increased drug dissolution rate, amorphous dispersions can be limited by their physical stability."( Physicochemical properties of the amorphous drug, cast films, and spray dried powders to predict formulation probability of success for solid dispersions: etravirine.
Brewster, ME; Craig, DQ; Davies, MC; De Cort, S; Leemans, R; Qi, S; Reading, M; Roberts, CJ; Segmuller, B; Stokbroekx, S; Turner, YT; Van Dycke, F; Voorspoels, J; Weuts, I; Xu, D, 2011
)
0.57
" We hypothesized that the presence of multiple drugs would reduce crystallization tendency, thereby providing stable, supersaturating formulations for bioavailability enhancement."( Multidrug, Anti-HIV Amorphous Solid Dispersions: Nature and Mechanisms of Impacts of Drugs on Each Other's Solution Concentrations.
Arca, HÇ; Dahal, D; Edgar, KJ; Mosquera-Giraldo, LI; Taylor, LS, 2017
)
0.46
"The aim of this study was to develop a fast, effective, and material sparing screening method to design amorphous solid dispersions (ASDs) of etravirine to drive more effectively the development process, leading to improved bioavailability (BA) and stability."( Five-Stage Approach for a Systematic Screening and Development of Etravirine Amorphous Solid Dispersions by Hot-Melt Extrusion.
Cadonau, S; Cardoso, S; Pereira, A; Pinto, RMA; Simões, MF; Simões, S; Werner, K, 2020
)
1
" The antiviral drugs Raltegravir, Indinavir, Tipranavir, Dolutegravir, and Etravirine also exhibited good bioavailability and drug-likeness properties."( Raltegravir, Indinavir, Tipranavir, Dolutegravir, and Etravirine against main protease and RNA-dependent RNA polymerase of SARS-CoV-2: A molecular docking and drug repurposing approach.
Al-Dhabi, NA; Arunagirinathan, N; Ignacimuthu, S; Indu, P; Rameshkumar, MR; Valan Arasu, M, 2020
)
1.04
"The inadequate bioavailability and toxicity potential of antiretroviral therapy limit their effectiveness in the complete eradication of HIV from viral reservoirs."( Multi-organ targeting of HIV-1 viral reservoirs with etravirine loaded nanostructured lipid carrier: An in-vivo proof of concept.
Fotooh Abadi, L; Kulkarni, S; Mahajan, K; Pai, R; Rojekar, S; Vavia, PR, 2021
)
0.87
"Many lead compounds fail to reach clinical trials despite being potent because of low bioavailability attributed to their insufficient solubility making solubility a primary and crucial factor in early phase drug discovery."( Structural modification aimed for improving solubility of lead compounds in early phase drug discovery.
Baidya, ATK; Das, B; Kumar, R; Mathew, AT; Yadav, AK, 2022
)
0.72

Dosage Studied

Etravirine (ETR) is a non-nucleoside reverse transcriptase inhibitor approved for 200 mg twice-daily dosing in conjunction with other antiretrovirals. Several drug interactions are expected and some may require dosage adjustment. Elvitegravir pharmacokinetic GMR was 6-7% higher following elvit.

ExcerptRelevanceReference
"Dietary dosing of the non-nucleoside reverse transcriptase inhibitor (NNRTI) TMC125, under development for treatment of HIV-1, resulted in a syndrome in male mice in a previous experiment that was termed hemorrhagic cardiomyopathy."( Hemorrhagic cardiomyopathy in male mice treated with an NNRTI: the role of vitamin K.
Bouche, MP; Coussement, W; De Jonghe, S; Lachau-Durand, S; Lammens, L; Starckx, S; Verbeeck, J; Vinken, P; Willems, B, 2008
)
0.35
"To evaluate the effect of 2 etravirine dosing regimens on QT/corrected QT interval (QTc) in HIV-negative volunteers and assess pharmacokinetic and additional safety parameters."( Etravirine has no effect on QT and corrected QT interval in HIV-negative volunteers.
De Smedt, G; Hoetelmans, RM; Janssen, K; Kakuda, TN; Lachaert, R; Peeters, M; Schöller-Gyüre, M; Woodfall, B, 2008
)
2.08
" Short-term dosing of etravirine in HIV-negative volunteers was generally safe and well tolerated."( Etravirine has no effect on QT and corrected QT interval in HIV-negative volunteers.
De Smedt, G; Hoetelmans, RM; Janssen, K; Kakuda, TN; Lachaert, R; Peeters, M; Schöller-Gyüre, M; Woodfall, B, 2008
)
2.1
"Administration of etravirine in a fasted state resulted in 51% lower mean exposure compared with dosing after a standard breakfast."( Effects of different meal compositions and fasted state on the oral bioavailability of etravirine.
Boffito, M; Hoetelmans, RM; Kakuda, TN; Leemans, R; Peeters, M; Pozniak, AL; Schöller-Gyüre, M; Vandermeulen, K; Vyncke, V; Woodfall, B, 2008
)
0.9
" Group 1 (n = 20) followed a sequence of 10-day dosing of elvitegravir/r (150/100 mg once daily) and elvitegravir/r plus etravirine (200 mg twice daily) or the reverse (n = 10 per sequence)."( Pharmacokinetics of elvitegravir and etravirine following coadministration of ritonavir-boosted elvitegravir and etravirine.
Kakuda, TN; Kearney, BP; Mack, R; Ramanathan, S; West, S, 2008
)
0.83
" Elvitegravir pharmacokinetic GMR was 6-7% higher following elvitegravir/r plus etravirine dosing versus elvitegravir/r."( Pharmacokinetics of elvitegravir and etravirine following coadministration of ritonavir-boosted elvitegravir and etravirine.
Kakuda, TN; Kearney, BP; Mack, R; Ramanathan, S; West, S, 2008
)
0.85
" The pharmacokinetics of etravirine in HIV-infected patients at the recommended dosage of 200 mg twice daily demonstrates moderate intersubject variability and no time dependency."( Clinical pharmacokinetics and pharmacodynamics of etravirine.
De Smedt, G; Hoetelmans, RM; Kakuda, TN; Raoof, A; Schöller-Gyüre, M, 2009
)
0.91
" Several drug interactions are expected with etravirine use, and some may require dosage adjustment or substitution of concurrent drugs."( Etravirine: A novel nonnucleoside reverse transcriptase inhibitor for managing human immunodeficiency virus infection.
Caldwell, DJ; Elsayed, RK, 2010
)
2.06
" During a 12 h dosing interval plasma and peripheral blood mononuclear cells were collected."( Intracellular and plasma steady-state pharmacokinetics of raltegravir, darunavir, etravirine and ritonavir in heavily pre-treated HIV-infected patients.
Beijnen, JH; Huitema, AD; Mulder, JW; Ter Heine, R; van Gorp, EC; Wagenaar, JF, 2010
)
0.59
" The ability to assess formulation risk in this regard early in development programs can not only help in guiding development strategies but can also point to critical design elements in the configuration of the dosage form."( Physicochemical properties of the amorphous drug, cast films, and spray dried powders to predict formulation probability of success for solid dispersions: etravirine.
Brewster, ME; Craig, DQ; Davies, MC; De Cort, S; Leemans, R; Qi, S; Reading, M; Roberts, CJ; Segmuller, B; Stokbroekx, S; Turner, YT; Van Dycke, F; Voorspoels, J; Weuts, I; Xu, D, 2011
)
0.57
"Addition of darunavir/ritonavir to etravirine, all dosed once daily, did not have a clinically significant effect on the pharmacokinetics of etravirine."( Pharmacokinetics of once-daily etravirine without and with once-daily darunavir/ritonavir in antiretroviral-naive HIV type-1-infected adults.
DeJesus, E; Kakuda, TN; Lalezari, JP; Osiyemi, OO; Ruane, PJ; Ryan, R; Witek, J, 2010
)
0.92
" However, ENF treatment is associated with injection site reactions and dosing fatigue."( A 48-week pilot study switching suppressed patients to darunavir/ritonavir and etravirine from enfuvirtide, protease inhibitor(s), and non-nucleoside reverse transcriptase inhibitor(s).
Alas, B; Perniciaro, A; Ruane, P; Ryan, R; Witek, J, 2010
)
0.59
"The need to adjust the dosage of raltegravir in HIV-infected patients who are also receiving etravirine is unclear."( Raltegravir dosage adjustment in HIV-infected patients receiving etravirine.
Do, VT; Fulco, PP; Higginson, RT, 2011
)
0.83
"74) were similar with both dosing schedules at the intracellular level."( Intracellular and plasma pharmacokinetics of 400 mg of etravirine once daily versus 200 mg of etravirine twice daily in HIV-infected patients.
Castillo-Ferrando, JR; Gutiérrez-Valencia, A; López-Cortés, LF; Martin-Peña, R; Ruiz-Valderas, R; Torres-Cornejo, A, 2012
)
0.63
"Our results show that intracellular etravirine levels were similar with both dosing regimens in patients with PI-sparing regimens, while etravirine plasma AUC(0-τ) and C(max) were 30% and 76% higher with the once-daily regimen, respectively."( Intracellular and plasma pharmacokinetics of 400 mg of etravirine once daily versus 200 mg of etravirine twice daily in HIV-infected patients.
Castillo-Ferrando, JR; Gutiérrez-Valencia, A; López-Cortés, LF; Martin-Peña, R; Ruiz-Valderas, R; Torres-Cornejo, A, 2012
)
0.9
" The use of SF and PK-PD disease models can be a valuable tool to predict dose-response of NMEs and support rational dose selection for monotherapy trials."( From in vitro EC₅₀ to in vivo dose-response for antiretrovirals using an HIV disease model. Part I: a framework.
Fang, J; Jadhav, PR, 2012
)
0.38
"44) for the concentration at the end of the dosing interval."( Herb-drug interaction between Echinacea purpurea and etravirine in HIV-infected patients.
Cedeño, S; Clotet, B; Miranda, C; Moltó, J; Negredo, E; Valle, M, 2012
)
0.63
"2 mg/kg twice daily (maximum dosage 200 mg twice daily) plus an optimized background therapy regimen achieved complete viral response (defined as HIV-1 RNA <50 copies/mL) in approximately half of treatment-experienced children and adolescents with HIV-1 infection and evidence of viral replication, and had an acceptable tolerability profile."( Etravirine: a guide to its use in treatment-experienced pediatric patients with HIV-1 infection in the US.
Lyseng-Williamson, KA, 2012
)
1.82
" Etravirine had no effects on S/GSK1265744 geometric mean ratios of the area under the curve from time zero until the end of the dosing interval (1."( Effects of etravirine on the pharmacokinetics of the integrase inhibitor S/GSK1265744.
Chen, S; Dumont, E; Ford, SL; Gould, E; Lou, Y; Piscitelli, S; Spreen, W, 2013
)
1.69
"Etravirine (ETR) is recommended as twice-daily dosing in pretreated patients."( Switch from etravirine twice daily to once daily in non-nucleoside reverse transcriptase inhibitor (NNRTI)-resistant HIV-infected patients with suppressed viremia: the Monetra study.
Agher, R; Assoumou, L; Blanc, C; Caby, F; Calvez, V; Courbon, E; Fourati, S; Katlama, C; Ktorza, N; Marcelin, AG; Peytavin, G; Schneider, L; Tindel, M,
)
1.95
" Once-daily dosing options are essential to treatment simplification strategies, which have been shown to enhance regimen compliance and durabiltiy."( New therapeutic landscape of NNRTIs for treatment of HIV: a look at recent data.
Dilanchian, P; Jayaweera, D, 2012
)
0.38
" Despite having a 30- to 40-h elimination half-life, etravirine was never considered for once-daily dosing during development due to an unacceptably high pill burden in phase II trials."( Does once-daily etravirine have a role in the management of HIV-1 infection?
McNicholl, IR, 2013
)
0.99
" Area under the curve over the dosing interval (AUC24h) and trough concentration (C0h) were estimated using a population pharmacokinetic model and compared with previous results using the 200-mg twice-daily dosage."( Pharmacokinetics and pharmacodynamics of etravirine 400 mg once daily in treatment-naïve patients.
Bickel, M; Di Perri, G; Faetkenheuer, G; Green, B; Hill, A; Kakuda, T; Kurowski, M; Morrish, G; van Delft, Y,
)
0.4
"In the SENSE trial, etravirine 400 mg once daily achieved similar exposures to historical reference data on etravirine when dosed at 200 mg twice daily."( Pharmacokinetics and pharmacodynamics of etravirine 400 mg once daily in treatment-naïve patients.
Bickel, M; Di Perri, G; Faetkenheuer, G; Green, B; Hill, A; Kakuda, T; Kurowski, M; Morrish, G; van Delft, Y,
)
0.72
"Two randomized studies in healthy adults investigated the single-dose pharmacokinetics of etravirine in various dosage strengths and the effects of dispersion in water and film-coating."( Single-dose pharmacokinetics of pediatric and adult formulations of etravirine and swallowability of the 200-mg tablet: results from three Phase 1 studies.
Berckmans, C; De Smedt, G; Hoetelmans, RM; Kakuda, TN; Leemans, R; Leopold, L; Nijs, S; Peeters, M; van Solingen-Ristea, R; Vyncke, V, 2013
)
0.85
" The complexity index was based on a score which takes into account the number of pills per day, the dosing schedule, the dosage form and any specific instructions linked to use of the drug."( Influence of adding etravirine on complexity index and patients' perceived complexity.
Almeida-González, CV; Calvo-Cidoncha, E; González-Bueno, J; Morillo-Verdugo, R, 2014
)
0.73
" The once-a-day ETV treatment was as effective as the twice daily dosing regimen."( Clinical and virological efficacy of etravirine plus two active Nucleos(t)ide analogs in an heterogeneous HIV-infected population.
de la Torre-Lima, J; Delgado-Fernández, M; Garcia-Lázaro, M; Girón-González, JA; López-Cortés, LF; López-Ruz, MA; Lozano, F; Márquez-Solero, M; Martinez-Perez, MA; Mohamed-Balghata, MO; Romero-Palacios, A; Téllez-Pérez, F; Viciana, P, 2014
)
0.68
"Etravirine (ETR), a non-nucleoside reverse transcriptase inhibitor approved for 200 mg twice-daily dosing in conjunction with other antiretrovirals (ARVs), has pharmacokinetic properties which support once-daily dosing."( Antiretroviral activity and safety of once-daily etravirine in treatment-naive HIV-infected adults: 48-week results.
Eron, JJ; Floris-Moore, MA; Francis, O; Johnson, MA; Kashuba, AD; Kronk, C; Mollan, K; Patterson, KB; Wilkin, AM; Wohl, DA, 2016
)
2.13
"Population pharmacokinetic (PopPK) analyses often rely on steady state and full adherence to prescribed dosage regimen assumptions from data gathered during therapeutic drug monitoring (TDM)."( Comparison of Population Pharmacokinetics Based on Steady-State Assumption Versus Electronically Monitored Adherence to Lopinavir, Atazanavir, Efavirenz, and Etravirine: A Retrospective Study.
Buclin, T; Bugnon, O; Cavassini, M; Csajka, C; Fuchs, A; Rotzinger, A; Schneider, MP, 2016
)
0.63
"Published PopPK models for lopinavir, atazanavir, efavirenz, and etravirine were applied to estimate PK parameters and individual concentrations in 140 HIV patients taking part in a medication adherence program using 2 dosing data sets."( Comparison of Population Pharmacokinetics Based on Steady-State Assumption Versus Electronically Monitored Adherence to Lopinavir, Atazanavir, Efavirenz, and Etravirine: A Retrospective Study.
Buclin, T; Bugnon, O; Cavassini, M; Csajka, C; Fuchs, A; Rotzinger, A; Schneider, MP, 2016
)
0.87
"Clearance estimates and likewise predicted concentrations did not markedly differ between the 2 dosing histories."( Comparison of Population Pharmacokinetics Based on Steady-State Assumption Versus Electronically Monitored Adherence to Lopinavir, Atazanavir, Efavirenz, and Etravirine: A Retrospective Study.
Buclin, T; Bugnon, O; Cavassini, M; Csajka, C; Fuchs, A; Rotzinger, A; Schneider, MP, 2016
)
0.63
"PopPK analysis assuming steady state with full adherence produced similar results to those based on detailed electronic dosing history reconciled with patients' allegations."( Comparison of Population Pharmacokinetics Based on Steady-State Assumption Versus Electronically Monitored Adherence to Lopinavir, Atazanavir, Efavirenz, and Etravirine: A Retrospective Study.
Buclin, T; Bugnon, O; Cavassini, M; Csajka, C; Fuchs, A; Rotzinger, A; Schneider, MP, 2016
)
0.63
" The CCR5-antagonist maraviroc (MVC), the non-nucleoside reverse transcriptase inhibitors (NNRTIs) etravirine (ETV) and rilpivirine (RPV), as well as the integrase strand transfer inhibitor (INSTI) raltegravir (RAL), have all been evaluated using both oral and non-oral dosing regimens, demonstrating a need for dynamic and sensitive bioanalytical tools for drug quantification in plasma and tissue."( Development and validation of a liquid chromatographic-tandem mass spectrometric method for the multiplexed quantification of etravirine, maraviroc, raltegravir, and rilpivirine in human plasma and tissue.
Marzinke, MA; Parsons, TL, 2016
)
0.86
" It is desirable for multidrug combinations to be coformulated into single dosage forms where possible, to promote patient convenience and adherence to dosage regimens, for which amorphous solid dispersion (ASD) is particularly well-suited."( Multidrug, Anti-HIV Amorphous Solid Dispersions: Nature and Mechanisms of Impacts of Drugs on Each Other's Solution Concentrations.
Arca, HÇ; Dahal, D; Edgar, KJ; Mosquera-Giraldo, LI; Taylor, LS, 2017
)
0.46
" The pharmacokinetic profile of etravirine and clinical data support twice-daily dosing, although once-daily dosing has been investigated in treatment-naïve and treatment-experienced persons."( Clinical Pharmacokinetics and Pharmacodynamics of Etravirine: An Updated Review.
Fletcher, CV; Havens, JP; Podany, AT; Scarsi, KK, 2020
)
1.09
"Solid dosage forms of amorphous solid dispersions (ASDs) have rarely been assessed for their crushability, although it might possibly be a more frequent practice than thought to facilitate oral administration in several clinical conditions (e."( The influence of crushing amorphous solid dispersion dosage forms on the in-vitro dissolution kinetics.
Appeltans, B; Pas, T; Van den Mooter, G; Verbert, S, 2020
)
0.56
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Roles (2)

RoleDescription
HIV-1 reverse transcriptase inhibitorAn entity which inhibits the activity of HIV-1 reverse transcriptase.
antiviral agentA substance that destroys or inhibits replication of viruses.
[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 (4)

ClassDescription
dinitrileA dinitrile is a compound containing two nitrile groups.
organobromine compoundA compound containing at least one carbon-bromine bond.
aminopyrimidineA member of the class of pyrimidines that is pyrimidine substituted by at least one amino group and its derivatives.
aromatic etherAny ether in which the oxygen is attached to at least one aryl substituent.
[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 (31)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Spike glycoproteinSevere acute respiratory syndrome-related coronavirusPotency35.48130.009610.525035.4813AID1479145
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Inhibition Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Chain A, Reverse transcriptase/ribonuclease HHIV-1 M:B_HXB2RIC50 (µMol)0.00210.00210.00210.0021AID977608
Chain B, p51 RTHIV-1 M:B_HXB2RIC50 (µMol)0.00210.00210.00210.0021AID977608
Chain A, Reverse transcriptase/ribonuclease HHIV-1 M:B_HXB2RIC50 (µMol)0.00210.00210.00210.0021AID977608
Chain B, p51 RTHIV-1 M:B_HXB2RIC50 (µMol)0.00210.00210.00210.0021AID977608
ATP-binding cassette sub-family C member 3Homo sapiens (human)IC50 (µMol)5.10000.63154.45319.3000AID586881
Cytochrome P450 1A2Homo sapiens (human)IC50 (µMol)0.02500.00011.774010.0000AID1561725
ATP-dependent translocase ABCB1Homo sapiens (human)IC50 (µMol)7.60000.00022.318510.0000AID586877
Cytochrome P450 3A4Homo sapiens (human)IC50 (µMol)0.01900.00011.753610.0000AID1561730
Cytochrome P450 2D6Homo sapiens (human)IC50 (µMol)0.03200.00002.015110.0000AID1561728
Cytochrome P450 2C9 Homo sapiens (human)IC50 (µMol)0.01700.00002.800510.0000AID1561726
Gag-Pol polyproteinHuman immunodeficiency virus type 1 (NEW YORK-5 ISOLATE)IC50 (µMol)0.37000.00020.10421.7000AID1801807
Potassium voltage-gated channel subfamily E member 1Homo sapiens (human)IC50 (µMol)1,258.93010.12004.048010.0000AID1207370
CruzipainTrypanosoma cruziIC50 (µMol)102.00000.00022.04508.0000AID484274; AID484275
Multidrug resistance-associated protein 1 Homo sapiens (human)IC50 (µMol)8.50000.00153.71109.6600AID586879
Glutamate receptor ionotropic, NMDA 1 Rattus norvegicus (Norway rat)IC50 (µMol)0.01000.00071.600310.0000AID1141979
Kappa-type opioid receptorCavia porcellus (domestic guinea pig)IC50 (µMol)0.09700.00030.71237.0700AID573468
Potassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)IC50 (µMol)1,258.93010.12004.048010.0000AID1207370
Glutamate receptor ionotropic, NMDA 2A Rattus norvegicus (Norway rat)IC50 (µMol)0.01000.00071.630610.0000AID1141979
Glutamate receptor ionotropic, NMDA 2BRattus norvegicus (Norway rat)IC50 (µMol)0.01000.00061.525710.0000AID1141979
Glutamate receptor ionotropic, NMDA 2CRattus norvegicus (Norway rat)IC50 (µMol)0.01000.00071.747210.0000AID1141979
Potassium voltage-gated channel subfamily H member 2Homo sapiens (human)IC50 (µMol)193.90500.00091.901410.0000AID1207464; AID1207495; AID1207524
Sodium channel protein type 5 subunit alphaHomo sapiens (human)IC50 (µMol)2,241.12850.00033.64849.2000AID1207310; AID1207341
Glutamate receptor ionotropic, NMDA 2DRattus norvegicus (Norway rat)IC50 (µMol)0.01000.00071.741110.0000AID1141979
Reverse transcriptase/RNaseH Human immunodeficiency virus 1IC50 (µMol)0.27110.00011.076810.0000AID1060625; AID1072797; AID1077216; AID1124998; AID1126511; AID1141975; AID1141976; AID1141978; AID1141979; AID1171593; AID1171594; AID1171595; AID1171596; AID1171597; AID1171598; AID1197835; AID1200848; AID1231487; AID1248227; AID1275539; AID1292014; AID1298247; AID1298248; AID1298249; AID1298250; AID1298251; AID1298253; AID1304407; AID1316359; AID1320869; AID1357809; AID1379964; AID1391088; AID1410405; AID1418468; AID1421298; AID1421299; AID1421300; AID1421301; AID1421302; AID1421310; AID1421311; AID1457063; AID1457064; AID1457065; AID1504705; AID1520066; AID1558858; AID1561723; AID1561724; AID1561725; AID1561726; AID1561727; AID1561728; AID1561729; AID1561730; AID1572075; AID1572530; AID1609110; AID1637387; AID1705194; AID1705195; AID1729163; AID1750722; AID1754648; AID1761021; AID1763907; AID1773463; AID1773843; AID1775803; AID1811038; AID1822283; AID1880369; AID1888706; AID517489; AID517490; AID517491; AID518734; AID518735; AID518737; AID518738; AID557019; AID557043; AID557044; AID573464; AID573465; AID573466; AID573468; AID573469; AID573470; AID648425; AID665276; AID698144; AID698145; AID698146; AID738336; AID779524
Glutamate receptor ionotropic, NMDA 3BRattus norvegicus (Norway rat)IC50 (µMol)0.01000.00071.741110.0000AID1141979
Canalicular multispecific organic anion transporter 1Homo sapiens (human)IC50 (µMol)7.80002.41006.343310.0000AID586880
Glutamate receptor ionotropic, NMDA 3ARattus norvegicus (Norway rat)IC50 (µMol)0.01000.00071.741110.0000AID1141979
Potassium voltage-gated channel subfamily D member 3Homo sapiens (human)IC50 (µMol)3,162.28001.40005.35009.3000AID1207430
Broad substrate specificity ATP-binding cassette transporter ABCG2Homo sapiens (human)IC50 (µMol)6.00000.00401.966610.0000AID586878
Reverse transcriptase Human immunodeficiency virus 1IC50 (µMol)0.01860.01000.53567.1300AID1815391; AID1815397; AID1815398; AID1815399; AID1815400; AID1815401; AID1815402
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Activation Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Reverse transcriptase/RNaseH Human immunodeficiency virus 1EC50 (µMol)0.01400.00040.61539.7000AID1326646; AID1326647; AID1410481; AID1410482; AID1410487; AID1421325; AID1456307; AID1456308; AID1483271; AID1483272; AID1483273; AID1483274; AID1483275; AID1483276; AID1483277; AID1750717; AID1750718; AID1750719; AID757627
Reverse transcriptase Human immunodeficiency virus 1EC50 (µMol)0.01210.00021.16839.0740AID1731746; AID1731747; AID1731749; AID1731750; AID1731751; AID1741393; AID1741394; AID1741395; AID1741396; AID1741397
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Other Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Reverse transcriptase/RNaseH Human immunodeficiency virus 1ID500.04550.00602.18989.0000AID1574384; AID1574385
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (183)

Processvia Protein(s)Taxonomy
xenobiotic metabolic processATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
bile acid and bile salt transportATP-binding cassette sub-family C member 3Homo sapiens (human)
glucuronoside transportATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transportATP-binding cassette sub-family C member 3Homo sapiens (human)
transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
leukotriene transportATP-binding cassette sub-family C member 3Homo sapiens (human)
monoatomic anion transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
transport across blood-brain barrierATP-binding cassette sub-family C member 3Homo sapiens (human)
steroid catabolic processCytochrome P450 1A2Homo sapiens (human)
porphyrin-containing compound metabolic processCytochrome P450 1A2Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 1A2Homo sapiens (human)
cholesterol metabolic processCytochrome P450 1A2Homo sapiens (human)
estrogen metabolic processCytochrome P450 1A2Homo sapiens (human)
toxin biosynthetic processCytochrome P450 1A2Homo sapiens (human)
post-embryonic developmentCytochrome P450 1A2Homo sapiens (human)
alkaloid metabolic processCytochrome P450 1A2Homo sapiens (human)
regulation of gene expressionCytochrome P450 1A2Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 1A2Homo sapiens (human)
dibenzo-p-dioxin metabolic processCytochrome P450 1A2Homo sapiens (human)
epoxygenase P450 pathwayCytochrome P450 1A2Homo sapiens (human)
lung developmentCytochrome P450 1A2Homo sapiens (human)
methylationCytochrome P450 1A2Homo sapiens (human)
monocarboxylic acid metabolic processCytochrome P450 1A2Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 1A2Homo sapiens (human)
retinol metabolic processCytochrome P450 1A2Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 1A2Homo sapiens (human)
cellular respirationCytochrome P450 1A2Homo sapiens (human)
aflatoxin metabolic processCytochrome P450 1A2Homo sapiens (human)
hydrogen peroxide biosynthetic processCytochrome P450 1A2Homo sapiens (human)
oxidative demethylationCytochrome P450 1A2Homo sapiens (human)
cellular response to cadmium ionCytochrome P450 1A2Homo sapiens (human)
omega-hydroxylase P450 pathwayCytochrome P450 1A2Homo sapiens (human)
G2/M transition of mitotic cell cycleATP-dependent translocase ABCB1Homo sapiens (human)
xenobiotic metabolic processATP-dependent translocase ABCB1Homo sapiens (human)
response to xenobiotic stimulusATP-dependent translocase ABCB1Homo sapiens (human)
phospholipid translocationATP-dependent translocase ABCB1Homo sapiens (human)
terpenoid transportATP-dependent translocase ABCB1Homo sapiens (human)
regulation of response to osmotic stressATP-dependent translocase ABCB1Homo sapiens (human)
transmembrane transportATP-dependent translocase ABCB1Homo sapiens (human)
transepithelial transportATP-dependent translocase ABCB1Homo sapiens (human)
stem cell proliferationATP-dependent translocase ABCB1Homo sapiens (human)
ceramide translocationATP-dependent translocase ABCB1Homo sapiens (human)
export across plasma membraneATP-dependent translocase ABCB1Homo sapiens (human)
transport across blood-brain barrierATP-dependent translocase ABCB1Homo sapiens (human)
positive regulation of anion channel activityATP-dependent translocase ABCB1Homo sapiens (human)
carboxylic acid transmembrane transportATP-dependent translocase ABCB1Homo sapiens (human)
xenobiotic detoxification by transmembrane export across the plasma membraneATP-dependent translocase ABCB1Homo sapiens (human)
xenobiotic transport across blood-brain barrierATP-dependent translocase ABCB1Homo sapiens (human)
regulation of chloride transportATP-dependent translocase ABCB1Homo sapiens (human)
lipid hydroxylationCytochrome P450 3A4Homo sapiens (human)
lipid metabolic processCytochrome P450 3A4Homo sapiens (human)
steroid catabolic processCytochrome P450 3A4Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 3A4Homo sapiens (human)
steroid metabolic processCytochrome P450 3A4Homo sapiens (human)
cholesterol metabolic processCytochrome P450 3A4Homo sapiens (human)
androgen metabolic processCytochrome P450 3A4Homo sapiens (human)
estrogen metabolic processCytochrome P450 3A4Homo sapiens (human)
alkaloid catabolic processCytochrome P450 3A4Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 3A4Homo sapiens (human)
calcitriol biosynthetic process from calciolCytochrome P450 3A4Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 3A4Homo sapiens (human)
vitamin D metabolic processCytochrome P450 3A4Homo sapiens (human)
vitamin D catabolic processCytochrome P450 3A4Homo sapiens (human)
retinol metabolic processCytochrome P450 3A4Homo sapiens (human)
retinoic acid metabolic processCytochrome P450 3A4Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 3A4Homo sapiens (human)
aflatoxin metabolic processCytochrome P450 3A4Homo sapiens (human)
oxidative demethylationCytochrome P450 3A4Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 2D6Homo sapiens (human)
steroid metabolic processCytochrome P450 2D6Homo sapiens (human)
cholesterol metabolic processCytochrome P450 2D6Homo sapiens (human)
estrogen metabolic processCytochrome P450 2D6Homo sapiens (human)
coumarin metabolic processCytochrome P450 2D6Homo sapiens (human)
alkaloid metabolic processCytochrome P450 2D6Homo sapiens (human)
alkaloid catabolic processCytochrome P450 2D6Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 2D6Homo sapiens (human)
isoquinoline alkaloid metabolic processCytochrome P450 2D6Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 2D6Homo sapiens (human)
retinol metabolic processCytochrome P450 2D6Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 2D6Homo sapiens (human)
negative regulation of bindingCytochrome P450 2D6Homo sapiens (human)
oxidative demethylationCytochrome P450 2D6Homo sapiens (human)
negative regulation of cellular organofluorine metabolic processCytochrome P450 2D6Homo sapiens (human)
arachidonic acid metabolic processCytochrome P450 2D6Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 2C9 Homo sapiens (human)
steroid metabolic processCytochrome P450 2C9 Homo sapiens (human)
cholesterol metabolic processCytochrome P450 2C9 Homo sapiens (human)
estrogen metabolic processCytochrome P450 2C9 Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 2C9 Homo sapiens (human)
epoxygenase P450 pathwayCytochrome P450 2C9 Homo sapiens (human)
urea metabolic processCytochrome P450 2C9 Homo sapiens (human)
monocarboxylic acid metabolic processCytochrome P450 2C9 Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 2C9 Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 2C9 Homo sapiens (human)
amide metabolic processCytochrome P450 2C9 Homo sapiens (human)
icosanoid biosynthetic processCytochrome P450 2C9 Homo sapiens (human)
oxidative demethylationCytochrome P450 2C9 Homo sapiens (human)
omega-hydroxylase P450 pathwayCytochrome P450 2C9 Homo sapiens (human)
epithelial cell maturationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
sensory perception of soundPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
male gonad developmentPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
vestibular nucleus developmentPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
secretory granule organizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cellular response to cAMPPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cellular response to acidic pHPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cellular response to light stimulusPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
potassium ion transmembrane transportPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cardiac muscle cell action potential involved in contractionPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cardiac muscle cell contractionPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane repolarizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane repolarization during action potentialPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
negative regulation of protein targeting to membranePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
positive regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
regulation of delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
negative regulation of delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
leukotriene metabolic processMultidrug resistance-associated protein 1 Homo sapiens (human)
xenobiotic metabolic processMultidrug resistance-associated protein 1 Homo sapiens (human)
response to xenobiotic stimulusMultidrug resistance-associated protein 1 Homo sapiens (human)
cobalamin transportMultidrug resistance-associated protein 1 Homo sapiens (human)
sphingolipid biosynthetic processMultidrug resistance-associated protein 1 Homo sapiens (human)
cellular response to oxidative stressMultidrug resistance-associated protein 1 Homo sapiens (human)
heme catabolic processMultidrug resistance-associated protein 1 Homo sapiens (human)
xenobiotic transportMultidrug resistance-associated protein 1 Homo sapiens (human)
phospholipid translocationMultidrug resistance-associated protein 1 Homo sapiens (human)
positive regulation of inflammatory responseMultidrug resistance-associated protein 1 Homo sapiens (human)
transmembrane transportMultidrug resistance-associated protein 1 Homo sapiens (human)
cell chemotaxisMultidrug resistance-associated protein 1 Homo sapiens (human)
transepithelial transportMultidrug resistance-associated protein 1 Homo sapiens (human)
cyclic nucleotide transportMultidrug resistance-associated protein 1 Homo sapiens (human)
leukotriene transportMultidrug resistance-associated protein 1 Homo sapiens (human)
monoatomic anion transmembrane transportMultidrug resistance-associated protein 1 Homo sapiens (human)
sphingolipid translocationMultidrug resistance-associated protein 1 Homo sapiens (human)
export across plasma membraneMultidrug resistance-associated protein 1 Homo sapiens (human)
transport across blood-brain barrierMultidrug resistance-associated protein 1 Homo sapiens (human)
cellular response to amyloid-betaMultidrug resistance-associated protein 1 Homo sapiens (human)
carboxylic acid transmembrane transportMultidrug resistance-associated protein 1 Homo sapiens (human)
xenobiotic transport across blood-brain barrierMultidrug resistance-associated protein 1 Homo sapiens (human)
glutathione transmembrane transportMultidrug resistance-associated protein 1 Homo sapiens (human)
gastrin-induced gastric acid secretionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
glucose metabolic processPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
heart developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
sensory perception of soundPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
rhythmic behaviorPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of heart contractionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of blood pressurePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
positive regulation of heart ratePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
iodide transportPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
erythrocyte differentiationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
intracellular chloride ion homeostasisPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
response to insulinPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
social behaviorPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
corticosterone secretionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
inner ear morphogenesisPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
inner ear developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
intestinal absorptionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
detection of mechanical stimulus involved in sensory perception of soundPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
potassium ion homeostasisPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cardiac muscle contractionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
auditory receptor cell developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of membrane repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of atrial cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
positive regulation of cardiac muscle contractionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of gastric acid secretionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
stomach developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
renal absorptionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
renal sodium ion absorptionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cellular response to cAMPPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cellular response to xenobiotic stimulusPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
potassium ion transmembrane transportPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cellular response to epinephrine stimulusPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
adrenergic receptor signaling pathwayPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cardiac muscle cell contractionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane repolarization during action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
atrial cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cochlea developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane repolarization during atrial cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
positive regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
non-motile cilium assemblyPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
potassium ion import across plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of heart rate by hormonePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of membrane potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
positive regulation of DNA-templated transcriptionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion homeostasisPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cardiac muscle contractionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cellular response to xenobiotic stimulusPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane depolarization during action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
negative regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
positive regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
negative regulation of potassium ion export across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion import across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of heart rateSodium channel protein type 5 subunit alphaHomo sapiens (human)
cardiac conduction system developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
cardiac ventricle developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
brainstem developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
sodium ion transportSodium channel protein type 5 subunit alphaHomo sapiens (human)
positive regulation of sodium ion transportSodium channel protein type 5 subunit alphaHomo sapiens (human)
response to denervation involved in regulation of muscle adaptationSodium channel protein type 5 subunit alphaHomo sapiens (human)
telencephalon developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
cerebellum developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
sodium ion transmembrane transportSodium channel protein type 5 subunit alphaHomo sapiens (human)
odontogenesis of dentin-containing toothSodium channel protein type 5 subunit alphaHomo sapiens (human)
positive regulation of action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
positive regulation of epithelial cell proliferationSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
cardiac muscle contractionSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of atrial cardiac muscle cell membrane depolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of atrial cardiac muscle cell membrane repolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of ventricular cardiac muscle cell membrane depolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
cellular response to calcium ionSodium channel protein type 5 subunit alphaHomo sapiens (human)
cardiac muscle cell action potential involved in contractionSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of cardiac muscle cell contractionSodium channel protein type 5 subunit alphaHomo sapiens (human)
ventricular cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
atrial cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
SA node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
AV node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
bundle of His cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during AV node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during SA node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during Purkinje myocyte cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during bundle of His cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
AV node cell to bundle of His cell communicationSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of heart rate by cardiac conductionSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during atrial cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of sodium ion transmembrane transportSodium channel protein type 5 subunit alphaHomo sapiens (human)
xenobiotic metabolic processCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
negative regulation of gene expressionCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bile acid and bile salt transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bilirubin transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
heme catabolic processCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic export from cellCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transepithelial transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
leukotriene transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
monoatomic anion transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transport across blood-brain barrierCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transport across blood-brain barrierCanalicular multispecific organic anion transporter 1Homo sapiens (human)
potassium ion transportPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
protein homooligomerizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
membrane repolarizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
action potentialPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
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 (95)

Processvia Protein(s)Taxonomy
ATP bindingATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type xenobiotic transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
glucuronoside transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type bile acid transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATP hydrolysis activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATPase-coupled transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
icosanoid transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
monooxygenase activityCytochrome P450 1A2Homo sapiens (human)
iron ion bindingCytochrome P450 1A2Homo sapiens (human)
protein bindingCytochrome P450 1A2Homo sapiens (human)
electron transfer activityCytochrome P450 1A2Homo sapiens (human)
oxidoreductase activityCytochrome P450 1A2Homo sapiens (human)
oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen, reduced flavin or flavoprotein as one donor, and incorporation of one atom of oxygenCytochrome P450 1A2Homo sapiens (human)
enzyme bindingCytochrome P450 1A2Homo sapiens (human)
heme bindingCytochrome P450 1A2Homo sapiens (human)
demethylase activityCytochrome P450 1A2Homo sapiens (human)
caffeine oxidase activityCytochrome P450 1A2Homo sapiens (human)
aromatase activityCytochrome P450 1A2Homo sapiens (human)
estrogen 16-alpha-hydroxylase activityCytochrome P450 1A2Homo sapiens (human)
estrogen 2-hydroxylase activityCytochrome P450 1A2Homo sapiens (human)
hydroperoxy icosatetraenoate dehydratase activityCytochrome P450 1A2Homo sapiens (human)
protein bindingATP-dependent translocase ABCB1Homo sapiens (human)
ATP bindingATP-dependent translocase ABCB1Homo sapiens (human)
ABC-type xenobiotic transporter activityATP-dependent translocase ABCB1Homo sapiens (human)
efflux transmembrane transporter activityATP-dependent translocase ABCB1Homo sapiens (human)
ATP hydrolysis activityATP-dependent translocase ABCB1Homo sapiens (human)
transmembrane transporter activityATP-dependent translocase ABCB1Homo sapiens (human)
ubiquitin protein ligase bindingATP-dependent translocase ABCB1Homo sapiens (human)
ATPase-coupled transmembrane transporter activityATP-dependent translocase ABCB1Homo sapiens (human)
xenobiotic transmembrane transporter activityATP-dependent translocase ABCB1Homo sapiens (human)
carboxylic acid transmembrane transporter activityATP-dependent translocase ABCB1Homo sapiens (human)
phosphatidylcholine floppase activityATP-dependent translocase ABCB1Homo sapiens (human)
phosphatidylethanolamine flippase activityATP-dependent translocase ABCB1Homo sapiens (human)
ceramide floppase activityATP-dependent translocase ABCB1Homo sapiens (human)
floppase activityATP-dependent translocase ABCB1Homo sapiens (human)
monooxygenase activityCytochrome P450 3A4Homo sapiens (human)
steroid bindingCytochrome P450 3A4Homo sapiens (human)
iron ion bindingCytochrome P450 3A4Homo sapiens (human)
protein bindingCytochrome P450 3A4Homo sapiens (human)
steroid hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
retinoic acid 4-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
oxidoreductase activityCytochrome P450 3A4Homo sapiens (human)
oxygen bindingCytochrome P450 3A4Homo sapiens (human)
enzyme bindingCytochrome P450 3A4Homo sapiens (human)
heme bindingCytochrome P450 3A4Homo sapiens (human)
vitamin D3 25-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
caffeine oxidase activityCytochrome P450 3A4Homo sapiens (human)
quinine 3-monooxygenase activityCytochrome P450 3A4Homo sapiens (human)
testosterone 6-beta-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
1-alpha,25-dihydroxyvitamin D3 23-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
anandamide 8,9 epoxidase activityCytochrome P450 3A4Homo sapiens (human)
anandamide 11,12 epoxidase activityCytochrome P450 3A4Homo sapiens (human)
anandamide 14,15 epoxidase activityCytochrome P450 3A4Homo sapiens (human)
aromatase activityCytochrome P450 3A4Homo sapiens (human)
vitamin D 24-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
estrogen 16-alpha-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
estrogen 2-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
1,8-cineole 2-exo-monooxygenase activityCytochrome P450 3A4Homo sapiens (human)
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)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
potassium channel regulator activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
telethonin bindingPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
protein-containing complex bindingPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
transmembrane transporter bindingPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
ATP bindingMultidrug resistance-associated protein 1 Homo sapiens (human)
ABC-type vitamin B12 transporter activityMultidrug resistance-associated protein 1 Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityMultidrug resistance-associated protein 1 Homo sapiens (human)
efflux transmembrane transporter activityMultidrug resistance-associated protein 1 Homo sapiens (human)
ATP hydrolysis activityMultidrug resistance-associated protein 1 Homo sapiens (human)
ATPase-coupled lipid transmembrane transporter activityMultidrug resistance-associated protein 1 Homo sapiens (human)
glutathione transmembrane transporter activityMultidrug resistance-associated protein 1 Homo sapiens (human)
ATPase-coupled transmembrane transporter activityMultidrug resistance-associated protein 1 Homo sapiens (human)
xenobiotic transmembrane transporter activityMultidrug resistance-associated protein 1 Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityMultidrug resistance-associated protein 1 Homo sapiens (human)
sphingolipid transporter activityMultidrug resistance-associated protein 1 Homo sapiens (human)
carboxylic acid transmembrane transporter activityMultidrug resistance-associated protein 1 Homo sapiens (human)
ABC-type transporter activityMultidrug resistance-associated protein 1 Homo sapiens (human)
ABC-type xenobiotic transporter activityMultidrug resistance-associated protein 1 Homo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
calmodulin bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
phosphatidylinositol-4,5-bisphosphate bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
protein phosphatase 1 bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
outward rectifier potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
protein kinase A catalytic subunit bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
protein kinase A regulatory subunit bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
transmembrane transporter bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in atrial cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
scaffold protein bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
transcription cis-regulatory region bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
inward rectifier potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
ubiquitin protein ligase bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
identical protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
protein homodimerization activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
C3HC4-type RING finger domain bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
scaffold protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated sodium channel activitySodium channel protein type 5 subunit alphaHomo sapiens (human)
protein bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
calmodulin bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
fibroblast growth factor bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
enzyme bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
protein kinase bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
protein domain specific bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
ankyrin bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
ubiquitin protein ligase bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
transmembrane transporter bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
nitric-oxide synthase bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in AV node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in bundle of His cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in Purkinje myocyte action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in SA node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
scaffold protein bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
protein bindingCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATP bindingCanalicular multispecific organic anion transporter 1Homo sapiens (human)
organic anion transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type xenobiotic transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bilirubin transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATP hydrolysis activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATPase-coupled transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
A-type (transient outward) potassium channel activityPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
transmembrane transporter bindingPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
metal ion bindingPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
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 (45)

Processvia Protein(s)Taxonomy
plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
basal plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
basolateral plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 1A2Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 1A2Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 1A2Homo sapiens (human)
cytoplasmATP-dependent translocase ABCB1Homo sapiens (human)
plasma membraneATP-dependent translocase ABCB1Homo sapiens (human)
cell surfaceATP-dependent translocase ABCB1Homo sapiens (human)
membraneATP-dependent translocase ABCB1Homo sapiens (human)
apical plasma membraneATP-dependent translocase ABCB1Homo sapiens (human)
extracellular exosomeATP-dependent translocase ABCB1Homo sapiens (human)
external side of apical plasma membraneATP-dependent translocase ABCB1Homo sapiens (human)
plasma membraneATP-dependent translocase ABCB1Homo sapiens (human)
cytoplasmCytochrome P450 3A4Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 3A4Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 3A4Homo sapiens (human)
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)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
lysosomePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cell surfacePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
apical plasma membranePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
Z discPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane raftPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
plasma membraneMultidrug resistance-associated protein 1 Homo sapiens (human)
basal plasma membraneMultidrug resistance-associated protein 1 Homo sapiens (human)
membraneMultidrug resistance-associated protein 1 Homo sapiens (human)
basolateral plasma membraneMultidrug resistance-associated protein 1 Homo sapiens (human)
apical plasma membraneMultidrug resistance-associated protein 1 Homo sapiens (human)
lateral plasma membraneMultidrug resistance-associated protein 1 Homo sapiens (human)
extracellular exosomeMultidrug resistance-associated protein 1 Homo sapiens (human)
basolateral plasma membraneMultidrug resistance-associated protein 1 Homo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 1 Rattus norvegicus (Norway rat)
plasma membraneGlutamate receptor ionotropic, NMDA 1 Rattus norvegicus (Norway rat)
early endosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
endoplasmic reticulumPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cytoplasmPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
lysosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
early endosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
late endosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
endoplasmic reticulumPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
basolateral plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
apical plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
transport vesiclePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cytoplasmic vesicle membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
neuron projectionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
neuronal cell bodyPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane raftPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
ciliary basePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
lumenal side of membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
basolateral part of cellPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
monoatomic ion channel complexPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
virion membraneSpike glycoproteinSevere acute respiratory syndrome-related coronavirus
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 2A Rattus norvegicus (Norway rat)
plasma membraneGlutamate receptor ionotropic, NMDA 2A Rattus norvegicus (Norway rat)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 2BRattus norvegicus (Norway rat)
plasma membraneGlutamate receptor ionotropic, NMDA 2BRattus norvegicus (Norway rat)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 2CRattus norvegicus (Norway rat)
plasma membraneGlutamate receptor ionotropic, NMDA 2CRattus norvegicus (Norway rat)
plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cell surfacePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
perinuclear region of cytoplasmPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
inward rectifier potassium channel complexPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
caveolaSodium channel protein type 5 subunit alphaHomo sapiens (human)
nucleoplasmSodium channel protein type 5 subunit alphaHomo sapiens (human)
nucleolusSodium channel protein type 5 subunit alphaHomo sapiens (human)
endoplasmic reticulumSodium channel protein type 5 subunit alphaHomo sapiens (human)
plasma membraneSodium channel protein type 5 subunit alphaHomo sapiens (human)
caveolaSodium channel protein type 5 subunit alphaHomo sapiens (human)
cell surfaceSodium channel protein type 5 subunit alphaHomo sapiens (human)
intercalated discSodium channel protein type 5 subunit alphaHomo sapiens (human)
membraneSodium channel protein type 5 subunit alphaHomo sapiens (human)
lateral plasma membraneSodium channel protein type 5 subunit alphaHomo sapiens (human)
Z discSodium channel protein type 5 subunit alphaHomo sapiens (human)
T-tubuleSodium channel protein type 5 subunit alphaHomo sapiens (human)
sarcolemmaSodium channel protein type 5 subunit alphaHomo sapiens (human)
perinuclear region of cytoplasmSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel complexSodium channel protein type 5 subunit alphaHomo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 2DRattus norvegicus (Norway rat)
plasma membraneGlutamate receptor ionotropic, NMDA 2DRattus norvegicus (Norway rat)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 3BRattus norvegicus (Norway rat)
plasma membraneGlutamate receptor ionotropic, NMDA 3BRattus norvegicus (Norway rat)
plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
cell surfaceCanalicular multispecific organic anion transporter 1Homo sapiens (human)
apical plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
intercellular canaliculusCanalicular multispecific organic anion transporter 1Homo sapiens (human)
apical plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 3ARattus norvegicus (Norway rat)
plasma membraneGlutamate receptor ionotropic, NMDA 3ARattus norvegicus (Norway rat)
plasma membranePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
sarcolemmaPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
GABA-ergic synapsePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
postsynaptic specialization membranePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
dendritic spinePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
neuronal cell bodyPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
postsynaptic membranePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
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 (1343)

Assay IDTitleYearJournalArticle
AID977608Experimentally measured binding affinity data (IC50) for protein-ligand complexes derived from PDB2010Bioorganic & medicinal chemistry letters, Jul-15, Volume: 20, Issue:14
Discovery of piperidin-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-benzyl derivatives with broad potency against resistant mutant viruses.
AID1775803Inhibition of recombinant wild type HIV1 reverse transcriptase assessed as inhibition of biotin-dUTP incorporation into template2021Journal of medicinal chemistry, 04-08, Volume: 64, Issue:7
2,4,5-Trisubstituted Pyrimidines as Potent HIV-1 NNRTIs: Rational Design, Synthesis, Activity Evaluation, and Crystallographic Studies.
AID1435517Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 expressing reverse transcriptase F227L/V106A double mutant
AID541252Antiviral activity against multidrug resistant HIV1 A17 containing reverse transcripatseK103N and Y181C mutation infected in human MT2 cells assessed as reduction in p24 antigen level after 4 days by ELISA2010Journal of medicinal chemistry, Dec-09, Volume: 53, Issue:23
Design, synthesis, and evaluation of diarylpyridines and diarylanilines as potent non-nucleoside HIV-1 reverse transcriptase inhibitors.
AID347633Antiviral activity against NNRTI-resistant HIV HXB2 isolate 32 with reverse transcriptase V106, V179, Y181, G190 mutation in human MT4 cells assessed as protection against virus-induced cell death after 5 days by MTT assay2008Journal of medicinal chemistry, Dec-11, Volume: 51, Issue:23
Design of annulated pyrazoles as inhibitors of HIV-1 reverse transcriptase.
AID1693808Antiviral activity against HIV-3B harboring reverse transcriptase Y181C mutant infected in human MT-4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2021Bioorganic & medicinal chemistry, 01-15, Volume: 30Novel indolylarylsulfone derivatives as covalent HIV-1 reverse transcriptase inhibitors specifically targeting the drug-resistant mutant Y181C.
AID508828Antiviral activity against Human immunodeficiency virus 1 subtype A isolate 92RW020 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1729153Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV HIV1 RES056 harboring K103N/Y181C double mutant infected in human MT4 cells2021European journal of medicinal chemistry, Mar-05, Volume: 213Exploiting the tolerant region I of the non-nucleoside reverse transcriptase inhibitor (NNRTI) binding pocket. Part 2: Discovery of diarylpyrimidine derivatives as potent HIV-1 NNRTIs with high Fsp
AID347630Antiviral activity against NNRTI-resistant HIV HXB2 isolate 21 with reverse transcriptase K103, V179, Y181 mutation in human MT4 cells assessed as protection against virus-induced cell death after 5 days by MTT assay2008Journal of medicinal chemistry, Dec-11, Volume: 51, Issue:23
Design of annulated pyrazoles as inhibitors of HIV-1 reverse transcriptase.
AID1565099Antiviral activity against drug resistant HIV-1 harboring reverse transcriptase Y188L mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay
AID573474Antiviral activity of wild-type Human immunodeficiency virus 1 isolate 5269 by cell based assay2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID1572107Sub-acute toxicity in Kunming mouse assessed as alveolar hemorrhage at 50 mg/kg/day, po administered for 2 weeks by hematoxylin and eosin staining-based assay2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1572554Solubility in water at pH 7
AID1418464Antiviral activity against HIV1 RES056 harboring K103N/Y181C double mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 4 days by MTT assay2018Bioorganic & medicinal chemistry letters, 12-01, Volume: 28, Issue:22
Design and synthesis of a novel series of non-nucleoside HIV-1 inhibitors bearing pyrimidine and N-substituted aromatic piperazine.
AID1357784Antiviral activity against HIV-1 RES056 harboring reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as inhibition of viral replication after 5 days by MTT assay2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID508646Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase Y181C, Y188L mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1483274Inhibition of HIV1 3B reverse transcriptase Y188L mutant infected in human MT4 cells assessed as protection against virus induced cytotoxicity after 5 days by MTT assay2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1583019Antiviral activity against HIV1 3B infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2020Journal of medicinal chemistry, 02-13, Volume: 63, Issue:3
Discovery and Characterization of Fluorine-Substituted Diarylpyrimidine Derivatives as Novel HIV-1 NNRTIs with Highly Improved Resistance Profiles and Low Activity for the hERG Ion Channel.
AID1165076Antiviral activity against HIV1 RES056 expressing reverse transcriptase K103N + Y181C mutant infected in human MT4 cells assessed as inhibition of virus-induced syncytium formation after 5 days by MTT assay2014European journal of medicinal chemistry, Nov-24, Volume: 87Design, synthesis and anti-HIV evaluation of novel diarylnicotinamide derivatives (DANAs) targeting the entrance channel of the NNRTI binding pocket through structure-guided molecular hybridization.
AID1705185Selectivity ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV-1 NL4-3 infected in human MT4 cells2020European journal of medicinal chemistry, Dec-15, Volume: 208New indolylarylsulfone non-nucleoside reverse transcriptase inhibitors show low nanomolar inhibition of single and double HIV-1 mutant strains.
AID1572072Resistance factor, ratio of EC50 for HIV1 harboring reverse transcriptase Y188L mutant to EC50 for wild type HIV12019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1583026Antiviral activity against HIV1 containing reverse transcriptase Y181C mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2020Journal of medicinal chemistry, 02-13, Volume: 63, Issue:3
Discovery and Characterization of Fluorine-Substituted Diarylpyrimidine Derivatives as Novel HIV-1 NNRTIs with Highly Improved Resistance Profiles and Low Activity for the hERG Ion Channel.
AID517495Antiviral activity against HIV1 harboring reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect by replication assay in presence of 10% FBS2010Bioorganic & medicinal chemistry letters, Oct-15, Volume: 20, Issue:20
Exploration of piperidine-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-Phenyl derivatives with broad potency against resistant mutant viruses.
AID1357794Resistance factor, ratio of EC50 for HIV1 harboring reverse transcriptase L100I mutant to EC50 for wild-type HIV-1 3B2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID508636Antiviral activity against Human immunodeficiency virus 1 subtype G infected in human MT4 cells assessed as reduction in virus induced cytopathicity2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID557044Inhibition of HIV1 isolate R8 reverse transcriptase K103N mutant after 90 mins by electrochemiluminescence analysis2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID1483272Inhibition of HIV1 3B reverse transcriptase K103N mutant infected in human MT4 cells assessed as protection against virus induced cytotoxicity after 5 days by MTT assay2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1866020Thermodynamic aqueous solubility of the compound2022Bioorganic & medicinal chemistry, 02-15, Volume: 56Structural modification aimed for improving solubility of lead compounds in early phase drug discovery.
AID1754646Antiviral activity against HIV1 harboring RT F227L/V106A mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect incubated for 5 days by MTT assay2021Bioorganic & medicinal chemistry, 07-15, Volume: 42Exploiting the hydrophobic channel of the NNIBP: Discovery of novel diarylpyrimidines as HIV-1 NNRTIs against wild-type and K103N mutant viruses.
AID1750718Inhibition of HIV-1 p66/p51 reverse transcriptase K103N mutant incubated for 40 mins by picogreen dye-based spectrofluorometric analysis2021Journal of medicinal chemistry, 04-22, Volume: 64, Issue:8
Hydrophobic Pocket Occupation Design of Difluoro-Biphenyl-Diarylpyrimidines as Non-Nucleoside HIV-1 Reverse Transcriptase Inhibitors: from
AID1446820Antiviral activity against HIV1 harboring F227L/V106A double mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect measured at 5 days post infection by MTT assay2017European journal of medicinal chemistry, Apr-21, Volume: 130Discovery of uracil-bearing DAPYs derivatives as novel HIV-1 NNRTIs via crystallographic overlay-based molecular hybridization.
AID1815402Inhibition of HIV1 reverse transcriptase F227L/V106A double mutant using oligo(dT)16 as primer measured after 40 mins by picogreen-dye based spectrofluorimetric analysis2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID584084Antiviral activity against HIV1 NL4-3 harboring reverse transcriptase Y181C mutant infected in human SupT1 cells assessed as inhibition of viral replication after 21 days relative to wild type HIV1 NL4-32010Antimicrobial agents and chemotherapy, Oct, Volume: 54, Issue:10
Lersivirine, a nonnucleoside reverse transcriptase inhibitor with activity against drug-resistant human immunodeficiency virus type 1.
AID1637399Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for human HIV-1 3B harboring L100I mutant infected in human MT4 cells2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Arylazolyl(azinyl)thioacetanilides. Part 20: Discovery of novel purinylthioacetanilides derivatives as potent HIV-1 NNRTIs via a structure-based bioisosterism approach.
AID1773449Antiviral activity against wild type HIV1 harboring E138K mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathogenic effect by MTT assay
AID1421311Inhibition of HIV1 reverse transcriptase K103N/Y188C double mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect by MTT assay2018European journal of medicinal chemistry, Oct-05, Volume: 158Advances in diarylpyrimidines and related analogues as HIV-1 nonnucleoside reverse transcriptase inhibitors.
AID1483269Antiviral activity against wild type HIV1 3B harboring reverse transcriptase infected in human MT4 cells assessed as protection against virus induced cytotoxicity after 5 days by MTT assay2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1165078Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for HIV1 3B infected in human MT4 cells2014European journal of medicinal chemistry, Nov-24, Volume: 87Design, synthesis and anti-HIV evaluation of novel diarylnicotinamide derivatives (DANAs) targeting the entrance channel of the NNRTI binding pocket through structure-guided molecular hybridization.
AID1446826Inhibition of wild-type HIV1 reverse transcriptase assessed as decrease in biotin-dUTP incorporation using poly(A)/oligo(dT)15 as template/primer after 1 hr by ELISA2017European journal of medicinal chemistry, Apr-21, Volume: 130Discovery of uracil-bearing DAPYs derivatives as novel HIV-1 NNRTIs via crystallographic overlay-based molecular hybridization.
AID508831Antiviral activity against Human immunodeficiency virus 1 subtype C isolate 92BR025 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1761015Antiviral activity against HIV-1 harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as inhibition of virus induced cytopathic effect measured after 5 days by MTT assay2021European journal of medicinal chemistry, Feb-05, Volume: 211Design, synthesis, and evaluation of "dual-site"-binding diarylpyrimidines targeting both NNIBP and the NNRTI adjacent site of the HIV-1 reverse transcriptase.
AID726438Antiviral activity against multidrug resistant HIV1 IIIB containing reverse transcriptase Y181C mutation infected in human MT2 cells assessed as cytoprotection from infection by MTT colorimetric method2013Bioorganic & medicinal chemistry letters, Feb-15, Volume: 23, Issue:4
Optimization of benzyloxazoles as non-nucleoside inhibitors of HIV-1 reverse transcriptase to enhance Y181C potency.
AID1487262Cytotoxicity against human MT4 cells assessed as reduction in cell viability after 5 days by MTT assay2017Bioorganic & medicinal chemistry, 08-15, Volume: 25, Issue:16
Discovery of novel DAPY-IAS hybrid derivatives as potential HIV-1 inhibitors using molecular hybridization based on crystallographic overlays.
AID557042Antiviral activity against HIV1 isolate R8 harboring wild type reverse transcriptase infected in human MT4 cells assessed as inhibition of viral replication after 72 hrs in presence of 10% FBS2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID1862564Selectivity index, ratio of CC50 for cytotoxicity against human MT4 cells to EC50 of Anti-HIV activity against HIV-1 IIIB infected in human MT4 cells
AID1298252Inhibition of HIV1 reverse transcriptase p66/p51 G190A mutant using poly(rA) template and measured after 40 mins by picogreen based spectrofluorometry2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
1,6-Bis[(benzyloxy)methyl]uracil derivatives-Novel antivirals with activity against HIV-1 and influenza H1N1 virus.
AID449196Antimalarial activity against Plasmodium falciparum W2mef assessed as DNA positive erythrocytes by hoechst 33342-thiazole orange stain based flow cytometry assay2009Bioorganic & medicinal chemistry letters, Sep-15, Volume: 19, Issue:18
Addressing the malaria drug resistance challenge using flow cytometry to discover new antimalarials.
AID1561712Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV1 expressing RT E138K mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect
AID1811039Anti-viral activity against HIV1 harboring RT L100I mutant infected MV4 cells assessed as protection against virus-induced cytopathic effect by MTT assay2021Journal of medicinal chemistry, 07-22, Volume: 64, Issue:14
Improving Druggability of Novel Diarylpyrimidine NNRTIs by a Fragment-Based Replacement Strategy: From Biphenyl-DAPYs to Heteroaromatic-Biphenyl-DAPYs.
AID508782Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase E138Q mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID517489Inhibition of HIV1 recombinant wild type reverse transcriptase by SPA heteropolymeric assay2010Bioorganic & medicinal chemistry letters, Oct-15, Volume: 20, Issue:20
Exploration of piperidine-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-Phenyl derivatives with broad potency against resistant mutant viruses.
AID517493Antiviral activity against wild type HIV1 infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect by replication assay in presence of 40% human serum2010Bioorganic & medicinal chemistry letters, Oct-15, Volume: 20, Issue:20
Exploration of piperidine-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-Phenyl derivatives with broad potency against resistant mutant viruses.
AID573464Inhibition of RNA-dependent DNA polymerase activity of wild-type Human immunodeficiency virus 1 subtype B reverse transcriptase by filter-based filtration assay2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID83554Potency evaluated against wild type HIV-1 strain IIIB2004Journal of medicinal chemistry, May-06, Volume: 47, Issue:10
Roles of conformational and positional adaptability in structure-based design of TMC125-R165335 (etravirine) and related non-nucleoside reverse transcriptase inhibitors that are highly potent and effective against wild-type and drug-resistant HIV-1 varian
AID449193Antiviral activity against HIV1 3B infected in human MT4 cells assessed as inhibition of viral replication2009Bioorganic & medicinal chemistry letters, Sep-15, Volume: 19, Issue:18
Discovery of diarylpyridine derivatives as novel non-nucleoside HIV-1 reverse transcriptase inhibitors.
AID1561738Solubility of the compound in pH 7.0 buffer by HPLC analysis
AID1811046Selectivity index, ratio of CC50 for mock infected human MT4 cells to IC50 for HIV1 IIIB infected human MT4 cells2021Journal of medicinal chemistry, 07-22, Volume: 64, Issue:14
Improving Druggability of Novel Diarylpyrimidine NNRTIs by a Fragment-Based Replacement Strategy: From Biphenyl-DAPYs to Heteroaromatic-Biphenyl-DAPYs.
AID518737Inhibition of DNA-dependent DNA polymerase activity of wild type HIV1 subtype B reverse transcriptase by gel-based primer extension assay2010Antimicrobial agents and chemotherapy, Jun, Volume: 54, Issue:6
The M230L nonnucleoside reverse transcriptase inhibitor resistance mutation in HIV-1 reverse transcriptase impairs enzymatic function and viral replicative capacity.
AID347622Antiviral activity against NNRTI-resistant HIV HXB2 isolate 1 with reverse transcriptase G190, Y188 mutation in human MT4 cells assessed as protection against virus-induced cell death after 5 days by MTT assay2008Journal of medicinal chemistry, Dec-11, Volume: 51, Issue:23
Design of annulated pyrazoles as inhibitors of HIV-1 reverse transcriptase.
AID533680Tmax in healthy human plasma at 200 mg, po twice a day for 8 days by reverse-phase HPLC-MS method2008Antimicrobial agents and chemotherapy, Dec, Volume: 52, Issue:12
Minimal pharmacokinetic interaction between the human immunodeficiency virus nonnucleoside reverse transcriptase inhibitor etravirine and the integrase inhibitor raltegravir in healthy subjects.
AID586904Activity at BCRP2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1197836Lipophilicity, log P of the compound at pH 7.0 by HPLC method2015European journal of medicinal chemistry, Mar-06, Volume: 92Fused heterocycles bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 3: optimization of [1,2,4]triazolo[1,5-a]pyrimidine core via structure-based and physicochemical property-driven approaches.
AID1811038Inhibition of recombinant wild-type HIV1 p66/p51 reverse transcriptase assessed as inhibition of [3H]dGTP incorporation using poly (rA) as templates incubated for 40 mins by spectrofluorometer2021Journal of medicinal chemistry, 07-22, Volume: 64, Issue:14
Improving Druggability of Novel Diarylpyrimidine NNRTIs by a Fragment-Based Replacement Strategy: From Biphenyl-DAPYs to Heteroaromatic-Biphenyl-DAPYs.
AID1572075Inhibition of recombinant HIV1 reverse transcriptase using (DIG)-dUTP and biotin-labeled dNTPs as substrate after 1 hr by ELISA2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID584237Antiviral activity against HIV1 NL4-3 harboring reverse transcriptase V108I mutant derived from 17 days viral passages with lersivirine infected in human SupT1 cells assessed as inhibition of viral replication after 21 days relative to wild type HIV1 NL4-2010Antimicrobial agents and chemotherapy, Oct, Volume: 54, Issue:10
Lersivirine, a nonnucleoside reverse transcriptase inhibitor with activity against drug-resistant human immunodeficiency virus type 1.
AID1352320Antiviral activity against HIV1 harboring reverse transcriptase E138K mutant infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 4 days by MTT assay2018European journal of medicinal chemistry, Feb-10, Volume: 145Discovery of biphenyl-substituted diarylpyrimidines as non-nucleoside reverse transcriptase inhibitors with high potency against wild-type and mutant HIV-1.
AID1773443Selectivity ratio of CC50 for human MT4 cells to IC50 for HIV-1 3B infected in MT4 cells
AID1884229Antiviral activity against HIV-1 RES056 infected in human MT4 cells assessed as reduction in virus induced cytotoxicity incubated for 5 to 7 days by MTT assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Chemical space exploration around indolylarylsulfone scaffold led to a novel class of highly active HIV-1 NNRTIs with spiro structural features.
AID1773456Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 harboring F227L/V106A double mutant infected in human MT4 cells
AID1249717Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 3B infected in human MT4 cells2015European journal of medicinal chemistry, Sep-18, Volume: 102Pyrimidine sulfonylacetanilides with improved potency against key mutant viruses of HIV-1 by specific targeting of a highly conserved residue.
AID1705189Antiviral activity against HIV1 harboring RT K103N/Y181C double mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect by MTT assay2020European journal of medicinal chemistry, Dec-15, Volume: 208New indolylarylsulfone non-nucleoside reverse transcriptase inhibitors show low nanomolar inhibition of single and double HIV-1 mutant strains.
AID1743631Antiviral activity against HIV1 harboring RT F227L/V106Amutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Nov-15, Volume: 206Targeting dual tolerant regions of binding pocket: Discovery of novel morpholine-substituted diarylpyrimidines as potent HIV-1 NNRTIs with significantly improved water solubility.
AID541244Antiviral activity against HIV1 NL4-3 infected in human TZM-bl cells after 4 days by luciferase reporter gene assay2010Journal of medicinal chemistry, Dec-09, Volume: 53, Issue:23
Design, synthesis, and evaluation of diarylpyridines and diarylanilines as potent non-nucleoside HIV-1 reverse transcriptase inhibitors.
AID465424Oral bioavailability in Beagle dog at 4 mg/kg2010Bioorganic & medicinal chemistry letters, Mar-01, Volume: 20, Issue:5
N1-Alkyl pyrimidinediones as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1862568Antiviral activity against HIV-1 Y188L mutant infected in human MT4 cells assessed as reduction in viral replication by MTT assay
AID1072808Antiviral activity against HIV1 harboring wild type reverse transcriptase infected in human MT4 cells2014Bioorganic & medicinal chemistry, Mar-15, Volume: 22, Issue:6
Discovery of 2-pyridone derivatives as potent HIV-1 NNRTIs using molecular hybridization based on crystallographic overlays.
AID476469Antiviral activity against HIV1 3B infected in human MT2 cells assessed as inhibition of virus replication by MTT assay2010Bioorganic & medicinal chemistry letters, Apr-15, Volume: 20, Issue:8
Eastern extension of azoles as non-nucleoside inhibitors of HIV-1 reverse transcriptase; cyano group alternatives.
AID1565095Antiviral activity against drug resistant HIV-1 harboring reverse transcriptase E138K mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1743630Antiviral activity against HIV1 harboring RT E138K mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Nov-15, Volume: 206Targeting dual tolerant regions of binding pocket: Discovery of novel morpholine-substituted diarylpyrimidines as potent HIV-1 NNRTIs with significantly improved water solubility.
AID619638Antiviral activity against HIV-1 3B harboring RT Y188L mutant infected in human MT4 cells assessed as inhibition of virus-induced syncytium formation after 5 days by MTT assay2011European journal of medicinal chemistry, Oct, Volume: 46, Issue:10
Arylazolylthioacetanilide. Part 8: Design, synthesis and biological evaluation of novel 2-(2-(2,4-dichlorophenyl)-2H-1,2,4-triazol-3-ylthio)-N-arylacetamides as potent HIV-1 inhibitors.
AID1207370Inhibition of slow delayed inward rectifying potassium current (Iks) in Chinese Hamster Ovary (CHO) cells expressing hKvLQT1/hminK measured using IonWorks Quattro automated patch clamp platform
AID1882476Cytotoxicity against human MT4 cells assessed as cell viability by MTT assay2022Journal of medicinal chemistry, 03-10, Volume: 65, Issue:5
Contemporary Medicinal Chemistry Strategies for the Discovery and Development of Novel HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors.
AID1884489Antiviral activity against drug-resistant HIV-1 L100I mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Expansion of the S-CN-DABO scaffold to exploit the impact on inhibitory activities against the non-nucleoside HIV-1 reverse transcriptase.
AID517496Antiviral activity against HIV1 harboring reverse transcriptase Y188L mutant infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect by replication assay in presence of 10% FBS2010Bioorganic & medicinal chemistry letters, Oct-15, Volume: 20, Issue:20
Exploration of piperidine-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-Phenyl derivatives with broad potency against resistant mutant viruses.
AID508658Antiviral activity against Human immunodeficiency virus 1 subtype O isolate BCF03 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID508642Antiviral activity against Human immunodeficiency virus 1 subtype AE infected in human MT4 cells assessed as reduction in virus induced cytopathicity2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1457063Inhibition of HIV1 NL4-3 reverse transcriptase His-tagged p66/p51 associated RNA dependent DNA polymerase activity expressed in Escherichia coli assessed as reduction in [3H]dTTP incorporation using poly(rA)/oligo(dT) as template/primer by scintillation c2017Journal of medicinal chemistry, 08-10, Volume: 60, Issue:15
Chiral Indolylarylsulfone Non-Nucleoside Reverse Transcriptase Inhibitors as New Potent and Broad Spectrum Anti-HIV-1 Agents.
AID1731749Inhibition of reverse transcriptase Y188L mutant in HIV-1 infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay
AID557035Antiviral activity against HIV1 isolate R8 harboring reverse transcriptase K103N and Y181C mutant infected in human MT4 cells assessed as inhibition of viral replication after 72 hrs in presence of 50% human serum2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID1705186Antiviral activity against HIV1 harboring RT K103N mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect by MTT assay2020European journal of medicinal chemistry, Dec-15, Volume: 208New indolylarylsulfone non-nucleoside reverse transcriptase inhibitors show low nanomolar inhibition of single and double HIV-1 mutant strains.
AID1435513Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 expressing reverse transcriptase K103N mutant
AID1561727Inhibition of HIV1 RT Y188L mutant in presence of reconstituted template and viral nucleotides [digoxigenin (DIG)-dUTP, biotin-dUTP and dTTP] incubated for 1 hr by ELISA method
AID1206937Cytotoxicity against human MT4 cells assessed as reduction of cell viability after 4 days by MTT assay2015European journal of medicinal chemistry, Jun-05, Volume: 97Discovery of piperidin-4-yl-aminopyrimidine derivatives as potent non-nucleoside HIV-1 reverse transcriptase inhibitors.
AID1743629Antiviral activity against HIV1 harboring RT Y188L mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Nov-15, Volume: 206Targeting dual tolerant regions of binding pocket: Discovery of novel morpholine-substituted diarylpyrimidines as potent HIV-1 NNRTIs with significantly improved water solubility.
AID1316343Selectivity index, ratio of CC50 for human mock-infected MT4 cells to EC50 for HIV1 3B harboring reverse transcriptase Y181C mutant infected in human MT4 cells2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID508775Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase Y181C mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1815384Antiviral activity against HIV1 harboring reverse transcriptase E138K mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID508785Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase E138A mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1773836Antiviral activity against HIV1 harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2021Journal of medicinal chemistry, 09-23, Volume: 64, Issue:18
Structure-Based Design and Discovery of Pyridyl-Bearing Fused Bicyclic HIV-1 Inhibitors: Synthesis, Biological Characterization, and Molecular Modeling Studies.
AID508633Cytotoxicity against human MT4 cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1157583Antiviral activity against N119-sensitive HIV1 harboring Y181C mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathogenicity after 4 days by MTT assay2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Synthesis of novel fluoro analogues of MKC442 as microbicides.
AID726439Antiviral activity against multidrug resistant HIV1 IIIB containing reverse transcriptase infected in human MT2 cells assessed as cytoprotection from infection by MTT colorimetric method2013Bioorganic & medicinal chemistry letters, Feb-15, Volume: 23, Issue:4
Optimization of benzyloxazoles as non-nucleoside inhibitors of HIV-1 reverse transcriptase to enhance Y181C potency.
AID586876Antiproliferative activity against MDCK2 cells assessed as growth inhibition2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1572527Antiviral activity against HIV1 harboring reverse transcriptase Y188L mutant infected in human MT4 cells assessed as inhibition of virus-induced cytotoxicity after 5 days by MTT assay
AID1691439Selectivity index, ratio of CC50 for cytotoxicity against human MT4 cells to EC50 for antiviral activity against HIV1 RES056 infected in human MT4 cells2020European journal of medicinal chemistry, May-01, Volume: 193In situ click chemistry-based rapid discovery of novel HIV-1 NNRTIs by exploiting the hydrophobic channel and tolerant regions of NNIBP.
AID1763897Antiviral activity against HIV-1 IIIB infected in human MT4 cells assessed as protection against virus-induced cytotoxicity after 5 days by MTT assay2021Bioorganic & medicinal chemistry, 06-15, Volume: 40Design, synthesis and anti-HIV evaluation of novel 5-substituted diarylpyrimidine derivatives as potent HIV-1 NNRTIs.
AID1171591Resistance index, ratio of EC50 for HIV1 NL4-3 expressing reverse transcriptase L100I mutant to EC50 for wild type HIV1 NL4-32014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID83553Potency evaluated against NNRTI-Resistant HIV-1 strain Val106Ala2004Journal of medicinal chemistry, May-06, Volume: 47, Issue:10
Roles of conformational and positional adaptability in structure-based design of TMC125-R165335 (etravirine) and related non-nucleoside reverse transcriptase inhibitors that are highly potent and effective against wild-type and drug-resistant HIV-1 varian
AID1231488Cytotoxicity against mock-infected human MT4 cells after 5 days by MTT assay2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Anti-HIV diarylpyrimidine-quinolone hybrids and their mode of action.
AID449198Antimalarial activity against Plasmodium falciparum W2mef ring form by hoechst 33342-thiazole orange stain based flow cytometry assay2009Bioorganic & medicinal chemistry letters, Sep-15, Volume: 19, Issue:18
Addressing the malaria drug resistance challenge using flow cytometry to discover new antimalarials.
AID522374Resistance index, ratio of EC50 for recombinant HIV1 harboring reverse transcriptase V106A mutant clone to EC50 for wild type HIV12010Antimicrobial agents and chemotherapy, Apr, Volume: 54, Issue:4
Combination of V106I and V179D polymorphic mutations in human immunodeficiency virus type 1 reverse transcriptase confers resistance to efavirenz and nevirapine but not etravirine.
AID1457058Antiviral activity against HIV1 harboring reverse transcriptase K103N /Y1881C double mutant infected in human ML4 cells assessed as protection against virus-induced cytopathic effect measured on day 5 post infection by MTT assay2017Journal of medicinal chemistry, 08-10, Volume: 60, Issue:15
Chiral Indolylarylsulfone Non-Nucleoside Reverse Transcriptase Inhibitors as New Potent and Broad Spectrum Anti-HIV-1 Agents.
AID1292014Inhibition of wild type HIV1 Reverse transcriptase assessed as reduction of biotin deoxyuridine triphosphate incorporation into the wild type HIV1 reverse transcriptase after 1 hr by ELISA2016European journal of medicinal chemistry, Jun-10, Volume: 115Design, synthesis and evaluation of novel HIV-1 NNRTIs with dual structural conformations targeting the entrance channel of the NNRTI binding pocket.
AID1558854Antiviral activity against HIV1 harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2019Journal of medicinal chemistry, 12-26, Volume: 62, Issue:24
Ligand-Based Design of Nondimethylphenyl-Diarylpyrimidines with Improved Metabolic Stability, Safety, and Oral Pharmacokinetic Profiles.
AID517491Inhibition of HIV1 recombinant reverse transcriptase Y188L mutant by SPA heteropolymeric assay2010Bioorganic & medicinal chemistry letters, Oct-15, Volume: 20, Issue:20
Exploration of piperidine-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-Phenyl derivatives with broad potency against resistant mutant viruses.
AID1483283Selectivity index, ratio of CC50 for human MT4 cells to EC50 for reverse transcriptase Y188L mutant in human MT4 cells infected HIV1 3B2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1457062Ratio of EC50 for HIV-1 harboring reverse transcriptase K103N/Y1881C double mutant infected in human MT4 cells to EC50 for HIV1 NL4-3 infected in human MT4 cells2017Journal of medicinal chemistry, 08-10, Volume: 60, Issue:15
Chiral Indolylarylsulfone Non-Nucleoside Reverse Transcriptase Inhibitors as New Potent and Broad Spectrum Anti-HIV-1 Agents.
AID417447Inhibition of HIV1 reverse transcriptase Y181C mutant2009European journal of medicinal chemistry, Feb, Volume: 44, Issue:2
QSAR studies for diarylpyrimidines against HIV-1 reverse transcriptase wild-type and mutant strains.
AID1773840Antiviral activity against HIV1 harboring reverse transcriptase F227L/V106A double mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2021Journal of medicinal chemistry, 09-23, Volume: 64, Issue:18
Structure-Based Design and Discovery of Pyridyl-Bearing Fused Bicyclic HIV-1 Inhibitors: Synthesis, Biological Characterization, and Molecular Modeling Studies.
AID508769Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase H221Y mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1775796Selectivity index, ratio of CC50 for cytotoxicity against human MT4 cells reduction in cell viability for 5 days by MTT assay to EC50 for antiviral activity against NNRTI-resistant HIV-1 RES056 harboring RT K103N/Y181C mutant infected in human MT4 cells a2021Journal of medicinal chemistry, 04-08, Volume: 64, Issue:7
2,4,5-Trisubstituted Pyrimidines as Potent HIV-1 NNRTIs: Rational Design, Synthesis, Activity Evaluation, and Crystallographic Studies.
AID1880374Selectivity index, ratio of CC50 for cytotoxicity against human MT4 cells to EC50 for antiviral activity against wild type HIV-1 IIIB infected in human MT4 cells2022Journal of medicinal chemistry, 06-23, Volume: 65, Issue:12
Structure-Based Discovery of Novel NH
AID1163255Antiplasmodial activity against chloroquine-resistant Plasmodium falciparum K1 infected in human O positive erythrocyte assessed as reduction in parasitemia after 72 hrs2014Bioorganic & medicinal chemistry, Oct-01, Volume: 22, Issue:19
From human immunodeficiency virus non-nucleoside reverse transcriptase inhibitors to potent and selective antitrypanosomal compounds.
AID779524Inhibition of recombinant wild type HIV-1 reverse transcriptase p66/p51 expressed in Escherichia coli JM109 using poly(rA)/oligo(dT)16 (1:1.2) as template/primer after 40 mins by spectrofluorometric analysis2013Bioorganic & medicinal chemistry, Nov-01, Volume: 21, Issue:21
Towards new C6-rigid S-DABO HIV-1 reverse transcriptase inhibitors: synthesis, biological investigation and molecular modeling studies.
AID1231489Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for HIV1 3B infected in human MT4 cells2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Anti-HIV diarylpyrimidine-quinolone hybrids and their mode of action.
AID508780Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase E138S mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID347623Antiviral activity against NNRTI-resistant HIV HXB2 isolate 2 with reverse transcriptase A98, K101, Y181, G190 mutation in human MT4 cells assessed as protection against virus-induced cell death after 5 days by MTT assay2008Journal of medicinal chemistry, Dec-11, Volume: 51, Issue:23
Design of annulated pyrazoles as inhibitors of HIV-1 reverse transcriptase.
AID648424Antiviral activity against HIV1 expressing reverse transcriptase K103N/Y181C mutant infected in human MT4 cells assessed as protection against virus-induced cytotoxicity after 5 days by MTT assay2012European journal of medicinal chemistry, May, Volume: 51Synthesis and biological evaluation of piperidine-substituted triazine derivatives as HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1761019Antiviral activity against HIV-1 harboring reverse transcriptase F227L/V106A mutant infected in human MT4 cells assessed as inhibition of virus induced cytopathic effect measured after 5 days by MTT assay2021European journal of medicinal chemistry, Feb-05, Volume: 211Design, synthesis, and evaluation of "dual-site"-binding diarylpyrimidines targeting both NNIBP and the NNRTI adjacent site of the HIV-1 reverse transcriptase.
AID508653Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase K103N, V179I, Y181C mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1171587Antiviral activity against HIV1 NL4-3 expressing reverse transcriptase K103N-Y181C mutant infected in human MT4 cells assessed as reduction in virus-induced cell death by MTT assay2014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1320866Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for HIV1 3B expressing wild type reverse transcriptase2016European journal of medicinal chemistry, Oct-04, Volume: 121Structural optimization of pyridine-type DAPY derivatives to exploit the tolerant regions of the NNRTI binding pocket.
AID1561704Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV1 expressing RT L100I mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect
AID1572523Antiviral activity against HIV1 harboring reverse transcriptase E138K mutant infected in human MT4 cells assessed as inhibition of virus-induced cytotoxicity after 5 days by MTT assay
AID1561707Resistance index, ratio of EC50 for antiviral activity against HIV1 expressing RT K103N mutant infected in human MT4 cells to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells
AID1446816Antiviral activity against HIV1 harboring K103N mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect measured at 5 days post infection by MTT assay2017European journal of medicinal chemistry, Apr-21, Volume: 130Discovery of uracil-bearing DAPYs derivatives as novel HIV-1 NNRTIs via crystallographic overlay-based molecular hybridization.
AID1636147Antiviral activity against NNRTI resistant HIV1 harboring reverse transcriptase V106I/Y181C/G190A/H221Y mutant assessed as inhibition of viral infection in human TZM-bl cells after 48 hrs by luciferase reporter gene assay2016Journal of medicinal chemistry, Aug-11, Volume: 59, Issue:15
Discovery and Structure-Based Optimization of 2-Ureidothiophene-3-carboxylic Acids as Dual Bacterial RNA Polymerase and Viral Reverse Transcriptase Inhibitors.
AID738336Inhibition of HIV 1 reverse transcriptase assessed as incorporation of biotin-dUTP into poly rA.dT by ELISA2013Bioorganic & medicinal chemistry, Apr-01, Volume: 21, Issue:7
Synthesis and biological evaluation of pyridazine derivatives as novel HIV-1 NNRTIs.
AID1558848Cytotoxicity against human MT4 cells assessed as reduction in cell viability incubated for 5 days by MTT assay2019Journal of medicinal chemistry, 12-26, Volume: 62, Issue:24
Ligand-Based Design of Nondimethylphenyl-Diarylpyrimidines with Improved Metabolic Stability, Safety, and Oral Pharmacokinetic Profiles.
AID1565092Cytotoxicity against human MT4 cells assessed as reduction in cell viability incubated for 5 days by MTT assay
AID1773460Resistance factor, ratio of EC50 for HIV1 harboring Y188L mutant mutant infected in human MT4 cells to EC50 for HIV1 3B infected in MT4 cells
AID1572106Sub-acute toxicity in Kunming mouse assessed as alveolar interstitial thickening at 50 mg/kg/day, po administered for 2 weeks by hematoxylin and eosin staining-based assay2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID632799Antiviral activity against Human immunodeficiency virus 1 NL4.3 infected in human MT4 cells assessed as inhibition of viral infection2011Journal of medicinal chemistry, Dec-22, Volume: 54, Issue:24
Computationally-guided optimization of a docking hit to yield catechol diethers as potent anti-HIV agents.
AID508656Antiviral activity against Human immunodeficiency virus 1 subtype O misolate BCF01 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1207495Inhibition of rapid delayed inward rectifying potassium current (IKr) in Chinese hamster ovary (CHO) cells stable expressing hERG measured using IonWorks Barracuda automated patch clamp platform
AID1561709Resistance index, ratio of EC50 for antiviral activity against HIV1 expressing RT Y181C mutant infected in human MT4 cells to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells
AID1888693Resistance index, ratio of EC50 for antiviral activity against HIV1 with RT L100I mutant infected in human MT4 cells to EC50 for antiviral activity against wild type HIV1 infected in human MT4 cells
AID508628Antiviral activity against Human immunodeficiency virus 1 subtype F isolate 93BR020 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1316344Selectivity index, ratio of CC50 for human mock-infected MT4 cells to EC50 for HIV1 3B harboring reverse transcriptase Y188L mutant infected in human MT4 cells2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID1410397Antiviral activity against HIV1 harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as inhibition of viral replication after 5 days by MTT assay2018ACS medicinal chemistry letters, Apr-12, Volume: 9, Issue:4
Discovery of Novel Diarylpyrimidine Derivatives as Potent HIV-1 NNRTIs Targeting the "NNRTI Adjacent" Binding Site.
AID541249Selectivity index, ratio of CC50 for human MT2 cells to EC50 for HIV1 3B2010Journal of medicinal chemistry, Dec-09, Volume: 53, Issue:23
Design, synthesis, and evaluation of diarylpyridines and diarylanilines as potent non-nucleoside HIV-1 reverse transcriptase inhibitors.
AID1072806Antiviral activity against HIV1 harboring reverse transcriptase Y181C mutant infected in human MT4 cells2014Bioorganic & medicinal chemistry, Mar-15, Volume: 22, Issue:6
Discovery of 2-pyridone derivatives as potent HIV-1 NNRTIs using molecular hybridization based on crystallographic overlays.
AID1572054Cytotoxicity against human MT4 cells after 5 days by MTT assay2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID490362Antiviral activity against wild type HIV1 infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect in presence of 10 % fetal bovine serum2010Bioorganic & medicinal chemistry letters, Jul-15, Volume: 20, Issue:14
Discovery of piperidin-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-benzyl derivatives with broad potency against resistant mutant viruses.
AID508657Antiviral activity against Human immunodeficiency virus 1 subtype O isolate BCF02 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID541248Selectivity index, ratio of CC50 for human TZM-bl cells to EC50 for HIV1 NL4-32010Journal of medicinal chemistry, Dec-09, Volume: 53, Issue:23
Design, synthesis, and evaluation of diarylpyridines and diarylanilines as potent non-nucleoside HIV-1 reverse transcriptase inhibitors.
AID1483285Selectivity index, ratio of CC50 for human MT4 cells to EC50 for reverse transcriptase F227L/V106A double mutant in human MT4 cells infected HIV1 3B2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1157592Antiviral activity against 6-[(3,5-Dimethylphenyl)fluoromethyl]-5-ethyl-1-{[[4-(3-hydroxyprop-1-ynyl)benzyl]oxy]methyl}pyrimidine-2,4(1H,3H)-dione-resistant HIV1 harboring RT 106I, 181C mutant infected in human MT4 cells assessed as inhibition of virus-in2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Synthesis of novel fluoro analogues of MKC442 as microbicides.
AID557062Antiviral activity against HIV1 isolate R8 harboring reverse transcriptase Y181C and K103N mutant relative to drug-sensitive HIV1 CNDO2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID1815380Antiviral activity against HIV1 harboring reverse transcriptase L100I mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1275544Antiviral activity against HIV1 3B expressing reverse transcriptase Y188L mutant infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 5 days by MTT assay2016European journal of medicinal chemistry, Feb-15, Volume: 109Design, synthesis and anti-HIV evaluation of novel diarylpyridine derivatives targeting the entrance channel of NNRTI binding pocket.
AID1069123Antiviral activity against HIV harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as inhibition of viral infection in presence of 50% normal human serum2014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Discovery of MK-1439, an orally bioavailable non-nucleoside reverse transcriptase inhibitor potent against a wide range of resistant mutant HIV viruses.
AID1457060Ratio of EC50 for HIV-1 harboring reverse transcriptase Y181C mutant infected in human MT4 cells to EC50 for HIV1 NL4-3 infected in human MT4 cells2017Journal of medicinal chemistry, 08-10, Volume: 60, Issue:15
Chiral Indolylarylsulfone Non-Nucleoside Reverse Transcriptase Inhibitors as New Potent and Broad Spectrum Anti-HIV-1 Agents.
AID1558856Antiviral activity against HIV1 harboring reverse transcriptase Y188L mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2019Journal of medicinal chemistry, 12-26, Volume: 62, Issue:24
Ligand-Based Design of Nondimethylphenyl-Diarylpyrimidines with Improved Metabolic Stability, Safety, and Oral Pharmacokinetic Profiles.
AID508640Antiviral activity against Human immunodeficiency virus 1 subtype BG infected in human MT4 cells assessed as reduction in virus induced cytopathicity2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1298253Inhibition of HIV1 3B reverse transcriptase p66/p51 K103N/Y181C mutant using poly(rA) template and measured after 40 mins by picogreen based spectrofluorometry2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
1,6-Bis[(benzyloxy)methyl]uracil derivatives-Novel antivirals with activity against HIV-1 and influenza H1N1 virus.
AID573465Inhibition of Human immunodeficiency virus 1 subtype B reverse transcriptase G190A mutant by filter-based filtration assay2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID1596753Antiviral activity against HIV-1 3B harboring reverse transcriptase RES056 mutant infected in MT4 cells measured after 5 days by MTT assay2019European journal of medicinal chemistry, Aug-15, Volume: 176Synthesis and biological evaluation of dihydroquinazoline-2-amines as potent non-nucleoside reverse transcriptase inhibitors of wild-type and mutant HIV-1 strains.
AID1773464Solubility in phosphate buffer at pH7
AID1171585Antiviral activity against HIV1 NL4-3 expressing reverse transcriptase Y188L mutant infected in human MT4 cells assessed as reduction in virus-induced cell death by MTT assay2014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID614142Antiviral activity against wild type Human immunodeficiency virus 1 3B infected in human MT4 cells assessed as protection against virus-induced cytopathogenicity after 4 days by MTT assay2011Bioorganic & medicinal chemistry, Sep-01, Volume: 19, Issue:17
Synthesis and structure-activity relationship of novel diarylpyrimidines with hydromethyl linker (CH(OH)-DAPYs) as HIV-1 NNRTIs.
AID1275539Inhibition of HIV1 reverse transcriptase assessed as reduction in biotin-dUTP incorporation using poly(rA)-oligo(dT) as template/primer and digoxigenin-dUTP/biotin-dUTP/dTTP nucleotides incubated for 1 hr by ELISA2016European journal of medicinal chemistry, Feb-15, Volume: 109Design, synthesis and anti-HIV evaluation of novel diarylpyridine derivatives targeting the entrance channel of NNRTI binding pocket.
AID1357809Inhibition of wild-type HIV1 reverse transcriptase assessed as decrease in digoxigenin and biotin-dUTP incorporation in to DNA using poly(A)/oligo(dT)15 as template/primer after 1 hr by ELISA2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID1504704Antiviral activity against HIV1 3B harboring reverse transcriptase RES056 double mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2017ACS medicinal chemistry letters, Nov-09, Volume: 8, Issue:11
Discovery of Thiophene[3,2-
AID1248222Antiviral activity against wild type HIV 1 3B infected in human MT4 cells assessed as protection against virus-induced cytopathogenicity after 5 days by MTT assay2015Bioorganic & medicinal chemistry, Oct-15, Volume: 23, Issue:20
A novel family of diarylpyrimidines (DAPYs) featuring a diatomic linker: Design, synthesis and anti-HIV activities.
AID1443668Antiviral activity against HIV1 harboring reverse transcriptase Y188L mutant infected in human MT4 cells assessed as inhibition of viral replication after 5 days by MTT assay2017Bioorganic & medicinal chemistry letters, 04-15, Volume: 27, Issue:8
Design and synthesis of hybrids of diarylpyrimidines and diketo acids as HIV-1 inhibitors.
AID1773839Antiviral activity against HIV1 harboring reverse transcriptase L100I mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2021Journal of medicinal chemistry, 09-23, Volume: 64, Issue:18
Structure-Based Design and Discovery of Pyridyl-Bearing Fused Bicyclic HIV-1 Inhibitors: Synthesis, Biological Characterization, and Molecular Modeling Studies.
AID1357791Antiviral activity against HIV1 harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as inhibition of viral replication after 5 days by MTT assay2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID1743620Cytotoxicity against human MT4 cells assessed as reduction in cell viability incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Nov-15, Volume: 206Targeting dual tolerant regions of binding pocket: Discovery of novel morpholine-substituted diarylpyrimidines as potent HIV-1 NNRTIs with significantly improved water solubility.
AID1421300Inhibition of HIV1 reverse transcriptase K103N mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect by MTT assay2018European journal of medicinal chemistry, Oct-05, Volume: 158Advances in diarylpyrimidines and related analogues as HIV-1 nonnucleoside reverse transcriptase inhibitors.
AID1731756Solubility in water at pH 7.4 at 0.1 to 10 mg
AID1750727Selectivity index, ratio of CC50 for cytotoxicity against human MT-4 cells to EC50 for inhibition of HIV-1 p66/51 reverse transcriptase E138K mutant2021Journal of medicinal chemistry, 04-22, Volume: 64, Issue:8
Hydrophobic Pocket Occupation Design of Difluoro-Biphenyl-Diarylpyrimidines as Non-Nucleoside HIV-1 Reverse Transcriptase Inhibitors: from
AID508626Antiviral activity against Human immunodeficiency virus 1 subtype E isolate CMU08 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1583021Cytotoxicity in mock-infected human MT4 cells assessed as reduction in cell viability incubated for 5 days by MTT assay2020Journal of medicinal chemistry, 02-13, Volume: 63, Issue:3
Discovery and Characterization of Fluorine-Substituted Diarylpyrimidine Derivatives as Novel HIV-1 NNRTIs with Highly Improved Resistance Profiles and Low Activity for the hERG Ion Channel.
AID1171412Antiviral activity against wild type HIV1 harboring reverse transcriptase Y181C mutant infected in human MT2 cells assessed as protection from virus-induced cytopathicity by MTT assay2014ACS medicinal chemistry letters, Nov-13, Volume: 5, Issue:11
Picomolar Inhibitors of HIV-1 Reverse Transcriptase: Design and Crystallography of Naphthyl Phenyl Ethers.
AID1410406Antiviral activity against wild type HIV1 3B infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2018ACS medicinal chemistry letters, Apr-12, Volume: 9, Issue:4
Discovery of Novel Diarylpyrimidine Derivatives as Potent HIV-1 NNRTIs Targeting the "NNRTI Adjacent" Binding Site.
AID1483292Fold resistance, ratio of EC50 for reverse transcriptase Y188L mutant in human MT4 cells infected HIV1 3B to EC50 for wild type HIV1 3B harboring reverse transcriptase infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1304408Antiviral activity against HIV-1 3B expressing wild type reverse transcriptase infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect by MTT assay2016Bioorganic & medicinal chemistry, 07-01, Volume: 24, Issue:13
Systematic evaluation of methyl ester bioisosteres in the context of developing alkenyldiarylmethanes (ADAMs) as non-nucleoside reverse transcriptase inhibitors (NNRTIs) for anti-HIV-1 chemotherapy.
AID1572074Resistance factor, ratio of EC50 for HIV1 harboring reverse transcriptase F227L/V106A double mutant to EC50 for wild type HIV12019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1248226Antiviral activity against wild type HIV 2 ROD infected in human MT4 cells assessed as protection against virus-induced cytopathogenicity after 5 days by MTT assay2015Bioorganic & medicinal chemistry, Oct-15, Volume: 23, Issue:20
A novel family of diarylpyrimidines (DAPYs) featuring a diatomic linker: Design, synthesis and anti-HIV activities.
AID1456311Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV-1 3B wild type reverse transcriptase infected in human MT4 cells2017Bioorganic & medicinal chemistry, 04-15, Volume: 25, Issue:8
Structural modifications of diarylpyrimidines (DAPYs) as HIV-1 NNRTIs: Synthesis, anti-HIV activities and SAR.
AID1561729Inhibition of HIV1 RT V106A/F227L mutant in presence of reconstituted template and viral nucleotides [digoxigenin (DIG)-dUTP, biotin-dUTP and dTTP] incubated for 1 hr by ELISA method
AID508764Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase L100I, K101E mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1126507Antiviral activity against HIV1 harboring RT K103N/Y181C double mutant infected in human MT4 cells assessed as inhibition of viral cytopathogenicity after 5 days by MTT assay2014Bioorganic & medicinal chemistry, Apr-15, Volume: 22, Issue:8
Structural modifications of CH(OH)-DAPYs as new HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1421302Inhibition of HIV1 reverse transcriptase Y188L mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect by MTT assay2018European journal of medicinal chemistry, Oct-05, Volume: 158Advances in diarylpyrimidines and related analogues as HIV-1 nonnucleoside reverse transcriptase inhibitors.
AID260238Cytotoxicity against human MT2 cells by MTT assay2006Bioorganic & medicinal chemistry letters, Feb, Volume: 16, Issue:3
Optimization of diarylamines as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID508792Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase K101P mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1320863Antiviral activity against HIV1 3B expressing wild type reverse transcriptase infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay2016European journal of medicinal chemistry, Oct-04, Volume: 121Structural optimization of pyridine-type DAPY derivatives to exploit the tolerant regions of the NNRTI binding pocket.
AID1572060Antiviral activity against HIV1 harboring reverse transcriptase Y188L mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect after 5 days by MTT assay2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1565091Antiviral activity against drug resistant HIV-1 RES056 harboring reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1504701Antiviral activity against HIV1 3B harboring reverse transcriptase Y188L mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2017ACS medicinal chemistry letters, Nov-09, Volume: 8, Issue:11
Discovery of Thiophene[3,2-
AID496628Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase E138A/K20R, V35T, E36A, T39D, V60I, I135V, T139I, K173A, Q174K, T200E, Q207A, R211K, F214L mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID1298246Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV-1 3B infected in human MT4 cells2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
1,6-Bis[(benzyloxy)methyl]uracil derivatives-Novel antivirals with activity against HIV-1 and influenza H1N1 virus.
AID1316346Selectivity index, ratio of CC50 for human mock-infected MT4 cells to EC50 for HIV1 3B harboring reverse transcriptase F227L/V106A double mutant infected in human MT4 cells2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID508624Antiviral activity against Human immunodeficiency virus 1 subtype E isolate 92TH006 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1773463Inhibition of wild type HIV1 reverse transcriptase assessed as reduction in biotin-dUTP incorporation into protein incubated for 1 hrs by ELISA
AID1197831Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for HIV1 3B infected in human MT4 cells2015European journal of medicinal chemistry, Mar-06, Volume: 92Fused heterocycles bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 3: optimization of [1,2,4]triazolo[1,5-a]pyrimidine core via structure-based and physicochemical property-driven approaches.
AID1773462Resistance factor, ratio of EC50 for HIV1 harboring F227L/V106A double mutant infected in human MT4 cells to EC50 for HIV1 3B infected in MT4 cells
AID1637401Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for human HIV-1 3B harboring Y181C mutant infected in human MT4 cells2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Arylazolyl(azinyl)thioacetanilides. Part 20: Discovery of novel purinylthioacetanilides derivatives as potent HIV-1 NNRTIs via a structure-based bioisosterism approach.
AID586895Induction of SLCO2B1 mRNA expression in human LS180 cells at 0.1 to 1 umol/liter after 4 days by RT-PCR analysis2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID368563Binding affinity to HIV1 reverse transcriptase treated for 3 mins by surface plasmon resonance2009Journal of medicinal chemistry, Feb-12, Volume: 52, Issue:3
Discovery of chiral cyclopropyl dihydro-alkylthio-benzyl-oxopyrimidine (S-DABO) derivatives as potent HIV-1 reverse transcriptase inhibitors with high activity against clinically relevant mutants.
AID1862565Antiviral activity against HIV-1 L100I mutant infected in human MT4 cells assessed as reduction in viral replication by MTT assay
AID1572065Selectivity ratio of CC50 for human MT4 cells to EC50 for HIV1 harboring reverse transcriptase Y181C mutant infected in human MT4 cells2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1069125Inhibition of HIV wild-type reverse transcriptase by electrochemiluminescence assay2014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Discovery of MK-1439, an orally bioavailable non-nucleoside reverse transcriptase inhibitor potent against a wide range of resistant mutant HIV viruses.
AID1572058Antiviral activity against HIV1 harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect after 5 days by MTT assay2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID557040Antiviral activity against HIV1 isolate R8 harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as inhibition of viral replication after 72 hrs in presence of 10% FBS2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID1163253Trypanocidal activity against nifurtimox-sensitive Trypanosoma cruzi Tulahuen CL2 infected in human MRC5 SV2 cells assessed as parasite growth inhibition after 168 hrs by beta-galactosidase assay2014Bioorganic & medicinal chemistry, Oct-01, Volume: 22, Issue:19
From human immunodeficiency virus non-nucleoside reverse transcriptase inhibitors to potent and selective antitrypanosomal compounds.
AID1743619Antiviral activity against HIV1 RES056 infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Nov-15, Volume: 206Targeting dual tolerant regions of binding pocket: Discovery of novel morpholine-substituted diarylpyrimidines as potent HIV-1 NNRTIs with significantly improved water solubility.
AID1435512Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 expressing reverse transcriptase L100I mutant
AID518736Selectivity ratio of IC50 for RNA-dependent DNA polymerase activity of HIV1 subtype B reverse transcriptase M230L mutant to IC50 for RNA-dependent DNA polymerase activity of wild type HIV1 subtype B reverse transcriptase2010Antimicrobial agents and chemotherapy, Jun, Volume: 54, Issue:6
The M230L nonnucleoside reverse transcriptase inhibitor resistance mutation in HIV-1 reverse transcriptase impairs enzymatic function and viral replicative capacity.
AID476468Cytotoxicity against human MT2 cells by MTT assay2010Bioorganic & medicinal chemistry letters, Apr-15, Volume: 20, Issue:8
Eastern extension of azoles as non-nucleoside inhibitors of HIV-1 reverse transcriptase; cyano group alternatives.
AID557061Antiviral activity against HIV1 isolate R8 harboring reverse transcriptase L100I and K103N mutant relative to drug-sensitive HIV1 CNDO2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID1583027Antiviral activity against HIV1 containing reverse transcriptase Y188L mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2020Journal of medicinal chemistry, 02-13, Volume: 63, Issue:3
Discovery and Characterization of Fluorine-Substituted Diarylpyrimidine Derivatives as Novel HIV-1 NNRTIs with Highly Improved Resistance Profiles and Low Activity for the hERG Ion Channel.
AID738335Antiviral activity against wild type HIV1 3B infected in human MT4 cells assessed as protection against virus-induced cytopathicity after 5 days by MTT assay2013Bioorganic & medicinal chemistry, Apr-01, Volume: 21, Issue:7
Synthesis and biological evaluation of pyridazine derivatives as novel HIV-1 NNRTIs.
AID1352317Antiviral activity against HIV1 harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 4 days by MTT assay2018European journal of medicinal chemistry, Feb-10, Volume: 145Discovery of biphenyl-substituted diarylpyrimidines as non-nucleoside reverse transcriptase inhibitors with high potency against wild-type and mutant HIV-1.
AID1775801Antiviral activity against HIV-1 harboring RT E138K mutant infected in human MT4 cells assessed as protection against virus-induced cytopathogenicity after 5 days by MTT assay2021Journal of medicinal chemistry, 04-08, Volume: 64, Issue:7
2,4,5-Trisubstituted Pyrimidines as Potent HIV-1 NNRTIs: Rational Design, Synthesis, Activity Evaluation, and Crystallographic Studies.
AID496620Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase K101E/K30E, I31P, V35Q, T39S, E44G, E53G, A62G, N81H, Q91H,I135V,S162A, K173S, Q174R, D177E, V179I, T200I, Q207N, R211K, F214L mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID1572108Sub-acute toxicity in Kunming mouse assessed as proximal convoluted tubule edema in kidney at 50 mg/kg/day, po administered for 2 weeks by hematoxylin and eosin staining-based assay2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1206935Antiviral activity against HIV-1 RES056 harboring reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as protection from virus-induced cytopathicity after 4 days by MTT assay2015European journal of medicinal chemistry, Jun-05, Volume: 97Discovery of piperidin-4-yl-aminopyrimidine derivatives as potent non-nucleoside HIV-1 reverse transcriptase inhibitors.
AID496631Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase V90I,Y181C/V35T, T39M, P119S, D123E, S162A, E169D, K173T, Q174K, D177E, T200A, Q207E, K219N, V245Q, E248D mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID1773455Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 harboring E138K mutant infected in human MT4 cells
AID1561720Resistance index, ratio of EC50 for antiviral activity against HIV1 3B infected in human MT4 cells grown under 30 passages in presence of (E)-4-((4-((4-(4-(2-Cyanovinyl)-2,6-dimethylphenoxy)thieno[2,3- d]pyrimidin-2-yl)amino)piperidin-1-yl)methyl)benzenes
AID465423Volume of distribution at steady state in Beagle dog at 0.5 mg/kg, iv or. 5 mg/kg, po2010Bioorganic & medicinal chemistry letters, Mar-01, Volume: 20, Issue:5
N1-Alkyl pyrimidinediones as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1292012Selectivity index, ratio of CC50 for mock infected human MT4 cells to EC50 for HIV1 3B infected in human MT4 cells2016European journal of medicinal chemistry, Jun-10, Volume: 115Design, synthesis and evaluation of novel HIV-1 NNRTIs with dual structural conformations targeting the entrance channel of the NNRTI binding pocket.
AID1815400Inhibition of HIV1 reverse transcriptase Y188L mutant using oligo(dT)16 as primer measured after 40 mins by picogreen-dye based spectrofluorimetric analysis2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1822281Antiviral activity against HIV-1 harboring F227L/V106A double mutant infected in human MT4 cells assessed as protection against virus-induced cytotoxicity incubated for 5 days by MTT assay2022Journal of medicinal chemistry, 02-10, Volume: 65, Issue:3
Discovery of Novel Pyridine-Dimethyl-Phenyl-DAPY Hybrids by Molecular Fusing of Methyl-Pyrimidine-DAPYs and Difluoro-Pyridinyl-DAPYs: Improving the Druggability toward High Inhibitory Activity, Solubility, Safety, and PK.
AID1729150Antiviral activity against HIV1 RES056 harboring K103N/Y181C double mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect by MTT assay2021European journal of medicinal chemistry, Mar-05, Volume: 213Exploiting the tolerant region I of the non-nucleoside reverse transcriptase inhibitor (NNRTI) binding pocket. Part 2: Discovery of diarylpyrimidine derivatives as potent HIV-1 NNRTIs with high Fsp
AID1435506Antiviral activity against HIV1 expressing reverse transcriptase Y188L mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay
AID1761014Antiviral activity against HIV-1 harboring reverse transcriptase L100I mutant infected in human MT4 cells assessed as inhibition of virus induced cytopathic effect measured after 5 days by MTT assay2021European journal of medicinal chemistry, Feb-05, Volume: 211Design, synthesis, and evaluation of "dual-site"-binding diarylpyrimidines targeting both NNIBP and the NNRTI adjacent site of the HIV-1 reverse transcriptase.
AID1165075Antiviral activity against HIV2 ROD infected in human MT4 cells assessed as inhibition of virus-induced syncytium formation after 5 days by MTT assay2014European journal of medicinal chemistry, Nov-24, Volume: 87Design, synthesis and anti-HIV evaluation of novel diarylnicotinamide derivatives (DANAs) targeting the entrance channel of the NNRTI binding pocket through structure-guided molecular hybridization.
AID1729152Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV 3B infected in human MT4 cells2021European journal of medicinal chemistry, Mar-05, Volume: 213Exploiting the tolerant region I of the non-nucleoside reverse transcriptase inhibitor (NNRTI) binding pocket. Part 2: Discovery of diarylpyrimidine derivatives as potent HIV-1 NNRTIs with high Fsp
AID1822278Antiviral activity against HIV-1 harboring Y181C mutant infected in human MT4 cells assessed as protection against virus-induced cytotoxicity incubated for 5 days by MTT assay2022Journal of medicinal chemistry, 02-10, Volume: 65, Issue:3
Discovery of Novel Pyridine-Dimethyl-Phenyl-DAPY Hybrids by Molecular Fusing of Methyl-Pyrimidine-DAPYs and Difluoro-Pyridinyl-DAPYs: Improving the Druggability toward High Inhibitory Activity, Solubility, Safety, and PK.
AID465421Binding affinity to human serum albumin2010Bioorganic & medicinal chemistry letters, Mar-01, Volume: 20, Issue:5
N1-Alkyl pyrimidinediones as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1561718Antiviral activity against HIV1 3B infected in human MT4 cells grown under 30 passages in presence of 4-[[4-[[4-(4-cyano-2,6-dimethylphenoxy)thieno[3,2-d]pyrimidin-2-yl]amino]-1-piperidyl]methyl]benzenesulfonamide assessed as protection against virus-indu
AID508637Antiviral activity against Human immunodeficiency virus 1 subtype D infected in human MT4 cells assessed as reduction in virus induced cytopathicity2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1535519Solubility of the compound2019Bioorganic & medicinal chemistry, 02-01, Volume: 27, Issue:3
Design, synthesis and biological evaluation of novel acetamide-substituted doravirine and its prodrugs as potent HIV-1 NNRTIs.
AID709859Antiviral activity against NNRTI-resistant HIV1 harboring reverse transcriptase Y181C/V179F double mutant infected in human MT4 cells assessed as inhibition of viral replication after 72 hrs by HeLa-CD4-LTR-beta-gal assay2012Journal of medicinal chemistry, Dec-13, Volume: 55, Issue:23
Rational design of potent non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1391084Antiviral activity against HIV1 harboring reverse transcriptase Y188L mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay2018Bioorganic & medicinal chemistry, 05-01, Volume: 26, Issue:8
The discovery of novel diarylpyri(mi)dine derivatives with high level activity against a wide variety of HIV-1 strains as well as against HIV-2.
AID517494Selectivity ratio of IC50 for wild type HIV1 in presence of 40% human serum to IC50 for wild type HIV1 in presence of 10% FBS2010Bioorganic & medicinal chemistry letters, Oct-15, Volume: 20, Issue:20
Exploration of piperidine-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-Phenyl derivatives with broad potency against resistant mutant viruses.
AID1235377Antiviral activity against HIV1 RES056 expressing reverse transcriptase K103N + Y181C mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathicity incubated for 4 days by MTT method2015Bioorganic & medicinal chemistry, Aug-01, Volume: 23, Issue:15
Hybrid chemistry. Part 4: Discovery of etravirine-VRX-480773 hybrids as potent HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1884490Antiviral activity against drug-resistant HIV-1 K103 N mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Expansion of the S-CN-DABO scaffold to exploit the impact on inhibitory activities against the non-nucleoside HIV-1 reverse transcriptase.
AID508627Antiviral activity against Human immunodeficiency virus 1 subtype F isolate 93BR019 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1741391Cytotoxicity against human MT4 cells assessed as reduction in cell viability incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Sep-15, Volume: 202Bioisosterism-based design and enantiomeric profiling of chiral hydroxyl-substituted biphenyl-diarylpyrimidine nonnucleoside HIV-1 reverse transcriptase inhibitors.
AID517490Inhibition of HIV1 recombinant reverse transcriptase K103N/Y181C double mutant by SPA heteropolymeric assay2010Bioorganic & medicinal chemistry letters, Oct-15, Volume: 20, Issue:20
Exploration of piperidine-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-Phenyl derivatives with broad potency against resistant mutant viruses.
AID1206934Antiviral activity against wild type HIV1 3B infected in human MT4 cells assessed as protection from virus-induced cytopathicity after 4 days by MTT assay2015European journal of medicinal chemistry, Jun-05, Volume: 97Discovery of piperidin-4-yl-aminopyrimidine derivatives as potent non-nucleoside HIV-1 reverse transcriptase inhibitors.
AID1561719Antiviral activity against HIV1 3B infected in human MT4 cells grown under 30 passages in presence of 4-[[4-[[4-[4-[2-cyanovinyl]-2,6-dimethylphenoxy]thieno[3,2-d]pyrimidin-2-yl]amino]-1-piperidyl]methyl]benzenesulfonamide assessed as protection against v
AID1705184Cytotoxicity against human MT-4 cells assessed as reduction in cell viability by MTT assay2020European journal of medicinal chemistry, Dec-15, Volume: 208New indolylarylsulfone non-nucleoside reverse transcriptase inhibitors show low nanomolar inhibition of single and double HIV-1 mutant strains.
AID1583022Selectivity index, ratio of CC50 for cytotoxicity in mock-infected human MT4 cells assessed as reduction in cell viability incubated for 5 days by MTT assay to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells assessed as protection 2020Journal of medicinal chemistry, 02-13, Volume: 63, Issue:3
Discovery and Characterization of Fluorine-Substituted Diarylpyrimidine Derivatives as Novel HIV-1 NNRTIs with Highly Improved Resistance Profiles and Low Activity for the hERG Ion Channel.
AID1687681Antiviral activity against HIV-1 K103N mutant strain infected in human MT4 cells assessed as reduction in virus-induced cytopathogenicity by MTT assay2020European journal of medicinal chemistry, Jan-15, Volume: 186Indazolyl-substituted piperidin-4-yl-aminopyrimidines as HIV-1 NNRTIs: Design, synthesis and biological activities.
AID484276Colloidal aggregation in fed state simulated intestinal fluid by dynamic light scattering assay in presence of 1% DMSO2010Journal of medicinal chemistry, May-27, Volume: 53, Issue:10
Colloid formation by drugs in simulated intestinal fluid.
AID1888707Solubility in water at pH 7.4
AID1316342Selectivity index, ratio of CC50 for human mock-infected MT4 cells to EC50 for HIV1 3B harboring reverse transcriptase L100I mutant infected in human MT4 cells2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID1446813Cytotoxicity against human MT4 cells after 5 days by MTT assay2017European journal of medicinal chemistry, Apr-21, Volume: 130Discovery of uracil-bearing DAPYs derivatives as novel HIV-1 NNRTIs via crystallographic overlay-based molecular hybridization.
AID1443662Antiviral activity against HIV1 RES056 harboring reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as inhibition of viral replication after 5 days by MTT assay2017Bioorganic & medicinal chemistry letters, 04-15, Volume: 27, Issue:8
Design and synthesis of hybrids of diarylpyrimidines and diketo acids as HIV-1 inhibitors.
AID1561723Inhibition of HIV1 RT in presence of reconstituted template and viral nucleotides [digoxigenin (DIG)-dUTP, biotin-dUTP and dTTP] incubated for 1 hr by ELISA method
AID491829Resistant fold, ratio of EC50 for HIV1 with reverse transcriptase A17 (K103N, Y181C) mutant to EC50 for wild type HIV1 3B2010Journal of medicinal chemistry, Jul-08, Volume: 53, Issue:13
Diarylaniline derivatives as a distinct class of HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1572518Antiviral activity against HIV1 3B infected in human MT4 cells assessed as inhibition of virus-induced cytotoxicity after 5 days by MTT assay
AID1443661Antiviral activity against HIV1 harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as inhibition of viral replication after 5 days by MTT assay2017Bioorganic & medicinal chemistry letters, 04-15, Volume: 27, Issue:8
Design and synthesis of hybrids of diarylpyrimidines and diketo acids as HIV-1 inhibitors.
AID583852Antiviral activity against HIV1 NL4-3 infected in human MT2 cells assessed as inhibition of virus-induced cell death at 0.005 multiplicities of infection after 6 days2010Antimicrobial agents and chemotherapy, Oct, Volume: 54, Issue:10
Lersivirine, a nonnucleoside reverse transcriptase inhibitor with activity against drug-resistant human immunodeficiency virus type 1.
AID1884227Antiviral activity against HIV-1 harboring Y188L mutant infected in human MT4 cells assessed as reduction in virus induced cytotoxicity incubated for 5 to 7 days by MTT assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Chemical space exploration around indolylarylsulfone scaffold led to a novel class of highly active HIV-1 NNRTIs with spiro structural features.
AID648422Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 3B2012European journal of medicinal chemistry, May, Volume: 51Synthesis and biological evaluation of piperidine-substituted triazine derivatives as HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID508777Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase V179F mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID347625Antiviral activity against NNRTI-resistant HIV HXB2 isolate 12 with reverse transcriptase L100, K103 mutation in human MT4 cells assessed as protection against virus-induced cell death after 5 days by MTT assay2008Journal of medicinal chemistry, Dec-11, Volume: 51, Issue:23
Design of annulated pyrazoles as inhibitors of HIV-1 reverse transcriptase.
AID1731745Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV IIIB
AID698144Inhibition of HIV1 reverse transcriptase L100I mutant RNA-dependent DNA polymerase activity using poly(rA)/oligo(dT)10:1 and [3H]-dTTP substrate2012Journal of medicinal chemistry, Jul-26, Volume: 55, Issue:14
New nitrogen containing substituents at the indole-2-carboxamide yield high potent and broad spectrum indolylarylsulfone HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1474166Liver toxicity in human assessed as induction of drug-induced liver injury by measuring severity class index2016Drug discovery today, Apr, Volume: 21, Issue:4
DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans.
AID1763899Antiviral activity against HIV-1 IIIB harboring RT K103N mutant infected in human MT4 cells assessed as protection against virus-induced cytotoxicity incubated for 5 days by MTT assay2021Bioorganic & medicinal chemistry, 06-15, Volume: 40Design, synthesis and anti-HIV evaluation of novel 5-substituted diarylpyrimidine derivatives as potent HIV-1 NNRTIs.
AID1596754Cytotoxicity against human MT4 cells measured after 5 days by MTT assay2019European journal of medicinal chemistry, Aug-15, Volume: 176Synthesis and biological evaluation of dihydroquinazoline-2-amines as potent non-nucleoside reverse transcriptase inhibitors of wild-type and mutant HIV-1 strains.
AID1171592Resistance index, ratio of EC50 for HIV1 NL4-3 expressing reverse transcriptase K103N-Y181C mutant to EC50 for wild type HIV1 NL4-32014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1200848Inhibition of HIV1 reverse transcriptase assessed as reduction in biotin-labeled dUTP incorporation into DNA using poly(A) x oligo(dT)15 as template/primer after 1 hr by ELISA2015European journal of medicinal chemistry, Mar-26, Volume: 93Fused heterocycles bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 4: design, synthesis and biological evaluation of novel imidazo[1,2-a]pyrazines.
AID106057Inhibitory activity against 103N strain and 181C strain of HIV-I in MT-4 cells2001Bioorganic & medicinal chemistry letters, Sep-03, Volume: 11, Issue:17
Evolution of anti-HIV drug candidates. Part 3: Diarylpyrimidine (DAPY) analogues.
AID1357790Antiviral activity against HIV1 harboring reverse transcriptase E138K mutant infected in human MT4 cells assessed as inhibition of viral replication after 5 days by MTT assay2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID1352319Antiviral activity against HIV1 harboring reverse transcriptase Y188L mutant infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 4 days by MTT assay2018European journal of medicinal chemistry, Feb-10, Volume: 145Discovery of biphenyl-substituted diarylpyrimidines as non-nucleoside reverse transcriptase inhibitors with high potency against wild-type and mutant HIV-1.
AID533679Cmax in healthy human plasma at 200 mg, po twice a day for 4 days coadministered with 400 mg of raltegravir, po twice a day for 4 days by reverse-phase HPLC-MS method2008Antimicrobial agents and chemotherapy, Dec, Volume: 52, Issue:12
Minimal pharmacokinetic interaction between the human immunodeficiency virus nonnucleoside reverse transcriptase inhibitor etravirine and the integrase inhibitor raltegravir in healthy subjects.
AID1574385Inhibition of recombinant HIV-1 His-tagged reverse transcriptase p66/p51 K103N mutant expressed in Escherichia coli assessed as reduction in [3H]dTTP incorporation using poly(rA)/oligo(dT) as template/primer after 20 mins by scintillation counting analysi2019Journal of medicinal chemistry, 01-24, Volume: 62, Issue:2
Effect of α-Methoxy Substitution on the Anti-HIV Activity of Dihydropyrimidin-4(3 H)-ones.
AID1583023Selectivity index, ratio of CC50 for cytotoxicity in mock-infected human MT4 cells assessed as reduction in cell viability incubated for 5 days by MTT assay to EC50 for antiviral activity against NNRTI-resistant HIV1 RES056 containing reverse transcriptas2020Journal of medicinal chemistry, 02-13, Volume: 63, Issue:3
Discovery and Characterization of Fluorine-Substituted Diarylpyrimidine Derivatives as Novel HIV-1 NNRTIs with Highly Improved Resistance Profiles and Low Activity for the hERG Ion Channel.
AID490371Antiviral activity against HIV1 expressing reverse transcriptase G190A mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect2010Bioorganic & medicinal chemistry letters, Jul-15, Volume: 20, Issue:14
Discovery of piperidin-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-benzyl derivatives with broad potency against resistant mutant viruses.
AID1235380Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 3B infected in human MT4 cells2015Bioorganic & medicinal chemistry, Aug-01, Volume: 23, Issue:15
Hybrid chemistry. Part 4: Discovery of etravirine-VRX-480773 hybrids as potent HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1819211Cytotoxicity against human MT4 cells assessed as reduction in cell viability incubated for 5 days by MTT assay
AID1200843Antiviral activity against HIV1 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect after 5 days by MTT assay2015European journal of medicinal chemistry, Mar-26, Volume: 93Fused heterocycles bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 4: design, synthesis and biological evaluation of novel imidazo[1,2-a]pyrazines.
AID508788Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase V106A mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1357803Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 harboring reverse transcriptase Y181C mutant2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID586909Induction of MRP3 activity2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1763900Antiviral activity against HIV-1 IIIB harboring RT Y181C mutant infected in human MT4 cells assessed as protection against virus-induced cytotoxicity incubated for 5 days by MTT assay2021Bioorganic & medicinal chemistry, 06-15, Volume: 40Design, synthesis and anti-HIV evaluation of novel 5-substituted diarylpyrimidine derivatives as potent HIV-1 NNRTIs.
AID767493Aqueous solubility of the compound2013Bioorganic & medicinal chemistry letters, Sep-15, Volume: 23, Issue:18
Optimization of diarylazines as anti-HIV agents with dramatically enhanced solubility.
AID491828Antiviral activity against HIV1 with reverse transcriptase A17 (K103N, Y181C) mutant infected in human MT2 cells assessed as inhibition of p24 antigen production after 4 days by ELISA2010Journal of medicinal chemistry, Jul-08, Volume: 53, Issue:13
Diarylaniline derivatives as a distinct class of HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1207558Inhibition of long-lasting type calcium current (hICa) in Chinese Hamster Ovary (CHO) cells expressing hCav1.2 measured using IonWorks Quattro automated patch clamp platform
AID383497Inhibition of HIV reverse transcriptase2008European journal of medicinal chemistry, Mar, Volume: 43, Issue:3
SAR and QSAR studies: modelling of new DAPY derivatives.
AID665276Inhibition of HIV1 wildtype reverse transcriptase using poly(rA)/oligo(dT)15 as template by colometric streptavidin alkaline phosphate reporter assay2012European journal of medicinal chemistry, Jul, Volume: 53Chiral resolution, absolute configuration assignment and biological activity of racemic diarylpyrimidine CH(OH)-DAPY as potent nonnucleoside HIV-1 reverse transcriptase inhibitors.
AID508830Antiviral activity against Human immunodeficiency virus 1 subtype B isolate WEJO infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1750720Inhibition of HIV-1 p66/p51 reverse transcriptase Y188L mutant incubated for 40 mins by picogreen dye-based spectrofluorometric analysis2021Journal of medicinal chemistry, 04-22, Volume: 64, Issue:8
Hydrophobic Pocket Occupation Design of Difluoro-Biphenyl-Diarylpyrimidines as Non-Nucleoside HIV-1 Reverse Transcriptase Inhibitors: from
AID1574377Antiviral activity against HIV-1 harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2019Journal of medicinal chemistry, 01-24, Volume: 62, Issue:2
Effect of α-Methoxy Substitution on the Anti-HIV Activity of Dihydropyrimidin-4(3 H)-ones.
AID1326643Inhibition of wild type HIV-1 3B reverse transcriptase infected in human MT4 cells assessed as inhibition of viral replication after 5 days by cell titer glo based luciferase reporter gene assay2016European journal of medicinal chemistry, Oct-21, Volume: 122Novel (2,6-difluorophenyl)(2-(phenylamino)pyrimidin-4-yl)methanones with restricted conformation as potent non-nucleoside reverse transcriptase inhibitors against HIV-1.
AID1888699Antiviral activity against HIV1 with RT RES056 mutant infected in human MT4 cells cells assessed as protection against viral-induced cytopathogenecity by MTT assay
AID1326645Selectivity index, ratio of CC50 for human MT4 cells to EC50 for wild type HIV-1 3B reverse transcriptase infected in human MT4 cells2016European journal of medicinal chemistry, Oct-21, Volume: 122Novel (2,6-difluorophenyl)(2-(phenylamino)pyrimidin-4-yl)methanones with restricted conformation as potent non-nucleoside reverse transcriptase inhibitors against HIV-1.
AID767501Antiviral activity against HIV1 3B harboring reverse transcriptase Y181C mutant infected in human MT2 cells assessed as protection against virus-induced effect by MTT assay2013Bioorganic & medicinal chemistry letters, Sep-15, Volume: 23, Issue:18
Optimization of diarylazines as anti-HIV agents with dramatically enhanced solubility.
AID1754637Antiviral activity against wild type HIV-1 IIIB infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect measured after 5 days by MTT assay2021Bioorganic & medicinal chemistry, 07-15, Volume: 42Exploiting the hydrophobic channel of the NNIBP: Discovery of novel diarylpyrimidines as HIV-1 NNRTIs against wild-type and K103N mutant viruses.
AID1763901Antiviral activity against HIV-1 IIIB harboring RT Y188L mutant infected in human MT4 cells assessed as protection against virus-induced cytotoxicity incubated for 5 days by MTT assay2021Bioorganic & medicinal chemistry, 06-15, Volume: 40Design, synthesis and anti-HIV evaluation of novel 5-substituted diarylpyrimidine derivatives as potent HIV-1 NNRTIs.
AID1443663Cytotoxicity against human MT4 cells assessed as decrease in cell viability after 5 days by MTT assay2017Bioorganic & medicinal chemistry letters, 04-15, Volume: 27, Issue:8
Design and synthesis of hybrids of diarylpyrimidines and diketo acids as HIV-1 inhibitors.
AID1572530Inhibition of HIV1 reverse transcriptase p66/p51 using poly(rA)/oligo(dT)16 as template/primer measured after 40 mins by pico-green based spectrofluorometric analysis
AID1750726Selectivity index, ratio of CC50 for cytotoxicity against human MT-4 cells to EC50 for inhibition of HIV-1 p66/51 reverse transcriptase Y188L mutant2021Journal of medicinal chemistry, 04-22, Volume: 64, Issue:8
Hydrophobic Pocket Occupation Design of Difluoro-Biphenyl-Diarylpyrimidines as Non-Nucleoside HIV-1 Reverse Transcriptase Inhibitors: from
AID1572064Selectivity ratio of CC50 for human MT4 cells to EC50 for HIV1 harboring reverse transcriptase K103N mutant infected in human MT4 cells2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1561708Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV1 expressing RT Y181C mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect
AID1421299Inhibition of HIV1 reverse transcriptase L100I mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect by MTT assay2018European journal of medicinal chemistry, Oct-05, Volume: 158Advances in diarylpyrimidines and related analogues as HIV-1 nonnucleoside reverse transcriptase inhibitors.
AID508763Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase L100I, K103N mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1880378Antiviral activity against HIV-1 IIIB harboring reverse transcriptase Y181C mutant infected in human MT4 cells by MTT assay2022Journal of medicinal chemistry, 06-23, Volume: 65, Issue:12
Structure-Based Discovery of Novel NH
AID1207310Inhibition of fast sodium current (INa) in Chinese Hamster Ovary (CHO) K1 cells transfected with human Nav1.5 measured using IonWorks Quattro automated patch clamp platform
AID1561724Inhibition of HIV1 RT L100I mutant in presence of reconstituted template and viral nucleotides [digoxigenin (DIG)-dUTP, biotin-dUTP and dTTP] incubated for 1 hr by ELISA method
AID496633Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase V90I/V35T, E36D, K43R, V60I, K101R, K122E, D123S, I135V, S162A, K173T, Q174K, D177E, V179I, G196E, T200A, Q207E mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID1483271Inhibition of HIV1 3B reverse transcriptase L100I mutant infected in human MT4 cells assessed as protection against virus induced cytotoxicity after 5 days by MTT assay2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID246283Effective concentration of the compound to inhibit HIV-1 mutant LAI replication in HIV-infected MT-4 cells2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
From 4,5,6,7-tetrahydro-5-methylimidazo[4,5,1-jk](1,4)benzodiazepin-2(1H)-one (TIBO) to etravirine (TMC125): fifteen years of research on non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1743636Resistance index, ratio of EC50 for antiviral activity against HIV1 expressing RT Y181C mutant infected in human MT4 cells to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells2020European journal of medicinal chemistry, Nov-15, Volume: 206Targeting dual tolerant regions of binding pocket: Discovery of novel morpholine-substituted diarylpyrimidines as potent HIV-1 NNRTIs with significantly improved water solubility.
AID1379962Cytotoxicity in mock-infected human MT4 cells assessed as reduction in cell viability by MTT assay2017European journal of medicinal chemistry, Nov-10, Volume: 140Design, synthesis and anti-HIV evaluation of novel diarylpyridine derivatives as potent HIV-1 NNRTIs.
AID465420Antiviral activity against NNRTI-resistant HIV1 harboring reverse transcriptase K103N mutant infected in human MT2 cells2010Bioorganic & medicinal chemistry letters, Mar-01, Volume: 20, Issue:5
N1-Alkyl pyrimidinediones as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1815387Resistance fold, ratio of EC50 for HIV1 harboring reverse transcriptase L100I mutant infected MV4 cells to EC50 for wild type HIV1 infected MV4 cells2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1435508Antiviral activity against HIV1 expressing reverse transcriptase F227L/V106A double mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay
AID541247Cytotoxicity in human MT2 cells after 4 day by XTT assay2010Journal of medicinal chemistry, Dec-09, Volume: 53, Issue:23
Design, synthesis, and evaluation of diarylpyridines and diarylanilines as potent non-nucleoside HIV-1 reverse transcriptase inhibitors.
AID347624Antiviral activity against NNRTI-resistant HIV HXB2 isolate 6 with reverse transcriptase L100, K103 mutation in human MT4 cells assessed as protection against virus-induced cell death after 5 days by MTT assay2008Journal of medicinal chemistry, Dec-11, Volume: 51, Issue:23
Design of annulated pyrazoles as inhibitors of HIV-1 reverse transcriptase.
AID106069Inhibitory activity against 188L strain of HIV-I in MT-4 cells2001Bioorganic & medicinal chemistry letters, Sep-03, Volume: 11, Issue:17
Evolution of anti-HIV drug candidates. Part 3: Diarylpyrimidine (DAPY) analogues.
AID476472Antiviral activity against HIV1 reverse transcriptase K103N/Y181C double mutant infected in human MT2 cells assessed as inhibition of viral replication by MTT assay2010Bioorganic & medicinal chemistry letters, Apr-15, Volume: 20, Issue:8
Eastern extension of azoles as non-nucleoside inhibitors of HIV-1 reverse transcriptase; cyano group alternatives.
AID1418467Selectivity index, ratio of CC50 for human MT4 cells to EC50 for wild-type HIV1 3B infected in human MT4 cells2018Bioorganic & medicinal chemistry letters, 12-01, Volume: 28, Issue:22
Design and synthesis of a novel series of non-nucleoside HIV-1 inhibitors bearing pyrimidine and N-substituted aromatic piperazine.
AID1884540Fold resistance, ratio of EC50 for antiviral activity against drug-resistant HIV-1 Y181C mutant infected in human MT4 cells to EC50 for antiviral activity against NNRTI resistant wild type HIV-1 infected in human MT4 cells2022European journal of medicinal chemistry, Aug-05, Volume: 238Expansion of the S-CN-DABO scaffold to exploit the impact on inhibitory activities against the non-nucleoside HIV-1 reverse transcriptase.
AID1390712Cytotoxicity against human MT4 cells after 5 days by MTT assay2018Bioorganic & medicinal chemistry letters, 05-01, Volume: 28, Issue:8
First discovery of a potential carbonate prodrug of NNRTI drug candidate RDEA427 with submicromolar inhibitory activity against HIV-1 K103N/Y181C double mutant strain.
AID1888696Antiviral activity against HIV1 with RT Y181C mutant infected in human MT4 cells cells assessed as protection against viral-induced cytopathogenecity by MTT assay
AID1882473Antiviral activity against wild type HIV-1 IIIB infected in human MT4 cells assessed as protection against virus-induced cytopathicity by MTT assay2022Journal of medicinal chemistry, 03-10, Volume: 65, Issue:5
Contemporary Medicinal Chemistry Strategies for the Discovery and Development of Novel HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors.
AID1316329Inhibition of recombinant wild type HIV1 reverse transcriptase using DIG-dUTP/biotin-dUTP/dTTP assessed as suppression of biotin-dUTP incorporation after 1 hr by ELISA2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID106065Inhibitory activity against 181C strain of HIV-I in MT-4 cells2001Bioorganic & medicinal chemistry letters, Sep-03, Volume: 11, Issue:17
Evolution of anti-HIV drug candidates. Part 3: Diarylpyrimidine (DAPY) analogues.
AID1729163Inhibition of HIV1 reverse transcriptase assessed as reduction in biotin-dUTP incorporation into template incubated for 1 hr by ELISA2021European journal of medicinal chemistry, Mar-05, Volume: 213Exploiting the tolerant region I of the non-nucleoside reverse transcriptase inhibitor (NNRTI) binding pocket. Part 2: Discovery of diarylpyrimidine derivatives as potent HIV-1 NNRTIs with high Fsp
AID1435514Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 expressing reverse transcriptase Y181C mutant
AID1561725Inhibition of HIV1 RT K103N mutant in presence of reconstituted template and viral nucleotides [digoxigenin (DIG)-dUTP, biotin-dUTP and dTTP] incubated for 1 hr by ELISA method
AID1157580Cytotoxicity against human MT4 cells after 96 hrs by MTT assay2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Synthesis of novel fluoro analogues of MKC442 as microbicides.
AID584238Antiviral activity against HIV1 NL4-3 harboring reverse transcriptase K103N, V108V/I mutant derived from 10 days viral passages with lersivirine infected in human SupT1 cells assessed as inhibition of viral replication after 21 days relative to wild type 2010Antimicrobial agents and chemotherapy, Oct, Volume: 54, Issue:10
Lersivirine, a nonnucleoside reverse transcriptase inhibitor with activity against drug-resistant human immunodeficiency virus type 1.
AID1298251Inhibition of HIV1 reverse transcriptase p66/p51 Y181C mutant using poly(rA) template and measured after 40 mins by picogreen based spectrofluorometry2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
1,6-Bis[(benzyloxy)methyl]uracil derivatives-Novel antivirals with activity against HIV-1 and influenza H1N1 virus.
AID1687686Cytotoxicity against human MT4 cells assessed as inhibition of cell viability by measuring reduction in absorbance at OD540 by MTT assay2020European journal of medicinal chemistry, Jan-15, Volume: 186Indazolyl-substituted piperidin-4-yl-aminopyrimidines as HIV-1 NNRTIs: Design, synthesis and biological activities.
AID1819234Aqueous solubility of the compound at pH 2.0 by HPLC analysis
AID1743634Resistance index, ratio of EC50 for antiviral activity against HIV1 expressing RT L100I mutant infected in human MT4 cells to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells2020European journal of medicinal chemistry, Nov-15, Volume: 206Targeting dual tolerant regions of binding pocket: Discovery of novel morpholine-substituted diarylpyrimidines as potent HIV-1 NNRTIs with significantly improved water solubility.
AID1275543Antiviral activity against HIV1 RES056 expressing reverse transcriptase F227L/V106A double mutant infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 5 days by MTT assay2016European journal of medicinal chemistry, Feb-15, Volume: 109Design, synthesis and anti-HIV evaluation of novel diarylpyridine derivatives targeting the entrance channel of NNRTI binding pocket.
AID1069124Antiviral activity against HIV harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as inhibition of viral infection in presence of 50% normal human serum2014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Discovery of MK-1439, an orally bioavailable non-nucleoside reverse transcriptase inhibitor potent against a wide range of resistant mutant HIV viruses.
AID1819217Antiviral activity against HIV-1 harboring reverse transcriptase Y188L mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1754641Antiviral activity against HIV1 harboring RT L100I mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect incubated for 5 days by MTT assay2021Bioorganic & medicinal chemistry, 07-15, Volume: 42Exploiting the hydrophobic channel of the NNIBP: Discovery of novel diarylpyrimidines as HIV-1 NNRTIs against wild-type and K103N mutant viruses.
AID1596755Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV-1 3B infected in MT4 cells2019European journal of medicinal chemistry, Aug-15, Volume: 176Synthesis and biological evaluation of dihydroquinazoline-2-amines as potent non-nucleoside reverse transcriptase inhibitors of wild-type and mutant HIV-1 strains.
AID586882Cmax in human plasma at 200 mg bid after 8 days2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID347636Antiviral activity against NNRTI-resistant HIV HXB2 isolate 43 with reverse transcriptase K103, Y181, P225 mutation in human MT4 cells assessed as protection against virus-induced cell death after 5 days by MTT assay2008Journal of medicinal chemistry, Dec-11, Volume: 51, Issue:23
Design of annulated pyrazoles as inhibitors of HIV-1 reverse transcriptase.
AID1561695Cytotoxicity against human MT4 cells assessed as reduction in cell viability incubated for 5 days by MTT assay
AID1357788Antiviral activity against HIV1 harboring reverse transcriptase L100I mutant infected in human MT4 cells assessed as inhibition of viral replication after 5 days by MTT assay2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID1750723Selectivity index, ratio of CC50 for cytotoxicity against human MT-4 cells to EC50 for inhibition of HIV-1 p66/51 reverse transcriptase L100I mutant2021Journal of medicinal chemistry, 04-22, Volume: 64, Issue:8
Hydrophobic Pocket Occupation Design of Difluoro-Biphenyl-Diarylpyrimidines as Non-Nucleoside HIV-1 Reverse Transcriptase Inhibitors: from
AID1165077Cytotoxicity against mock-infected human MT4 cells after 5 days by MTT assay2014European journal of medicinal chemistry, Nov-24, Volume: 87Design, synthesis and anti-HIV evaluation of novel diarylnicotinamide derivatives (DANAs) targeting the entrance channel of the NNRTI binding pocket through structure-guided molecular hybridization.
AID573466Inhibition of Human immunodeficiency virus 1 subtype B reverse transcriptase Y181C mutant by filter-based filtration assay2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID1687683Antiviral activity against HIV-1 E138K mutant strain infected in human MT4 cells assessed as reduction in virus-induced cytopathogenicity by MTT assay2020European journal of medicinal chemistry, Jan-15, Volume: 186Indazolyl-substituted piperidin-4-yl-aminopyrimidines as HIV-1 NNRTIs: Design, synthesis and biological activities.
AID1884491Antiviral activity against drug-resistant HIV-1 Y181C mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Expansion of the S-CN-DABO scaffold to exploit the impact on inhibitory activities against the non-nucleoside HIV-1 reverse transcriptase.
AID1357783Antiviral activity against wild-type HIV-1 3B infected in human MT4 cells assessed as inhibition of viral replication after 5 days by MTT assay2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID533678Cmax in healthy human plasma at 200 mg, po twice a day for 8 days by reverse-phase HPLC-MS method2008Antimicrobial agents and chemotherapy, Dec, Volume: 52, Issue:12
Minimal pharmacokinetic interaction between the human immunodeficiency virus nonnucleoside reverse transcriptase inhibitor etravirine and the integrase inhibitor raltegravir in healthy subjects.
AID586878Inhibition of BCRP expressed in MDCK2 cells assessed as cell growth inhibition by calcein and pheophorbide A efflux assays2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1157582Antiviral activity against efavirenz-resistant HIV1 harboring RT 100I, 103R, 179D, 225H mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathogenicity after 4 days by MTT assay2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Synthesis of novel fluoro analogues of MKC442 as microbicides.
AID1249719Antiviral activity against HIV1 expressing reverse transcriptase K103N mutant2015European journal of medicinal chemistry, Sep-18, Volume: 102Pyrimidine sulfonylacetanilides with improved potency against key mutant viruses of HIV-1 by specific targeting of a highly conserved residue.
AID1520059Cytotoxicity against human MT4 cells assessed as reduction in cell viability incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Fragment hopping-based discovery of novel sulfinylacetamide-diarylpyrimidines (DAPYs) as HIV-1 nonnucleoside reverse transcriptase inhibitors.
AID508796Ratio of EC50 for HIV1 in presence of 1 mg/ml alpha-1 acid-glycoprotein to EC50 for HIV1 in absence of serum proteins by GFP assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID584240Antiviral activity against HIV1 NL4-3 harboring reverse transcriptase Y181C, V108V/I, V179V/D mutant derived from 17 days viral passages with lersivirine infected in human SupT1 cells assessed as inhibition of viral replication after 21 days relative to w2010Antimicrobial agents and chemotherapy, Oct, Volume: 54, Issue:10
Lersivirine, a nonnucleoside reverse transcriptase inhibitor with activity against drug-resistant human immunodeficiency virus type 1.
AID573471Inhibition of DNA-dependent DNA polymerase activity of wild-type Human immunodeficiency virus 1 subtype B reverse transcriptase G190A-81C mutant by filter-based filtration assay2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID586911Induction of SLCO1B1 activity2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1357801Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 harboring reverse transcriptase K103N mutant2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID557039Antiviral activity against HIV1 isolate R8 harboring reverse transcriptase K103N and Y181C mutant infected in human MT4 cells assessed as inhibition of viral replication after 72 hrs in presence of 10% FBS2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID584241Ratio of EC50 for wild type HIV1 NL4-3 to EC50 for HIV1 NL4-3 harboring reverse transcriptase V179V/D mutant2010Antimicrobial agents and chemotherapy, Oct, Volume: 54, Issue:10
Lersivirine, a nonnucleoside reverse transcriptase inhibitor with activity against drug-resistant human immunodeficiency virus type 1.
AID508791Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase K101Q mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1574375Antiviral activity against HIV 1 harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2019Journal of medicinal chemistry, 01-24, Volume: 62, Issue:2
Effect of α-Methoxy Substitution on the Anti-HIV Activity of Dihydropyrimidin-4(3 H)-ones.
AID1815383Antiviral activity against HIV1 harboring reverse transcriptase Y188L mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1060625Inhibition of wild-type HIV1 reverse transcriptase assessed as incorporation of biotin-labeled dUTP2014Bioorganic & medicinal chemistry, Jan-01, Volume: 22, Issue:1
Design, synthesis and preliminary SAR studies of novel N-arylmethyl substituted piperidine-linked aniline derivatives as potent HIV-1 NNRTIs.
AID1637387Inhibition of HIV-1 reverse transcriptase assessed as reduction in biotin-duTP incorporation using biotin-labeled dUTP and hybrid poly(A)-oligo (dT)15 template primer after 2 hrs by ELISA2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Arylazolyl(azinyl)thioacetanilides. Part 20: Discovery of novel purinylthioacetanilides derivatives as potent HIV-1 NNRTIs via a structure-based bioisosterism approach.
AID1435505Antiviral activity against HIV1 expressing reverse transcriptase Y181C mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay
AID1888703Resistance index, ratio of EC50 for antiviral activity against HIV1 with RT E138K mutant infected in human MT4 cells to EC50 for antiviral activity against wild type HIV1 infected in human MT4 cells
AID484390Colloidal aggregation in fed state simulated intestinal fluid by dynamic light scattering-based beads autocorrelation assay in presence of 1% DMSO2010Journal of medicinal chemistry, May-27, Volume: 53, Issue:10
Colloid formation by drugs in simulated intestinal fluid.
AID1819219Antiviral activity against HIV-1 harboring reverse transcriptase F227L/V106A mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1775791Antiviral activity against HIV-1 strain IIIB infected in human MT4 cells assessed as protection against virus-induced cytopathicity after 5 days by MTT assay2021Journal of medicinal chemistry, 04-08, Volume: 64, Issue:7
2,4,5-Trisubstituted Pyrimidines as Potent HIV-1 NNRTIs: Rational Design, Synthesis, Activity Evaluation, and Crystallographic Studies.
AID1775798Antiviral activity against HIV-1 harboring RT K103N mutant infected in human MT4 cells assessed as protection against virus-induced cytopathogenicity after 5 days by MTT assay2021Journal of medicinal chemistry, 04-08, Volume: 64, Issue:7
2,4,5-Trisubstituted Pyrimidines as Potent HIV-1 NNRTIs: Rational Design, Synthesis, Activity Evaluation, and Crystallographic Studies.
AID1761018Antiviral activity against HIV-1 harboring reverse transcriptase E138K mutant infected in human MT4 cells assessed as inhibition of virus induced cytopathic effect measured after 5 days by MTT assay2021European journal of medicinal chemistry, Feb-05, Volume: 211Design, synthesis, and evaluation of "dual-site"-binding diarylpyrimidines targeting both NNIBP and the NNRTI adjacent site of the HIV-1 reverse transcriptase.
AID586890Induction of SLCO1B1 mRNA expression in human LS180 cells at 0.1 to 1 umol/liter after 4 days by RT-PCR analysis2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1761016Antiviral activity against HIV-1 harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as inhibition of virus induced cytopathic effect measured after 5 days by MTT assay2021European journal of medicinal chemistry, Feb-05, Volume: 211Design, synthesis, and evaluation of "dual-site"-binding diarylpyrimidines targeting both NNIBP and the NNRTI adjacent site of the HIV-1 reverse transcriptase.
AID347637Antiviral activity against NNRTI-resistant HIV HXB2 isolate 47 with reverse transcriptase A98, V108, G190, F227 mutation in human MT4 cells assessed as protection against virus-induced cell death after 5 days by MTT assay2008Journal of medicinal chemistry, Dec-11, Volume: 51, Issue:23
Design of annulated pyrazoles as inhibitors of HIV-1 reverse transcriptase.
AID508786Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase V108I mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID508776Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase V179T mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1761008Selectivity index, ratio of CC50 for human TZM-bl cells to EC50 for wild type HIV1 NL4-3 infected in human TZM-bl cells2021European journal of medicinal chemistry, Feb-05, Volume: 211Design, synthesis, and evaluation of "dual-site"-binding diarylpyrimidines targeting both NNIBP and the NNRTI adjacent site of the HIV-1 reverse transcriptase.
AID1884541Fold resistance, ratio of EC50 for antiviral activity against drug-resistant HIV-1 Y188L mutant infected in human MT4 cells to EC50 for antiviral activity against NNRTI resistant wild type HIV-1 infected in human MT4 cells2022European journal of medicinal chemistry, Aug-05, Volume: 238Expansion of the S-CN-DABO scaffold to exploit the impact on inhibitory activities against the non-nucleoside HIV-1 reverse transcriptase.
AID522376Resistance index, ratio of EC50 for recombinant HIV1 harboring reverse transcriptase V179D mutant clone to EC50 for wild type HIV12010Antimicrobial agents and chemotherapy, Apr, Volume: 54, Issue:4
Combination of V106I and V179D polymorphic mutations in human immunodeficiency virus type 1 reverse transcriptase confers resistance to efavirenz and nevirapine but not etravirine.
AID1775797Antiviral activity against HIV-1 harboring RT L100I mutant infected in human MT4 cells assessed as protection against virus-induced cytopathogenicity after 5 days by MTT assay2021Journal of medicinal chemistry, 04-08, Volume: 64, Issue:7
2,4,5-Trisubstituted Pyrimidines as Potent HIV-1 NNRTIs: Rational Design, Synthesis, Activity Evaluation, and Crystallographic Studies.
AID1207430Inhibition of transient outward potassium current (Ito) current in Chinese Hamster Ovary (CHO) K1 cells expressing human Kv4.3 measured using IonWorks Quattro automated patch clamp platform
AID1298245Cytotoxicity against human MT4 cells after 5 days MTT assay2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
1,6-Bis[(benzyloxy)methyl]uracil derivatives-Novel antivirals with activity against HIV-1 and influenza H1N1 virus.
AID1352312Antiviral activity against wild type HIV1 3B infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 4 days by MTT assay2018European journal of medicinal chemistry, Feb-10, Volume: 145Discovery of biphenyl-substituted diarylpyrimidines as non-nucleoside reverse transcriptase inhibitors with high potency against wild-type and mutant HIV-1.
AID1636150Ratio of IC50 for NNRTI resistant HIV1 harboring reverse transcriptase L100I/K103N/H221Y mutant infected in human TZM-bl cells to IC50 for wild type HIV1 NL4-3 infected in human TZM-bl cells2016Journal of medicinal chemistry, Aug-11, Volume: 59, Issue:15
Discovery and Structure-Based Optimization of 2-Ureidothiophene-3-carboxylic Acids as Dual Bacterial RNA Polymerase and Viral Reverse Transcriptase Inhibitors.
AID665552Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 RES056 harboring reverse transcriptase K103N/Y181C RT mutant2012Bioorganic & medicinal chemistry, Jun-15, Volume: 20, Issue:12
Design, synthesis, anti-HIV evaluation and molecular modeling of piperidine-linked amino-triazine derivatives as potent non-nucleoside reverse transcriptase inhibitors.
AID1583028Antiviral activity against HIV1 containing reverse transcriptase E138K mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2020Journal of medicinal chemistry, 02-13, Volume: 63, Issue:3
Discovery and Characterization of Fluorine-Substituted Diarylpyrimidine Derivatives as Novel HIV-1 NNRTIs with Highly Improved Resistance Profiles and Low Activity for the hERG Ion Channel.
AID1483273Inhibition of HIV1 3B reverse transcriptase Y181C mutant infected in human MT4 cells assessed as protection against virus induced cytotoxicity after 5 days by MTT assay2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1572520Cytotoxicity against human MT4 cells assessed as reduction in cell viability after 5 days by MTT assay
AID1316332Antiviral activity against HIV1 RES056 harboring reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as protection against virus induced cytotoxicity after 5 days by MTT assay2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID1483278Cytotoxicity against human MT4 cells assessed as cell viability after 5 days by MTT assay2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1275546Antiviral activity against HIV1 3B expressing reverse transcriptase E138K mutant infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 5 days by MTT assay2016European journal of medicinal chemistry, Feb-15, Volume: 109Design, synthesis and anti-HIV evaluation of novel diarylpyridine derivatives targeting the entrance channel of NNRTI binding pocket.
AID614141Antiviral activity against wild type Human immunodeficiency virus 1 3B bearing reverse transcriptase K103N and Y181C double mutation infected in human MT4 cells assessed as protection against virus-induced cytopathogenicity after 4 days by MTT assay2011Bioorganic & medicinal chemistry, Sep-01, Volume: 19, Issue:17
Synthesis and structure-activity relationship of novel diarylpyrimidines with hydromethyl linker (CH(OH)-DAPYs) as HIV-1 NNRTIs.
AID557043Inhibition of HIV1 isolate R8 reverse transcriptase Y181C mutant after 90 mins by electrochemiluminescence analysis2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID1888706Inhibition of HIV1 reverse transcriptase assessed as inhibition of biotin-dUTP incorporation
AID635346Cytotoxicity against human MT4 cells measured by MTT assay2011Bioorganic & medicinal chemistry, Dec-01, Volume: 19, Issue:23
Design, synthesis and biological evaluation of cycloalkyl arylpyrimidines (CAPYs) as HIV-1 NNRTIs.
AID1275545Antiviral activity against HIV1 3B expressing reverse transcriptase Y181C mutant infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 5 days by MTT assay2016European journal of medicinal chemistry, Feb-15, Volume: 109Design, synthesis and anti-HIV evaluation of novel diarylpyridine derivatives targeting the entrance channel of NNRTI binding pocket.
AID449276Cytotoxicity against human erythrocytes assessed as change in internal complexity by flow cytometry2009Bioorganic & medicinal chemistry letters, Sep-15, Volume: 19, Issue:18
Addressing the malaria drug resistance challenge using flow cytometry to discover new antimalarials.
AID693584Antiviral activity against HIV1 RES056 harbouring RT K103N/Y181C mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2012European journal of medicinal chemistry, Dec, Volume: 58Synthesis, biological evaluation and molecular modeling of 4,6-diarylpyrimidines and diarylbenzenes as novel non-nucleosides HIV-1 reverse transcriptase inhibitors.
AID1520058Antiviral activity against HIV1 3B harbouring NNRTI K103N mutant infected in human MT4 cells assessed as virus-induced cytopathic effect incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Fragment hopping-based discovery of novel sulfinylacetamide-diarylpyrimidines (DAPYs) as HIV-1 nonnucleoside reverse transcriptase inhibitors.
AID276232Cytotoxicity against MT2 cells2006Bioorganic & medicinal chemistry letters, Nov-01, Volume: 16, Issue:21
Optimization of pyrimidinyl- and triazinyl-amines as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1775794Cytotoxicity in mock-infected human MT4 assessed as reduction in cell viability incubated for 5 days by MTT assay2021Journal of medicinal chemistry, 04-08, Volume: 64, Issue:7
2,4,5-Trisubstituted Pyrimidines as Potent HIV-1 NNRTIs: Rational Design, Synthesis, Activity Evaluation, and Crystallographic Studies.
AID1882475Antiviral activity against HIV-1 harboring K103N mutant infected in human MT4 cells assessed as protection against virus-induced cytopathicity by MTT assay2022Journal of medicinal chemistry, 03-10, Volume: 65, Issue:5
Contemporary Medicinal Chemistry Strategies for the Discovery and Development of Novel HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors.
AID1152374Selectivity index, ratio CC50 for human MT4 cells to EC50 for HIV 1 3B2014Bioorganic & medicinal chemistry, Jun-15, Volume: 22, Issue:12
Synthesis and biological evaluation of CHX-DAPYs as HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1446817Antiviral activity against HIV1 harboring Y181C mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect measured at 5 days post infection by MTT assay2017European journal of medicinal chemistry, Apr-21, Volume: 130Discovery of uracil-bearing DAPYs derivatives as novel HIV-1 NNRTIs via crystallographic overlay-based molecular hybridization.
AID1884223Selectivity index, ratio of CC50 for cytotoxicity against human MT4 cells to EC50 for antiviral activity against wildtype HIV-1 3B infected in human MT4 cells2022European journal of medicinal chemistry, Aug-05, Volume: 238Chemical space exploration around indolylarylsulfone scaffold led to a novel class of highly active HIV-1 NNRTIs with spiro structural features.
AID1687685Antiviral activity against HIV-2 ROD strain infected in human MT4 cells assessed as reduction in virus-induced cytopathogenicity by MTT assay2020European journal of medicinal chemistry, Jan-15, Volume: 186Indazolyl-substituted piperidin-4-yl-aminopyrimidines as HIV-1 NNRTIs: Design, synthesis and biological activities.
AID1483284Selectivity index, ratio of CC50 for human MT4 cells to EC50 for reverse transcriptase E138K mutant in human MT4 cells infected HIV1 3B2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID586906Inhibition of BCRP activity2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1558847Antiviral activity against HIV1 3B infected in human MT4 cells incubated assessed as reduction in virus-induced cytopathic effect for 5 days by MTT assay2019Journal of medicinal chemistry, 12-26, Volume: 62, Issue:24
Ligand-Based Design of Nondimethylphenyl-Diarylpyrimidines with Improved Metabolic Stability, Safety, and Oral Pharmacokinetic Profiles.
AID1731748Inhibition of reverse transcriptase Y183C mutant in HIV-1 infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay
AID518735Inhibition of RNA-dependent DNA polymerase activity of HIV1 subtype B reverse transcriptase M230L mutant by filter-based filtration assay2010Antimicrobial agents and chemotherapy, Jun, Volume: 54, Issue:6
The M230L nonnucleoside reverse transcriptase inhibitor resistance mutation in HIV-1 reverse transcriptase impairs enzymatic function and viral replicative capacity.
AID573468Inhibition of Human immunodeficiency virus 1 subtype B reverse transcriptase DNA-dependent DNA polymerase activity by gel-based primer extension assay2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID586891Induction of PXR mRNA expression in human LS180 cells at 0.1 to 1 umol/liter after 4 days by RT-PCR analysis2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID584083Antiviral activity against HIV1 NL4-3 harboring reverse transcriptase K103N mutant infected in human SupT1 cells assessed as inhibition of viral replication after 21 days relative to wild type HIV1 NL4-32010Antimicrobial agents and chemotherapy, Oct, Volume: 54, Issue:10
Lersivirine, a nonnucleoside reverse transcriptase inhibitor with activity against drug-resistant human immunodeficiency virus type 1.
AID522371Antiviral activity against recombinant HIV1 harboring reverse transcriptase V179D mutant clone infected in MAGIC-5 cells using 5-bromo4-chloro-3-indolyl-beta-D-galactopyranoside staining based light microscopy2010Antimicrobial agents and chemotherapy, Apr, Volume: 54, Issue:4
Combination of V106I and V179D polymorphic mutations in human immunodeficiency virus type 1 reverse transcriptase confers resistance to efavirenz and nevirapine but not etravirine.
AID508629Antiviral activity against Human immunodeficiency virus 1 subtype F isolate 93BR029 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID464765Antiviral activity against HIV1 harboring reverse transcriptase Y181C mutant infected in human MT2 cells2010Bioorganic & medicinal chemistry letters, Mar-01, Volume: 20, Issue:5
N1-Heterocyclic pyrimidinediones as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1231485Antiviral activity against wild-type HIV1 3B infected in human MT4 cells assessed as protection of cells from virus-induced cytopathic after 5 days by MTT assay2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Anti-HIV diarylpyrimidine-quinolone hybrids and their mode of action.
AID1773461Resistance factor, ratio of EC50 for HIV1 harboring E138K mutant infected in human MT4 cells to EC50 for HIV1 3B infected in MT4 cells
AID1457059Ratio of EC50 for HIV-1 harboring reverse transcriptase K103N mutant infected in human MT4 cells to EC50 for HIV1 NL4-3 infected in human MT4 cells2017Journal of medicinal chemistry, 08-10, Volume: 60, Issue:15
Chiral Indolylarylsulfone Non-Nucleoside Reverse Transcriptase Inhibitors as New Potent and Broad Spectrum Anti-HIV-1 Agents.
AID1304407Inhibition of HIV-1 BH10 recombinant reverse transcriptase K103N mutant expressed in Escherichia coli assessed as incorporation of [32P]GTP into poly(rA)/oligo(dT) as template primer2016Bioorganic & medicinal chemistry, 07-01, Volume: 24, Issue:13
Systematic evaluation of methyl ester bioisosteres in the context of developing alkenyldiarylmethanes (ADAMs) as non-nucleoside reverse transcriptase inhibitors (NNRTIs) for anti-HIV-1 chemotherapy.
AID491834Antiviral activity against wild type HIV1 3B infected in human MT2 cells assessed as inhibition of p24 antigen production after 4 days by ELISA2010Journal of medicinal chemistry, Jul-08, Volume: 53, Issue:13
Diarylaniline derivatives as a distinct class of HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID709863Selectivity ratio of EC50 for NNRTI-resistant HIV1 harboring reverse transcriptase Y181V mutant to EC50 for HIV1 expressing wild type reverse transcriptase2012Journal of medicinal chemistry, Dec-13, Volume: 55, Issue:23
Rational design of potent non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID586889Induction of SLCO1B3 mRNA expression in human LS180 cells at 0.1 to 1 umol/liter after 4 days by RT-PCR analysis2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID619636Antiviral activity against HIV-1 3B harboring RT L100I mutant infected in human MT4 cells assessed as inhibition of virus-induced syncytium formation after 5 days by MTT assay2011European journal of medicinal chemistry, Oct, Volume: 46, Issue:10
Arylazolylthioacetanilide. Part 8: Design, synthesis and biological evaluation of novel 2-(2-(2,4-dichlorophenyl)-2H-1,2,4-triazol-3-ylthio)-N-arylacetamides as potent HIV-1 inhibitors.
AID1822275Selectivity index, ratio of CC50 for cytotoxicity against human MT4 cells to EC50 for antiviral activity against HIV-1 IIIB infected in human MT4 cells2022Journal of medicinal chemistry, 02-10, Volume: 65, Issue:3
Discovery of Novel Pyridine-Dimethyl-Phenyl-DAPY Hybrids by Molecular Fusing of Methyl-Pyrimidine-DAPYs and Difluoro-Pyridinyl-DAPYs: Improving the Druggability toward High Inhibitory Activity, Solubility, Safety, and PK.
AID1171594Inhibition of wild type HIV1 reverse transcriptase K103N mutant assessed as reduction in enzyme activity2014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1761020Antiviral activity against HIV-1 harboring reverse transcriptase RES056 mutant infected in human MT4 cells assessed as inhibition of virus induced cytopathic effect measured after 5 days by MTT assay2021European journal of medicinal chemistry, Feb-05, Volume: 211Design, synthesis, and evaluation of "dual-site"-binding diarylpyrimidines targeting both NNIBP and the NNRTI adjacent site of the HIV-1 reverse transcriptase.
AID1884492Antiviral activity against drug-resistant HIV-1 Y188L mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Expansion of the S-CN-DABO scaffold to exploit the impact on inhibitory activities against the non-nucleoside HIV-1 reverse transcriptase.
AID1077216Inhibition of wild type HIV-1 reverse transcriptase using poly (rC)/oligo (dG) as template/primer assessed as inhibition of biotin-dUTP incorporation after 1 hr by microtiter plate ELISA reader analysis2014European journal of medicinal chemistry, Apr-09, Volume: 76Discovery of nitropyridine derivatives as potent HIV-1 non-nucleoside reverse transcriptase inhibitors via a structure-based core refining approach.
AID665553Fold resistance, ratio of EC50 for HIV1 RES056 harboring reverse transcriptase K103N/Y181C RT mutant to EC50 for wild type HIV1 3B2012Bioorganic & medicinal chemistry, Jun-15, Volume: 20, Issue:12
Design, synthesis, anti-HIV evaluation and molecular modeling of piperidine-linked amino-triazine derivatives as potent non-nucleoside reverse transcriptase inhibitors.
AID1731750Inhibition of reverse transcriptase E138K mutant in HIV-1 infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1884221Antiviral activity against wild type HIV-1 3B infected in human MT4 cells assessed as reduction in virus induced cytotoxicity incubated for 5 to 7 days by MTT assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Chemical space exploration around indolylarylsulfone scaffold led to a novel class of highly active HIV-1 NNRTIs with spiro structural features.
AID1561721Resistance index, ratio of EC50 for antiviral activity against HIV1 3B infected in human MT4 cells grown under 30 passages in presence of 4-[[4-[[4-(4-cyano-2,6-dimethylphenoxy)thieno[3,2-d]pyrimidin-2-yl]amino]-1-piperidyl]methyl]benzenesulfonamide to EC
AID1561702Antiviral activity against HIV1 expressing RT E138K mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1884542Fold resistance, ratio of EC50 for antiviral activity against drug-resistant HIV-1 E138K mutant infected in human MT4 cells to EC50 for antiviral activity against NNRTI resistant wild type HIV-1 infected in human MT4 cells2022European journal of medicinal chemistry, Aug-05, Volume: 238Expansion of the S-CN-DABO scaffold to exploit the impact on inhibitory activities against the non-nucleoside HIV-1 reverse transcriptase.
AID517498Clearance in human liver microsomes2010Bioorganic & medicinal chemistry letters, Oct-15, Volume: 20, Issue:20
Exploration of piperidine-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-Phenyl derivatives with broad potency against resistant mutant viruses.
AID1200844Antiviral activity against HIV1 RES056 expressing reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect after 5 days by MTT assay2015European journal of medicinal chemistry, Mar-26, Volume: 93Fused heterocycles bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 4: design, synthesis and biological evaluation of novel imidazo[1,2-a]pyrazines.
AID1235382Antiviral activity against HIV1 3B infected in human MT4 cells assessed as inhibition of virus-induced cytopathicity at 0.19 uM added 3 to 5 hrs post infection and measured 31 hrs post infection by p24 enzyme-linked immunosorbent assay2015Bioorganic & medicinal chemistry, Aug-01, Volume: 23, Issue:15
Hybrid chemistry. Part 4: Discovery of etravirine-VRX-480773 hybrids as potent HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID508756Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase V179D, Y181C mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1157595Antiviral activity against TMC125-resistant HIV1 harboring RT 109M, 138K, 190E mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathogenicity after 4 days by MTT assay2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Synthesis of novel fluoro analogues of MKC442 as microbicides.
AID508765Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase M236L mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1882468Antiviral activity against wild type HIV-1 IIIB infected in human MT2 cells assessed as protection against virus-induced cytopathicity by MTT assay2022Journal of medicinal chemistry, 03-10, Volume: 65, Issue:5
Contemporary Medicinal Chemistry Strategies for the Discovery and Development of Novel HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors.
AID1754648Inhibition of HIV-1 reverse transcriptase assessed as inhibition of biotin-dUTP incorporation into cDNA measured after 1 hr by ELISA2021Bioorganic & medicinal chemistry, 07-15, Volume: 42Exploiting the hydrophobic channel of the NNIBP: Discovery of novel diarylpyrimidines as HIV-1 NNRTIs against wild-type and K103N mutant viruses.
AID1357789Antiviral activity against HIV1 harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as inhibition of viral replication after 5 days by MTT assay2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID1171411Antiviral activity against wild type HIV1 3B infected in human MT2 cells assessed as protection from virus-induced cytopathicity by MTT assay2014ACS medicinal chemistry letters, Nov-13, Volume: 5, Issue:11
Picomolar Inhibitors of HIV-1 Reverse Transcriptase: Design and Crystallography of Naphthyl Phenyl Ethers.
AID1572062Antiviral activity against HIV1 harboring reverse transcriptase F227L/V106A double mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect after 5 days by MTT assay2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1561714Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV1 expressing RT F227L + V106A mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect
AID1248227Inhibition of recombinant HIV1 Reverse transcriptase p66/p51 using poly (rA)-oligo (dT) as template primer after 40 mins by spectrofluorometric analysis2015Bioorganic & medicinal chemistry, Oct-15, Volume: 23, Issue:20
A novel family of diarylpyrimidines (DAPYs) featuring a diatomic linker: Design, synthesis and anti-HIV activities.
AID1773453Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 harboring Y18IC mutant infected in human MT4 cells
AID490370Antiviral activity against HIV1 expressing reverse transcriptase K103N/L100I double mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect2010Bioorganic & medicinal chemistry letters, Jul-15, Volume: 20, Issue:14
Discovery of piperidin-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-benzyl derivatives with broad potency against resistant mutant viruses.
AID1126511Inhibition of wild-type HIV1 reverse transcriptase p66/p51 after 40 mins by spectrophotometry2014Bioorganic & medicinal chemistry, Apr-15, Volume: 22, Issue:8
Structural modifications of CH(OH)-DAPYs as new HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1446814Selectivity index, ratio of CC50 for human MT4 cells to EC50 for wild-type HIV1 3B infected in human MT4 cells2017European journal of medicinal chemistry, Apr-21, Volume: 130Discovery of uracil-bearing DAPYs derivatives as novel HIV-1 NNRTIs via crystallographic overlay-based molecular hybridization.
AID106061Inhibitory activity against 103N strain of HIV-I in MT-4 cells2001Bioorganic & medicinal chemistry letters, Sep-03, Volume: 11, Issue:17
Evolution of anti-HIV drug candidates. Part 3: Diarylpyrimidine (DAPY) analogues.
AID665275Selectivity index ratio of CC50 for human MT4 cells to EC50 for HIV1 3B2012European journal of medicinal chemistry, Jul, Volume: 53Chiral resolution, absolute configuration assignment and biological activity of racemic diarylpyrimidine CH(OH)-DAPY as potent nonnucleoside HIV-1 reverse transcriptase inhibitors.
AID1391079Cytotoxicity against human MT4 cells assessed as reduction in cell viability after 5 days by MTT assay2018Bioorganic & medicinal chemistry, 05-01, Volume: 26, Issue:8
The discovery of novel diarylpyri(mi)dine derivatives with high level activity against a wide variety of HIV-1 strains as well as against HIV-2.
AID1815392Resistance fold, ratio of EC50 for HIV1 harboring reverse transcriptase Y188L mutant infected MV4 cells to EC50 for wild type HIV1 infected MV4 cells2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1884220Antiviral activity against HIV-1 harboring F227L/V106A double mutant infected in human MT4 cells assessed as reduction in virus induced cytotoxicity incubated for 5 to 7 days by MTT assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Chemical space exploration around indolylarylsulfone scaffold led to a novel class of highly active HIV-1 NNRTIs with spiro structural features.
AID1352314Cytotoxicity against human MT4 cells assessed as reduction in cell viability after 4 days by MTT assay2018European journal of medicinal chemistry, Feb-10, Volume: 145Discovery of biphenyl-substituted diarylpyrimidines as non-nucleoside reverse transcriptase inhibitors with high potency against wild-type and mutant HIV-1.
AID541246Antiviral activity against HIV1 3B infected in human MT2 cells assessed as reduction in p24 antigen level after 4 days by ELISA2010Journal of medicinal chemistry, Dec-09, Volume: 53, Issue:23
Design, synthesis, and evaluation of diarylpyridines and diarylanilines as potent non-nucleoside HIV-1 reverse transcriptase inhibitors.
AID417448Inhibition of HIV1 reverse transcriptase Y188L mutant2009European journal of medicinal chemistry, Feb, Volume: 44, Issue:2
QSAR studies for diarylpyrimidines against HIV-1 reverse transcriptase wild-type and mutant strains.
AID1565097Antiviral activity against drug resistant HIV-1 harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1558861Aqueous solubility of compound at 2 mg measured for 24 hrs by HPLC analysis2019Journal of medicinal chemistry, 12-26, Volume: 62, Issue:24
Ligand-Based Design of Nondimethylphenyl-Diarylpyrimidines with Improved Metabolic Stability, Safety, and Oral Pharmacokinetic Profiles.
AID1446829Solubility of the compound in water at pH 7.42017European journal of medicinal chemistry, Apr-21, Volume: 130Discovery of uracil-bearing DAPYs derivatives as novel HIV-1 NNRTIs via crystallographic overlay-based molecular hybridization.
AID1691435Antiviral activity against HIV1 3B infected in human MT4 cells assessed as reduction in virus-induced cell death incubated for 5 days by MTT assay2020European journal of medicinal chemistry, May-01, Volume: 193In situ click chemistry-based rapid discovery of novel HIV-1 NNRTIs by exploiting the hydrophobic channel and tolerant regions of NNIBP.
AID1410483Cytotoxicity against human MT4 cells assessed as reduction in cell viability by MTT assay2018ACS medicinal chemistry letters, Apr-12, Volume: 9, Issue:4
Further Exploring Solvent-Exposed Tolerant Regions of Allosteric Binding Pocket for Novel HIV-1 NNRTIs Discovery.
AID1574382Resistance index, ratio of EC50 for HIV-1 harboring reverse transcriptase K103N mutant to EC50 for wild type HIV-1 NL4-32019Journal of medicinal chemistry, 01-24, Volume: 62, Issue:2
Effect of α-Methoxy Substitution on the Anti-HIV Activity of Dihydropyrimidin-4(3 H)-ones.
AID1773448Antiviral activity against wild type HIV1 harboring Y188L mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathogenic effect by MTT assay
AID1705197Inhibition of RNA-dependent DNA polymerase activity of recombinant HIV-1 p66/p51 reverse transcriptase K103N/Y181C mutant assessed as inhibition of [3H]dTTP incorporation using poly(rA)/oligo(dT) as templates incubated for 15 mins by MicroBeta scintillati2020European journal of medicinal chemistry, Dec-15, Volume: 208New indolylarylsulfone non-nucleoside reverse transcriptase inhibitors show low nanomolar inhibition of single and double HIV-1 mutant strains.
AID1761006Antiviral activity against wild type HIV1 NL4-3 infected in human TZM-bl cells assessed as reduction in viral infection measured after 1 day by luciferase reporter gene assay based luminiscence assay2021European journal of medicinal chemistry, Feb-05, Volume: 211Design, synthesis, and evaluation of "dual-site"-binding diarylpyrimidines targeting both NNIBP and the NNRTI adjacent site of the HIV-1 reverse transcriptase.
AID508650Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase L100I, K103N, Y181C mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID496629Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase E138A/K20R, V35T, E36D, T39N, V60I, D123N, I135V, S162A, K173T, Q174K, D177E, T200A, Q207E, R211K mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID508774Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase Y181I mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1171414Cytotoxicity against human MT2 cells assessed as reduction in cell viability by MTT assay2014ACS medicinal chemistry letters, Nov-13, Volume: 5, Issue:11
Picomolar Inhibitors of HIV-1 Reverse Transcriptase: Design and Crystallography of Naphthyl Phenyl Ethers.
AID508638Antiviral activity against Human immunodeficiency virus 1 subtype F1 infected in human MT4 cells assessed as reduction in virus induced cytopathicity2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1207524Inhibition of rapid delayed inward rectifying potassium current (IKr) measured using manual patch clamp assay
AID1207281Inhibition of long-lasting type calcium current (ICaL) in HEK293 cells (alpha1C/beta2a/alpha2delta1) cells measured using IonWorks Barracuda automated patch clamp platform
AID1443666Antiviral activity against HIV1 harboring reverse transcriptase L100I mutant infected in human MT4 cells assessed as inhibition of viral replication after 5 days by MTT assay2017Bioorganic & medicinal chemistry letters, 04-15, Volume: 27, Issue:8
Design and synthesis of hybrids of diarylpyrimidines and diketo acids as HIV-1 inhibitors.
AID1754642Antiviral activity against HIV1 harboring K103N mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect incubated for 5 days by MTT assay2021Bioorganic & medicinal chemistry, 07-15, Volume: 42Exploiting the hydrophobic channel of the NNIBP: Discovery of novel diarylpyrimidines as HIV-1 NNRTIs against wild-type and K103N mutant viruses.
AID1124998Inhibition of recombinant HIV1 reverse transcriptase assessed as inhibition of biotin-dUTP incorporation in to poly [A] x oligo[dT]15 by ELISA2014Bioorganic & medicinal chemistry, Apr-01, Volume: 22, Issue:7
Fused heterocyclic compounds bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 1: design, synthesis and biological evaluation of novel 5,7-disubstituted pyrazolo[1,5-a]pyrimidine derivatives.
AID1687687Selectivity index, ratio of CC50 for cytotoxicity against human MT4 cells reduction in cell viability by MTT assay to EC50 for antiviral activity against wild type HIV-1 strain IIIB infected in human MT4 cells assessed as reduction in virus-induced cytopa2020European journal of medicinal chemistry, Jan-15, Volume: 186Indazolyl-substituted piperidin-4-yl-aminopyrimidines as HIV-1 NNRTIs: Design, synthesis and biological activities.
AID738334Antiviral activity against HIV1 RES056 harboring reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as protection against virus-induced cytopathicity after 5 days by MTT assay2013Bioorganic & medicinal chemistry, Apr-01, Volume: 21, Issue:7
Synthesis and biological evaluation of pyridazine derivatives as novel HIV-1 NNRTIs.
AID1743640Inhibition of HIV1 reverse transcriptase assessed as reduction in biotin-dUTP incorporation into template preincubated for 1 hr followed by template/primer and dNTP addition and measured after 1 hr by ELISA2020European journal of medicinal chemistry, Nov-15, Volume: 206Targeting dual tolerant regions of binding pocket: Discovery of novel morpholine-substituted diarylpyrimidines as potent HIV-1 NNRTIs with significantly improved water solubility.
AID417449Hydrophobic index, Log P of the compound2009European journal of medicinal chemistry, Feb, Volume: 44, Issue:2
QSAR studies for diarylpyrimidines against HIV-1 reverse transcriptase wild-type and mutant strains.
AID1773458Resistance factor, ratio of EC50 for HIV1 harboring K103N mutant infected in human MT4 cells to EC50 for HIV1 3B infected in MT4 cells
AID533674Trough drug concentration at 12 hrs in healthy human plasma at 200 mg, po twice a day for 8 days by reverse-phase HPLC-MS method2008Antimicrobial agents and chemotherapy, Dec, Volume: 52, Issue:12
Minimal pharmacokinetic interaction between the human immunodeficiency virus nonnucleoside reverse transcriptase inhibitor etravirine and the integrase inhibitor raltegravir in healthy subjects.
AID1456307Inhibition of HIV-1 reverse transcriptase E138K mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect measured at 5 days post infection by MTT assay2017Bioorganic & medicinal chemistry, 04-15, Volume: 25, Issue:8
Structural modifications of diarylpyrimidines (DAPYs) as HIV-1 NNRTIs: Synthesis, anti-HIV activities and SAR.
AID1574383Resistance index, ratio of EC50 for HIV-1 harboring reverse transcriptase K103N/Y181C double mutant to EC50 for wild type HIV-1 NL4-32019Journal of medicinal chemistry, 01-24, Volume: 62, Issue:2
Effect of α-Methoxy Substitution on the Anti-HIV Activity of Dihydropyrimidin-4(3 H)-ones.
AID1731751Inhibition of reverse transcriptase F227L/V106A double mutant in HIV-1 infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay
AID465422Half life in Beagle dog at 0.5 mg/kg, iv or. 5 mg/kg, po2010Bioorganic & medicinal chemistry letters, Mar-01, Volume: 20, Issue:5
N1-Alkyl pyrimidinediones as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID449194Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 3B2009Bioorganic & medicinal chemistry letters, Sep-15, Volume: 19, Issue:18
Discovery of diarylpyridine derivatives as novel non-nucleoside HIV-1 reverse transcriptase inhibitors.
AID496742Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase E138A/V35T, K122E, I135V, T139A, S162A, K173T, Q174K, N175Y, D177E, T200A, Q207E, R211K mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID1171581Antiviral activity against HIV1 3B infected in human CEM cells assessed as protection against virus-induced cytopathicity by giant cell formation assay2014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID496626Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase E138A/K22N, V35T, T39A, K122E, I135V, T139A, S162A, K173T, Q174A, D177E, T200A, Q207E, R211K, F214L mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID1141977Inhibition of HIV1 wild-type reverse transcriptase Y181I/Y181C mutant using [3H]dTTP by scintillation counting2014European journal of medicinal chemistry, Jun-10, Volume: 80New indolylarylsulfones as highly potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1435518Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 RES056 expressing reverse transcriptase mutant
AID1298250Inhibition of HIV1 reverse transcriptase p66/p51 V106A mutant using poly(rA) template and measured after 40 mins by picogreen based spectrofluorometry2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
1,6-Bis[(benzyloxy)methyl]uracil derivatives-Novel antivirals with activity against HIV-1 and influenza H1N1 virus.
AID1357786Selectivity index, ratio of CC50 for human MT4 cells to EC50 for wild-type HIV1 3B2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID1435511Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 3B expressing wild type reverse transcriptase
AID1326644Cytotoxicity against human MT4 cells assessed as decrease in cell viability after 5 days by cell titer glo based luciferase reporter gene assay2016European journal of medicinal chemistry, Oct-21, Volume: 122Novel (2,6-difluorophenyl)(2-(phenylamino)pyrimidin-4-yl)methanones with restricted conformation as potent non-nucleoside reverse transcriptase inhibitors against HIV-1.
AID522370Antiviral activity against recombinant HIV1 harboring reverse transcriptase V106I mutant clone infected in MAGIC-5 cells using 5-bromo4-chloro-3-indolyl-beta-D-galactopyranoside staining based light microscopy2010Antimicrobial agents and chemotherapy, Apr, Volume: 54, Issue:4
Combination of V106I and V179D polymorphic mutations in human immunodeficiency virus type 1 reverse transcriptase confers resistance to efavirenz and nevirapine but not etravirine.
AID1572519Antiviral activity against HIV1 RES056 harboring reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as inhibition of virus-induced cytotoxicity after 5 days by MTT assay
AID1504699Antiviral activity against HIV1 3B harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2017ACS medicinal chemistry letters, Nov-09, Volume: 8, Issue:11
Discovery of Thiophene[3,2-
AID1811050Oral bioavailability in rat2021Journal of medicinal chemistry, 07-22, Volume: 64, Issue:14
Improving Druggability of Novel Diarylpyrimidine NNRTIs by a Fragment-Based Replacement Strategy: From Biphenyl-DAPYs to Heteroaromatic-Biphenyl-DAPYs.
AID586884Induction of CYP2B6 mRNA expression in human LS180 cells at 0.1 to 1 umol/liter after 4 days by RT-PCR analysis2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1141975Inhibition of HIV1 wild-type reverse transcriptase using [3H]dTTP by scintillation counting2014European journal of medicinal chemistry, Jun-10, Volume: 80New indolylarylsulfones as highly potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1249723Antiviral activity against HIV1 expressing reverse transcriptase F227L + V106A mutant2015European journal of medicinal chemistry, Sep-18, Volume: 102Pyrimidine sulfonylacetanilides with improved potency against key mutant viruses of HIV-1 by specific targeting of a highly conserved residue.
AID1729161Antiviral activity against HIV1 harboring F227L/V106A mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect by MTT assay2021European journal of medicinal chemistry, Mar-05, Volume: 213Exploiting the tolerant region I of the non-nucleoside reverse transcriptase inhibitor (NNRTI) binding pocket. Part 2: Discovery of diarylpyrimidine derivatives as potent HIV-1 NNRTIs with high Fsp
AID573480Fold resistance, ratio of EC50 for Human immunodeficiency virus 1 isolate 8116 harboring A98S, G190A mutation in reverse transcriptase to wild-type2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID508623Antiviral activity against Human immunodeficiency virus 1 subtype D isolate 92UG035 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1596752Antiviral activity against HIV-1 3B harboring reverse transcriptase E138K mutant infected in MT4 cells measured after 5 days by MTT assay2019European journal of medicinal chemistry, Aug-15, Volume: 176Synthesis and biological evaluation of dihydroquinazoline-2-amines as potent non-nucleoside reverse transcriptase inhibitors of wild-type and mutant HIV-1 strains.
AID757627Inhibition of HIV RT K103N/Y181C mutant by cell based assay2013European journal of medicinal chemistry, Jul, Volume: 65Molecular design, synthesis and biological evaluation of BP-O-DAPY and O-DAPY derivatives as non-nucleoside HIV-1 reverse transcriptase inhibitors.
AID1316341Antiviral activity against HIV1 3B harboring reverse transcriptase F227L/V106A double mutant infected in human MT4 cells assessed as protection against virus induced cytotoxicity after 5 days by MTT assay2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID1741398Inhibition of reverse transcriptase F227L and V106A mutant in HIV-1 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Sep-15, Volume: 202Bioisosterism-based design and enantiomeric profiling of chiral hydroxyl-substituted biphenyl-diarylpyrimidine nonnucleoside HIV-1 reverse transcriptase inhibitors.
AID1637397Antiviral activity against human HIV-1 3B harboring reverse transcriptase E138K mutant infected in human MT4 cells assessed as protection from virus-induced cytopathic effect measured 5 days post infection by MTT assay2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Arylazolyl(azinyl)thioacetanilides. Part 20: Discovery of novel purinylthioacetanilides derivatives as potent HIV-1 NNRTIs via a structure-based bioisosterism approach.
AID1157585Antiviral activity against zidovudine-resistant HIV1 harboring RT 67N, 70R, 215F, 219Q mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathogenicity after 4 days by MTT assay2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Synthesis of novel fluoro analogues of MKC442 as microbicides.
AID1390715Resistance index, ratio of EC50 for HIV1 RES056 harboring K103N/Y181C double mutant infected in human MT4 cells to EC50 for wild-type HIV1 3B infected in human MT4 cells2018Bioorganic & medicinal chemistry letters, 05-01, Volume: 28, Issue:8
First discovery of a potential carbonate prodrug of NNRTI drug candidate RDEA427 with submicromolar inhibitory activity against HIV-1 K103N/Y181C double mutant strain.
AID1357785Cytotoxicity against human MT4 cells assessed as decrease in cell viability after 5 days by MTT assay2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID1231487Inhibition of wild-type HIV1 Reverse transcriptase p66/p51 assessed as relative fluorescence signal after 40 mins2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Anti-HIV diarylpyrimidine-quinolone hybrids and their mode of action.
AID1390711Antiviral activity against HIV1 RES056 expressing reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay2018Bioorganic & medicinal chemistry letters, 05-01, Volume: 28, Issue:8
First discovery of a potential carbonate prodrug of NNRTI drug candidate RDEA427 with submicromolar inhibitory activity against HIV-1 K103N/Y181C double mutant strain.
AID508790Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase K103N mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID508829Antiviral activity against Human immunodeficiency virus 1 subtype B isolate JR-CSF infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1773450Antiviral activity against HIV1 harboring F227L/V106A mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathogenic effect by MTT assay
AID1483282Selectivity index, ratio of CC50 for human MT4 cells to EC50 for reverse transcriptase Y181C mutant in human MT4 cells infected HIV1 3B2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1141979Inhibition of HIV1 wild-type reverse transcriptase V106A mutant using [3H]dTTP by scintillation counting2014European journal of medicinal chemistry, Jun-10, Volume: 80New indolylarylsulfones as highly potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1352315Selectivity index, ratio of CC50 for human MT4 cells to EC50 for wild type HIV1 3B2018European journal of medicinal chemistry, Feb-10, Volume: 145Discovery of biphenyl-substituted diarylpyrimidines as non-nucleoside reverse transcriptase inhibitors with high potency against wild-type and mutant HIV-1.
AID1072805Antiviral activity against HIV1 harboring reverse transcriptase K101N/Y181C double mutant infected in human MT4 cells2014Bioorganic & medicinal chemistry, Mar-15, Volume: 22, Issue:6
Discovery of 2-pyridone derivatives as potent HIV-1 NNRTIs using molecular hybridization based on crystallographic overlays.
AID1773827Antiviral activity against HIV1 RES056 infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2021Journal of medicinal chemistry, 09-23, Volume: 64, Issue:18
Structure-Based Design and Discovery of Pyridyl-Bearing Fused Bicyclic HIV-1 Inhibitors: Synthesis, Biological Characterization, and Molecular Modeling Studies.
AID1884228Antiviral activity against HIV-1 harboring E138K mutant infected in human MT4 cells assessed as reduction in virus induced cytotoxicity incubated for 5 to 7 days by MTT assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Chemical space exploration around indolylarylsulfone scaffold led to a novel class of highly active HIV-1 NNRTIs with spiro structural features.
AID1773829Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for HIV1 3B infected in human MT4 cells2021Journal of medicinal chemistry, 09-23, Volume: 64, Issue:18
Structure-Based Design and Discovery of Pyridyl-Bearing Fused Bicyclic HIV-1 Inhibitors: Synthesis, Biological Characterization, and Molecular Modeling Studies.
AID1390714Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 RES056 expressing reverse transcriptase K103N/Y181C double mutant2018Bioorganic & medicinal chemistry letters, 05-01, Volume: 28, Issue:8
First discovery of a potential carbonate prodrug of NNRTI drug candidate RDEA427 with submicromolar inhibitory activity against HIV-1 K103N/Y181C double mutant strain.
AID1574373Cytotoxicity against human MT4 cells assessed as reduction in cell viability after 5 days by MTT assay2019Journal of medicinal chemistry, 01-24, Volume: 62, Issue:2
Effect of α-Methoxy Substitution on the Anti-HIV Activity of Dihydropyrimidin-4(3 H)-ones.
AID1773837Antiviral activity against HIV1 harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2021Journal of medicinal chemistry, 09-23, Volume: 64, Issue:18
Structure-Based Design and Discovery of Pyridyl-Bearing Fused Bicyclic HIV-1 Inhibitors: Synthesis, Biological Characterization, and Molecular Modeling Studies.
AID347628Antiviral activity against NNRTI-resistant HIV HXB2 isolate 17 with reverse transcriptase A98, K103, Y181 mutation in human MT4 cells assessed as protection against virus-induced cell death after 5 days by MTT assay2008Journal of medicinal chemistry, Dec-11, Volume: 51, Issue:23
Design of annulated pyrazoles as inhibitors of HIV-1 reverse transcriptase.
AID1152370Antiviral activity against HIV1 3B infected in human MT4 cells assessed as inhibition of virus-induced cytopathicity measured 5 days post viral infection by MTT assay2014Bioorganic & medicinal chemistry, Jun-15, Volume: 22, Issue:12
Synthesis and biological evaluation of CHX-DAPYs as HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1435525Inhibition of recombinant wild type HIV1 reverse transcriptase using poly (A).oligo (dT)15 as template/primer assessed as decrease in biotin-dUTP incorporation after 1 hr by ELISA
AID1743628Antiviral activity against HIV1 harboring RT Y181C mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Nov-15, Volume: 206Targeting dual tolerant regions of binding pocket: Discovery of novel morpholine-substituted diarylpyrimidines as potent HIV-1 NNRTIs with significantly improved water solubility.
AID1191400Antiviral activity against HIV-1 RES056 harboring reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 5 days by MTT assay2015Bioorganic & medicinal chemistry, Feb-01, Volume: 23, Issue:3
Synthesis and biological evaluation of DAPY-DPEs hybrids as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1819215Antiviral activity against HIV-1 harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1171595Inhibition of wild type HIV1 reverse transcriptase Y181I/Y181C mutant assessed as reduction in enzyme activity2014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID496617Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase V90I/V35T, E36D, T39K, I50V, V60I, S68G, I135V, S162A, K173V, Q174K,D177E, T200A, E203Q, Q207D, R211K mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID522368Antiviral activity against wild type HIV1 infected in MAGIC-5 cells using 5-bromo4-chloro-3-indolyl-beta-D-galactopyranoside staining based light microscopy2010Antimicrobial agents and chemotherapy, Apr, Volume: 54, Issue:4
Combination of V106I and V179D polymorphic mutations in human immunodeficiency virus type 1 reverse transcriptase confers resistance to efavirenz and nevirapine but not etravirine.
AID648425Inhibition of HIV1 reverse transcriptase assessed as incorporation of biotin-dUTP into poly(rA)-oligo(dT)2012European journal of medicinal chemistry, May, Volume: 51Synthesis and biological evaluation of piperidine-substituted triazine derivatives as HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1320869Inhibition of recombinant wild type HIV1 p66/p51 using poly(rA) template/oligo(dT)16 primer after 40 mins by PicoGreen-based spectrofluorometric method2016European journal of medicinal chemistry, Oct-04, Volume: 121Structural optimization of pyridine-type DAPY derivatives to exploit the tolerant regions of the NNRTI binding pocket.
AID1636927Antiviral activity against HIV1 3B infected in human MT2 cells assessed as protection against viral infection by MTT method2016Bioorganic & medicinal chemistry, 10-15, Volume: 24, Issue:20
Computer-aided discovery of anti-HIV agents.
AID1572066Selectivity ratio of CC50 for human MT4 cells to EC50 for HIV1 harboring reverse transcriptase Y188L mutant infected in human MT4 cells2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1446819Antiviral activity against HIV1 harboring E138K mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect measured at 5 days post infection by MTT assay2017European journal of medicinal chemistry, Apr-21, Volume: 130Discovery of uracil-bearing DAPYs derivatives as novel HIV-1 NNRTIs via crystallographic overlay-based molecular hybridization.
AID1637390Cytotoxicity against mock-infected human MT4 cells assessed as reduction of cell viability after 5 days by MTT assay2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Arylazolyl(azinyl)thioacetanilides. Part 20: Discovery of novel purinylthioacetanilides derivatives as potent HIV-1 NNRTIs via a structure-based bioisosterism approach.
AID1822282Antiviral activity against HIV-1 harboring RES056 mutant infected in human MT4 cells assessed as protection against virus-induced cytotoxicity incubated for 5 days by MTT assay2022Journal of medicinal chemistry, 02-10, Volume: 65, Issue:3
Discovery of Novel Pyridine-Dimethyl-Phenyl-DAPY Hybrids by Molecular Fusing of Methyl-Pyrimidine-DAPYs and Difluoro-Pyridinyl-DAPYs: Improving the Druggability toward High Inhibitory Activity, Solubility, Safety, and PK.
AID508652Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase K101P, K103N, V108I mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID347635Antiviral activity against NNRTI-resistant HIV HXB2 isolate 36 with reverse transcriptase K103, V106, Y181 mutation in human MT4 cells assessed as protection against virus-induced cell death after 5 days by MTT assay2008Journal of medicinal chemistry, Dec-11, Volume: 51, Issue:23
Design of annulated pyrazoles as inhibitors of HIV-1 reverse transcriptase.
AID246301Effective concentration of the compound to inhibit HIV-1 mutant Y188L replication in HIV-infected MT-4 cells2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
From 4,5,6,7-tetrahydro-5-methylimidazo[4,5,1-jk](1,4)benzodiazepin-2(1H)-one (TIBO) to etravirine (TMC125): fifteen years of research on non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1862562Antiviral activity against HIV-1 ROD infected in human MT4 cells assessed as reduction in viral replication by MTT assay
AID1822277Antiviral activity against HIV-1 harboring K103N mutant infected in human MT4 cells assessed as protection against virus-induced cytotoxicity incubated for 5 days by MTT assay2022Journal of medicinal chemistry, 02-10, Volume: 65, Issue:3
Discovery of Novel Pyridine-Dimethyl-Phenyl-DAPY Hybrids by Molecular Fusing of Methyl-Pyrimidine-DAPYs and Difluoro-Pyridinyl-DAPYs: Improving the Druggability toward High Inhibitory Activity, Solubility, Safety, and PK.
AID1421325Inhibition of wild-type HIV1 3B reverse transcriptase infected in human MT4 cells assessed as protection against virus-induced cytopathic effect measured 5 days post infection by MTT assay2018European journal of medicinal chemistry, Oct-05, Volume: 158Advances in diarylpyrimidines and related analogues as HIV-1 nonnucleoside reverse transcriptase inhibitors.
AID557030Ratio of EC95 for HIV1 isolate R8 harboring reverse transcriptase Y181C mutant in presence of 50% human serum to EC95 for HIV1 isolate R8 harboring wild type reverse transcriptase in presence of 50% human serum2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID1815390Selectivity index, ratio of CC50 for cytotoxicity against human MT4 cells to EC50 of Anti-HIV activity against HIV-1 IIIB infected in human MT42021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID541245Cytotoxicity in human TZM-bl cells after 4 day by XTT assay2010Journal of medicinal chemistry, Dec-09, Volume: 53, Issue:23
Design, synthesis, and evaluation of diarylpyridines and diarylanilines as potent non-nucleoside HIV-1 reverse transcriptase inhibitors.
AID1811034Anti-viral activity against HIV1 harboring RT E138K mutant infected MV4 cells assessed as protection against virus-induced cytopathic effect by MTT assay2021Journal of medicinal chemistry, 07-22, Volume: 64, Issue:14
Improving Druggability of Novel Diarylpyrimidine NNRTIs by a Fragment-Based Replacement Strategy: From Biphenyl-DAPYs to Heteroaromatic-Biphenyl-DAPYs.
AID1763906Cytotoxicity against human MT4 cells assessed as reduction in cell viability after 5 days by MTT assay2021Bioorganic & medicinal chemistry, 06-15, Volume: 40Design, synthesis and anti-HIV evaluation of novel 5-substituted diarylpyrimidine derivatives as potent HIV-1 NNRTIs.
AID1565096Antiviral activity against drug resistant HIV-1 harboring reverse transcriptase L100I mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1435502Antiviral activity against HIV1 3B expressing wild type reverse transcriptase infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay
AID1750715Cytotoxicity against in human MT-4 cells after 5 days by MTT assay2021Journal of medicinal chemistry, 04-22, Volume: 64, Issue:8
Hydrophobic Pocket Occupation Design of Difluoro-Biphenyl-Diarylpyrimidines as Non-Nucleoside HIV-1 Reverse Transcriptase Inhibitors: from
AID1124993Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV2 ROD2014Bioorganic & medicinal chemistry, Apr-01, Volume: 22, Issue:7
Fused heterocyclic compounds bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 1: design, synthesis and biological evaluation of novel 5,7-disubstituted pyrazolo[1,5-a]pyrimidine derivatives.
AID1316359Inhibition of recombinant HIV1 reverse transcriptase K103N/Y181C double mutant using DIG-dUTP/biotin-dUTP/dTTP assessed as suppression of biotin-dUTP incorporation after 1 hr by ELISA2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID1357795Resistance factor, ratio of EC50 for HIV1 harboring reverse transcriptase K103N mutant to EC50 for wild-type HIV-1 3B2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID1443669Antiviral activity against HIV1 harboring reverse transcriptase E138K mutant infected in human MT4 cells assessed as inhibition of viral replication after 5 days by MTT assay2017Bioorganic & medicinal chemistry letters, 04-15, Volume: 27, Issue:8
Design and synthesis of hybrids of diarylpyrimidines and diketo acids as HIV-1 inhibitors.
AID1574380Resistance index, ratio of EC50 for HIV-1 harboring reverse transcriptase Y181C mutant to EC50 for wild type HIV-1 NL4-32019Journal of medicinal chemistry, 01-24, Volume: 62, Issue:2
Effect of α-Methoxy Substitution on the Anti-HIV Activity of Dihydropyrimidin-4(3 H)-ones.
AID1275554Selectivity index, ratio of CC50 for human MT4 cells to EC50 for wild type HIV2 ROD infected in human MT4 cells2016European journal of medicinal chemistry, Feb-15, Volume: 109Design, synthesis and anti-HIV evaluation of novel diarylpyridine derivatives targeting the entrance channel of NNRTI binding pocket.
AID508772Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase Y188L mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1750717Inhibition of HIV-1 p66/p51 reverse transcriptase L100I mutant incubated for 40 mins by picogreen dye-based spectrofluorometric analysis2021Journal of medicinal chemistry, 04-22, Volume: 64, Issue:8
Hydrophobic Pocket Occupation Design of Difluoro-Biphenyl-Diarylpyrimidines as Non-Nucleoside HIV-1 Reverse Transcriptase Inhibitors: from
AID83549Potency evaluated against NNRTI-Resistant HIV-1 strain Leu100Ile2004Journal of medicinal chemistry, May-06, Volume: 47, Issue:10
Roles of conformational and positional adaptability in structure-based design of TMC125-R165335 (etravirine) and related non-nucleoside reverse transcriptase inhibitors that are highly potent and effective against wild-type and drug-resistant HIV-1 varian
AID1391077Antiviral activity against wild type HIV1 3B infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay2018Bioorganic & medicinal chemistry, 05-01, Volume: 26, Issue:8
The discovery of novel diarylpyri(mi)dine derivatives with high level activity against a wide variety of HIV-1 strains as well as against HIV-2.
AID522372Antiviral activity against recombinant HIV1 harboring reverse transcriptase V106A/V179D mutant clone infected in MAGIC-5 cells using 5-bromo4-chloro-3-indolyl-beta-D-galactopyranoside staining based light microscopy2010Antimicrobial agents and chemotherapy, Apr, Volume: 54, Issue:4
Combination of V106I and V179D polymorphic mutations in human immunodeficiency virus type 1 reverse transcriptase confers resistance to efavirenz and nevirapine but not etravirine.
AID1435515Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 expressing reverse transcriptase Y188L mutant
AID1390710Antiviral activity against HIV1 3B expressing wild type reverse transcriptase infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay2018Bioorganic & medicinal chemistry letters, 05-01, Volume: 28, Issue:8
First discovery of a potential carbonate prodrug of NNRTI drug candidate RDEA427 with submicromolar inhibitory activity against HIV-1 K103N/Y181C double mutant strain.
AID1773835Antiviral activity against HIV1 harboring reverse transcriptase E138K mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2021Journal of medicinal chemistry, 09-23, Volume: 64, Issue:18
Structure-Based Design and Discovery of Pyridyl-Bearing Fused Bicyclic HIV-1 Inhibitors: Synthesis, Biological Characterization, and Molecular Modeling Studies.
AID1815393Resistance fold, ratio of EC50 for HIV1 harboring reverse transcriptase E138K mutant infected MV4 cells to EC50 for wild type HIV1 infected MV4 cells2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1304406Inhibition of wild type HIV-1 BH10 recombinant reverse transcriptase expressed in Escherichia coli assessed as incorporation of [32P]GTP into poly(rA)/oligo(dT) as template primer2016Bioorganic & medicinal chemistry, 07-01, Volume: 24, Issue:13
Systematic evaluation of methyl ester bioisosteres in the context of developing alkenyldiarylmethanes (ADAMs) as non-nucleoside reverse transcriptase inhibitors (NNRTIs) for anti-HIV-1 chemotherapy.
AID1352318Antiviral activity against HIV1 harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 4 days by MTT assay2018European journal of medicinal chemistry, Feb-10, Volume: 145Discovery of biphenyl-substituted diarylpyrimidines as non-nucleoside reverse transcriptase inhibitors with high potency against wild-type and mutant HIV-1.
AID1754639Selectivity index, ratio of CC50 for human MT4 cells to EC50 for Antiviral activity against HIV-1 IIIB infected in human MT4 cells2021Bioorganic & medicinal chemistry, 07-15, Volume: 42Exploiting the hydrophobic channel of the NNIBP: Discovery of novel diarylpyrimidines as HIV-1 NNRTIs against wild-type and K103N mutant viruses.
AID1561701Antiviral activity against HIV1 expressing RT Y188L mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay
AID541253Resistance index, ratio of EC50 for multidrug resistant HIV1 A17 to EC50 for wild type HIV1 3B2010Journal of medicinal chemistry, Dec-09, Volume: 53, Issue:23
Design, synthesis, and evaluation of diarylpyridines and diarylanilines as potent non-nucleoside HIV-1 reverse transcriptase inhibitors.
AID1583020Antiviral activity against NNRTI-resistant HIV1 RES056 containing reverse transcriptase K103N+Y181C mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2020Journal of medicinal chemistry, 02-13, Volume: 63, Issue:3
Discovery and Characterization of Fluorine-Substituted Diarylpyrimidine Derivatives as Novel HIV-1 NNRTIs with Highly Improved Resistance Profiles and Low Activity for the hERG Ion Channel.
AID496615Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase V90I/V35T, K64R, K102Q, D123E, S162A, K173T, Q174K, D177E, G196E,T200A, Q207E, R211K mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID1761011Cytotoxicity against human MT4 cells assessed as reduction in cell viability measured after 5 days by MTT assay2021European journal of medicinal chemistry, Feb-05, Volume: 211Design, synthesis, and evaluation of "dual-site"-binding diarylpyrimidines targeting both NNIBP and the NNRTI adjacent site of the HIV-1 reverse transcriptase.
AID508631Antiviral activity against Human immunodeficiency virus 1 subtype G isolate JV1083 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID518742Selectivity ratio of EC50 for antiviral activity against HIV 1 subtype B harboring reverse transcriptase M230L mutant to EC50 for antiviral activity against HIV 1 subtype B harboring wild type reverse transcriptase2010Antimicrobial agents and chemotherapy, Jun, Volume: 54, Issue:6
The M230L nonnucleoside reverse transcriptase inhibitor resistance mutation in HIV-1 reverse transcriptase impairs enzymatic function and viral replicative capacity.
AID1316330Antiviral activity against wild type HIV1 3B harboring reverse transcriptase infected in human MT4 cells assessed as protection against virus induced cytotoxicity after 5 days by MTT assay2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID1862570Antiviral activity against HIV-1 F227L-V106A mutant infected in human MT4 cells assessed as reduction in viral replication by MTT assay
AID1561706Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV1 expressing RT K103N mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect
AID1410405Inhibition of recombinant wild type HIV1 reverse transcriptase assessed as decrease in biotin-dUTP incorporation using DIG-labeled dUTP/biotin-labeled dUTP and dTTP as template and viral nucleotides after 1 hr by ELISA2018ACS medicinal chemistry letters, Apr-12, Volume: 9, Issue:4
Discovery of Novel Diarylpyrimidine Derivatives as Potent HIV-1 NNRTIs Targeting the "NNRTI Adjacent" Binding Site.
AID586897Induction of CYP3A4 mRNA expression in human LS180 cells at 1 umol/liter after 4 days by RT-PCR analysis relative to beta-glucuronidase gene expression2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID508654Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase V179F, Y181C, F227C mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID496624Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase E138A/A33E, V35T, T39A, D123E, S162A, Q174G, T200E, I202V, Q207E mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID508794Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase L100I mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID693586Cytotoxicity against human MT4 cells assessed as decrease in cell viability after 5 days by MTT assay2012European journal of medicinal chemistry, Dec, Volume: 58Synthesis, biological evaluation and molecular modeling of 4,6-diarylpyrimidines and diarylbenzenes as novel non-nucleosides HIV-1 reverse transcriptase inhibitors.
AID1743618Antiviral activity against HIV1 3B infected in human MT4 cells assessed as virus-induced cytopathic effect incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Nov-15, Volume: 206Targeting dual tolerant regions of binding pocket: Discovery of novel morpholine-substituted diarylpyrimidines as potent HIV-1 NNRTIs with significantly improved water solubility.
AID1157581Antiviral activity against wild type HIV1 3B infected in human MT4 cells assessed as inhibition of virus-induced cytopathogenicity after 4 days by MTT assay2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Synthesis of novel fluoro analogues of MKC442 as microbicides.
AID1819214Antiviral activity against HIV1 L1001 infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay
AID508832Antiviral activity against Human immunodeficiency virus 1 subtype C isolate 93IN101 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1819209Antiviral activity against HIV-1 IIIB infected in human MT4 cells assessed as protection against virus-induced cytotoxicity incubated for 5 days by MTT assay
AID1561728Inhibition of HIV1 RT E138K mutant in presence of reconstituted template and viral nucleotides [digoxigenin (DIG)-dUTP, biotin-dUTP and dTTP] incubated for 1 hr by ELISA method
AID1822274Cytotoxicity against human MT4 cells assessed as reduction in cell viability measured after 5 days by MTT assay2022Journal of medicinal chemistry, 02-10, Volume: 65, Issue:3
Discovery of Novel Pyridine-Dimethyl-Phenyl-DAPY Hybrids by Molecular Fusing of Methyl-Pyrimidine-DAPYs and Difluoro-Pyridinyl-DAPYs: Improving the Druggability toward High Inhibitory Activity, Solubility, Safety, and PK.
AID1316334Selectivity index, ratio of CC50 for human mock-infected MT4 cells to EC50 for wild type HIV1 3B harboring reverse transcriptase infected in human MT4 cells2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID347629Antiviral activity against NNRTI-resistant HIV HXB2 isolate 20 with reverse transcriptase K103, Y181 mutation in human MT4 cells assessed as protection against virus-induced cell death after 5 days by MTT assay2008Journal of medicinal chemistry, Dec-11, Volume: 51, Issue:23
Design of annulated pyrazoles as inhibitors of HIV-1 reverse transcriptase.
AID1761021Inhibition of HIV-1 reverse transcriptase assessed as reduction in biotin-dUTP incorporation into cDNA incubated for 1 hr with DIG-dUTP, biotin-dUTP and dTTP followed by transferring onto streptavidin-coated plate and measured after 1 hr by ELISA2021European journal of medicinal chemistry, Feb-05, Volume: 211Design, synthesis, and evaluation of "dual-site"-binding diarylpyrimidines targeting both NNIBP and the NNRTI adjacent site of the HIV-1 reverse transcriptase.
AID1572071Resistance factor, ratio of EC50 for HIV1 harboring reverse transcriptase Y181C mutant to EC50 for wild type HIV12019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1457055Antiviral activity against HIV1 harboring reverse transcriptase K103N mutant infected in human ML4 cells assessed as protection against virus-induced cytopathic effect measured on day 5 post infection by MTT assay2017Journal of medicinal chemistry, 08-10, Volume: 60, Issue:15
Chiral Indolylarylsulfone Non-Nucleoside Reverse Transcriptase Inhibitors as New Potent and Broad Spectrum Anti-HIV-1 Agents.
AID1435507Antiviral activity against HIV1 expressing reverse transcriptase E138K mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay
AID496634Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase V90I,A98G/V35T, E36D, T39K, S48A, I50V, V60I, S68G, I135V, S162A, K173T, Q174K, D177E, G196E, T200A, E203G, Q207D, R211K, Q222P mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID632800Antiviral activity against Human immunodeficiency virus 1 NL4.3 reverse transcriptase Y181C mutant infected in human MT4 cells assessed as inhibition of viral infection2011Journal of medicinal chemistry, Dec-22, Volume: 54, Issue:24
Computationally-guided optimization of a docking hit to yield catechol diethers as potent anti-HIV agents.
AID1418468Inhibition of recombinant wild-type HIV1 GST-fused reverse transcriptase p66/p51 RNA-dependent DNA polymerase activity expressed in Escherichia coli assessed as reduction in biotin-dUTP incorporation using poly(rA)/oligo(dT)16 as template/primer after 40 2018Bioorganic & medicinal chemistry letters, 12-01, Volume: 28, Issue:22
Design and synthesis of a novel series of non-nucleoside HIV-1 inhibitors bearing pyrimidine and N-substituted aromatic piperazine.
AID1304409Antiviral activity against HIV-1 3B expressing reverse transcriptase K103N mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect by MTT assay2016Bioorganic & medicinal chemistry, 07-01, Volume: 24, Issue:13
Systematic evaluation of methyl ester bioisosteres in the context of developing alkenyldiarylmethanes (ADAMs) as non-nucleoside reverse transcriptase inhibitors (NNRTIs) for anti-HIV-1 chemotherapy.
AID1888709Solubility in water at pH 2
AID1443670Antiviral activity against HIV1 harboring reverse transcriptase F227L/V106A double mutant infected in human MT4 cells assessed as inhibition of viral replication after 5 days by MTT assay2017Bioorganic & medicinal chemistry letters, 04-15, Volume: 27, Issue:8
Design and synthesis of hybrids of diarylpyrimidines and diketo acids as HIV-1 inhibitors.
AID1124992Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 3B2014Bioorganic & medicinal chemistry, Apr-01, Volume: 22, Issue:7
Fused heterocyclic compounds bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 1: design, synthesis and biological evaluation of novel 5,7-disubstituted pyrazolo[1,5-a]pyrimidine derivatives.
AID586899Induction of ABCG2 protein expression in human LS180 cells at 1 umol/liter after 7 days by RT-PCR analysis relative to control2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1456310Cytotoxicity against human MT4 cells assessed as reduction in cell viability after 5 days by MTT assay2017Bioorganic & medicinal chemistry, 04-15, Volume: 25, Issue:8
Structural modifications of diarylpyrimidines (DAPYs) as HIV-1 NNRTIs: Synthesis, anti-HIV activities and SAR.
AID1171593Inhibition of wild type HIV1 reverse transcriptase assessed as reduction in enzyme activity2014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1561717Antiviral activity against HIV1 3B infected in human MT4 cells grown under 30 passages in presence of (E)-4-((4-((4-(4-(2-Cyanovinyl)-2,6-dimethylphenoxy)thieno[2,3- d]pyrimidin-2-yl)amino)piperidin-1-yl)methyl)benzenesulfonamide assessed as protection ag
AID1822273Antiviral activity against wild type HIV-1 IIIB infected in human MT4 cells assessed as protection against virus-induced cytotoxicity incubated for 5 days by MTT assay2022Journal of medicinal chemistry, 02-10, Volume: 65, Issue:3
Discovery of Novel Pyridine-Dimethyl-Phenyl-DAPY Hybrids by Molecular Fusing of Methyl-Pyrimidine-DAPYs and Difluoro-Pyridinyl-DAPYs: Improving the Druggability toward High Inhibitory Activity, Solubility, Safety, and PK.
AID1483286Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 RES056 reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1561705Resistance index, ratio of EC50 for antiviral activity against HIV1 expressing RT L100I mutant infected in human MT4 cells to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells
AID1574381Resistance index, ratio of EC50 for HIV-1 harboring reverse transcriptase Y188L mutant to EC50 for wild type HIV-1 NL4-32019Journal of medicinal chemistry, 01-24, Volume: 62, Issue:2
Effect of α-Methoxy Substitution on the Anti-HIV Activity of Dihydropyrimidin-4(3 H)-ones.
AID1773459Resistance factor, ratio of EC50 for HIV1 harboring Y18IC mutant infected in human MT4 cells to EC50 for HIV1 3B infected in MT4 cells
AID1235379Cytotoxicity against human MT4 cells assessed as reduction in cell viability incubated for 4 days MTT method2015Bioorganic & medicinal chemistry, Aug-01, Volume: 23, Issue:15
Hybrid chemistry. Part 4: Discovery of etravirine-VRX-480773 hybrids as potent HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1637396Antiviral activity against human HIV-1 3B harboring reverse transcriptase Y188L mutant infected in human MT4 cells assessed as protection from virus-induced cytopathic effect measured 5 days post infection by MTT assay2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Arylazolyl(azinyl)thioacetanilides. Part 20: Discovery of novel purinylthioacetanilides derivatives as potent HIV-1 NNRTIs via a structure-based bioisosterism approach.
AID1126510Selectivity index, ratio of CC50 for mock-infected human MT4 cells to IC50 for HIV1 3B infected in human MT4 cells2014Bioorganic & medicinal chemistry, Apr-15, Volume: 22, Issue:8
Structural modifications of CH(OH)-DAPYs as new HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1191399Antiviral activity against wild type HIV-1 3B infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 5 days by MTT assay2015Bioorganic & medicinal chemistry, Feb-01, Volume: 23, Issue:3
Synthesis and biological evaluation of DAPY-DPEs hybrids as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID496616Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase V90I/V35T, T39M, K46Q, V60I, S68G, D123E, I135V, S162A, K173A, Q174K,D177E, T200A, Q207E, R211K mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID1126509Cytotoxicity against mock-infected human MT4 cells after 5 days by MTT assay2014Bioorganic & medicinal chemistry, Apr-15, Volume: 22, Issue:8
Structural modifications of CH(OH)-DAPYs as new HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1357792Antiviral activity against HIV1 harboring reverse transcriptase Y188L mutant infected in human MT4 cells assessed as inhibition of viral replication after 5 days by MTT assay2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID508768Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase M230I mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1191403Selectivity index, ratio of CC50 for human MT4 cells to EC50 for wild type HIV-1 3B infected in human MT4 cells2015Bioorganic & medicinal chemistry, Feb-01, Volume: 23, Issue:3
Synthesis and biological evaluation of DAPY-DPEs hybrids as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1705183Antiviral activity against HIV-1 NL4-3 infected in human MT-4 cells assessed as virus induced cytopathic effect by MTT assay2020European journal of medicinal chemistry, Dec-15, Volume: 208New indolylarylsulfone non-nucleoside reverse transcriptase inhibitors show low nanomolar inhibition of single and double HIV-1 mutant strains.
AID1750725Selectivity index, ratio of CC50 for cytotoxicity against human MT-4 cells to EC50 for inhibition of HIV-1 p66/51 reverse transcriptase Y181C mutant2021Journal of medicinal chemistry, 04-22, Volume: 64, Issue:8
Hydrophobic Pocket Occupation Design of Difluoro-Biphenyl-Diarylpyrimidines as Non-Nucleoside HIV-1 Reverse Transcriptase Inhibitors: from
AID508630Antiviral activity against Human immunodeficiency virus 1 subtype G isolate G3 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1316328Selectivity index, ratio of CC50 for human mock-infected MT4 cells to EC50 for HIV1 3B harboring reverse transcriptase K103N mutant infected in human MT4 cells2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID1888697Antiviral activity against HIV1 with RT Y188L mutant infected in human MT4 cells cells assessed as protection against viral-induced cytopathogenecity by MTT assay
AID1754640Antiviral activity against wild type HIV-2 ROD infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect measured after 5 days by MTT assay2021Bioorganic & medicinal chemistry, 07-15, Volume: 42Exploiting the hydrophobic channel of the NNIBP: Discovery of novel diarylpyrimidines as HIV-1 NNRTIs against wild-type and K103N mutant viruses.
AID508648Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase L100I, K103N, T386A mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID508643Antiviral activity against Human immunodeficiency virus 1 subtype A1 infected in human MT4 cells assessed as reduction in virus induced cytopathicity2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1572069Resistance factor, ratio of EC50 for HIV1 harboring reverse transcriptase L100I mutant to EC50 for wild type HIV12019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1483291Fold resistance, ratio of EC50 for reverse transcriptase Y181C mutant in human MT4 cells infected HIV1 3B to EC50 for wild type HIV1 3B harboring reverse transcriptase infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID533677AUC (0 to 12 hrs) in healthy human plasma at 200 mg, po twice a day for 4 days coadministered with 400 mg of raltegravir, po twice a day for 4 days by reverse-phase HPLC-MS method2008Antimicrobial agents and chemotherapy, Dec, Volume: 52, Issue:12
Minimal pharmacokinetic interaction between the human immunodeficiency virus nonnucleoside reverse transcriptase inhibitor etravirine and the integrase inhibitor raltegravir in healthy subjects.
AID1248225Antiviral activity against HIV 1 RES056 harboring reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as protection against virus-induced cytopathogenicity after 5 days by MTT assay2015Bioorganic & medicinal chemistry, Oct-15, Volume: 23, Issue:20
A novel family of diarylpyrimidines (DAPYs) featuring a diatomic linker: Design, synthesis and anti-HIV activities.
AID1391083Antiviral activity against HIV1 harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay2018Bioorganic & medicinal chemistry, 05-01, Volume: 26, Issue:8
The discovery of novel diarylpyri(mi)dine derivatives with high level activity against a wide variety of HIV-1 strains as well as against HIV-2.
AID1819232Aqueous solubility of the compound at pH 7.4 by HPLC analysis
AID1572526Antiviral activity against HIV1 harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as inhibition of virus-induced cytotoxicity after 5 days by MTT assay
AID1520066Inhibition of recombinant wild type HIV-1 His-tagged reverse transcriptase p66/p51 expressed in Escherichia coli JM109 using poly(rA)/oligo(dT)16 as template/primer incubated for 40 mins by pico-green based spectrofluorometric analysis2020European journal of medicinal chemistry, Jan-01, Volume: 185Fragment hopping-based discovery of novel sulfinylacetamide-diarylpyrimidines (DAPYs) as HIV-1 nonnucleoside reverse transcriptase inhibitors.
AID1763907Inhibition of wild type HIV-1 reverse transcriptase assessed as reduction in biotin-dUTP incorporation in presence of template-primer incubated for 60 mins followed by transferring to streptavidin-coated plate and further incubation for 60 mins by ELISA2021Bioorganic & medicinal chemistry, 06-15, Volume: 40Design, synthesis and anti-HIV evaluation of novel 5-substituted diarylpyrimidine derivatives as potent HIV-1 NNRTIs.
AID1483302Inhibition of recombinant wild type HIV1 3B reverse transcriptase infected in human MT4 cells assessed as decrease in biotin-dUTP incorporation after 1 hr2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID417450Inhibition of HIV1 wild type reverse transcriptase2009European journal of medicinal chemistry, Feb, Volume: 44, Issue:2
QSAR studies for diarylpyrimidines against HIV-1 reverse transcriptase wild-type and mutant strains.
AID1410487Inhibition of HIV1 reverse transcriptase K103N mutant infected in human MT4 cells assessed as protection against virus-induced cytotoxicity by MTT assay2018ACS medicinal chemistry letters, Apr-12, Volume: 9, Issue:4
Further Exploring Solvent-Exposed Tolerant Regions of Allosteric Binding Pocket for Novel HIV-1 NNRTIs Discovery.
AID586900Activity at ABCB12011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID586903Activity at MRP32011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID464771Volume of distribution at steady state in Beagle dog at 0.5 mg/kg, iv or 0.5 mg/kg, po2010Bioorganic & medicinal chemistry letters, Mar-01, Volume: 20, Issue:5
N1-Heterocyclic pyrimidinediones as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1483277Inhibition of HIV1 3B reverse transcriptase F227L/V106A double mutant infected in human MT4 cells assessed as protection against virus induced cytotoxicity after 5 days by MTT assay2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1520060Selectivity index, ratio of CC50 for cytotoxicity against human MT4 cells to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells2020European journal of medicinal chemistry, Jan-01, Volume: 185Fragment hopping-based discovery of novel sulfinylacetamide-diarylpyrimidines (DAPYs) as HIV-1 nonnucleoside reverse transcriptase inhibitors.
AID1637394Antiviral activity against human HIV-1 3B harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as protection from virus-induced cytopathic effect measured 5 days post infection by MTT assay2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Arylazolyl(azinyl)thioacetanilides. Part 20: Discovery of novel purinylthioacetanilides derivatives as potent HIV-1 NNRTIs via a structure-based bioisosterism approach.
AID586885Induction of ABCC1 mRNA expression in human LS180 cells at 0.1 to 1 umol/liter after 4 days by RT-PCR analysis2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1320867Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for HIV1 RES056 expressing reverse transcriptase K103N/Y181C double mutant2016European journal of medicinal chemistry, Oct-04, Volume: 121Structural optimization of pyridine-type DAPY derivatives to exploit the tolerant regions of the NNRTI binding pocket.
AID522378Resistance index, ratio of EC50 for recombinant HIV1 harboring reverse transcriptase V106I/V179D mutant clone to EC50 for wild type HIV12010Antimicrobial agents and chemotherapy, Apr, Volume: 54, Issue:4
Combination of V106I and V179D polymorphic mutations in human immunodeficiency virus type 1 reverse transcriptase confers resistance to efavirenz and nevirapine but not etravirine.
AID619639Antiviral activity against HIV-1 3B harboring RT F227L and V106A mutant infected in human MT4 cells assessed as inhibition of virus-induced syncytium formation after 5 days by MTT assay2011European journal of medicinal chemistry, Oct, Volume: 46, Issue:10
Arylazolylthioacetanilide. Part 8: Design, synthesis and biological evaluation of novel 2-(2-(2,4-dichlorophenyl)-2H-1,2,4-triazol-3-ylthio)-N-arylacetamides as potent HIV-1 inhibitors.
AID246299Effective concentration of the compound to inhibit HIV-1 mutant L100I replication in HIV-infected MT-4 cells2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
From 4,5,6,7-tetrahydro-5-methylimidazo[4,5,1-jk](1,4)benzodiazepin-2(1H)-one (TIBO) to etravirine (TMC125): fifteen years of research on non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1235376Antiviral activity against HIV1 3B infected in human MT4 cells assessed as inhibition of virus-induced cytopathicity incubated for 4 days by MTT method2015Bioorganic & medicinal chemistry, Aug-01, Volume: 23, Issue:15
Hybrid chemistry. Part 4: Discovery of etravirine-VRX-480773 hybrids as potent HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1316345Selectivity index, ratio of CC50 for human mock-infected MT4 cells to EC50 for HIV1 3B harboring reverse transcriptase E138K mutant infected in human MT4 cells2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID648420Antiviral activity against HIV1 3B infected in human MT4 cells assessed as protection against virus-induced cytotoxicity after 5 days by MTT assay2012European journal of medicinal chemistry, May, Volume: 51Synthesis and biological evaluation of piperidine-substituted triazine derivatives as HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID496622Antiviral activity against HIV-1 subtype CRF06_cpx ontaining reverse transcriptase V106I/V21I, V35T, V60I, K122T, D123R, I135V, S162A, K173T, Q174K, D177E, I178L, T200A, Q207D, R211K, V245Q, E248D mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID1811049Aqueous solubility of compound in PBS at pH 7.4 by HPLC-UV analysis2021Journal of medicinal chemistry, 07-22, Volume: 64, Issue:14
Improving Druggability of Novel Diarylpyrimidine NNRTIs by a Fragment-Based Replacement Strategy: From Biphenyl-DAPYs to Heteroaromatic-Biphenyl-DAPYs.
AID1316339Antiviral activity against HIV1 3B harboring reverse transcriptase Y188L mutant infected in human MT4 cells assessed as protection against virus induced cytotoxicity after 5 days by MTT assay2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID518741Antiviral activity against HIV 1 subtype B harboring reverse transcriptase M230L mutant infected in human TZM-bl cells assessed as inhibition of viral growth by luciferase reporter gene assay2010Antimicrobial agents and chemotherapy, Jun, Volume: 54, Issue:6
The M230L nonnucleoside reverse transcriptase inhibitor resistance mutation in HIV-1 reverse transcriptase impairs enzymatic function and viral replicative capacity.
AID1572051Antiviral activity against wild type HIV1 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect after 5 days by MTT assay2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1443660Antiviral activity against wild type HIV1 3B infected in human MT4 cells assessed as inhibition of viral replication after 5 days by MTT assay2017Bioorganic & medicinal chemistry letters, 04-15, Volume: 27, Issue:8
Design and synthesis of hybrids of diarylpyrimidines and diketo acids as HIV-1 inhibitors.
AID1773441Antiviral activity against wild type HIV2 ROD infected in human MT4 cells assessed as reduction in virus-induced cytopathogenic effect by MTT assay
AID1862569Antiviral activity against HIV-1 E138K mutant infected in human MT4 cells assessed as reduction in viral replication by MTT assay
AID709860Antiviral activity against NNRTI-resistant HIV1 harboring reverse transcriptase Y181C/K103N double mutant infected in human MT4 cells assessed as inhibition of viral replication after 72 hrs by HeLa-CD4-LTR-beta-gal assay2012Journal of medicinal chemistry, Dec-13, Volume: 55, Issue:23
Rational design of potent non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID260237Antiviral activity against HIV1 3B in MT2 cells by MTT assay2006Bioorganic & medicinal chemistry letters, Feb, Volume: 16, Issue:3
Optimization of diarylamines as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID508758Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase K103N, Y181I mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1743626Antiviral activity against HIV1 harboring RT L100I mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Nov-15, Volume: 206Targeting dual tolerant regions of binding pocket: Discovery of novel morpholine-substituted diarylpyrimidines as potent HIV-1 NNRTIs with significantly improved water solubility.
AID586908Induction of MRP1 activity2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID508825Antiviral activity against Human immunodeficiency virus 1 subtype A isolate 92UG029 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID517497Antiviral activity against HIV1 harboring reverse transcriptase V106A mutant infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect by replication assay in presence of 10% FBS2010Bioorganic & medicinal chemistry letters, Oct-15, Volume: 20, Issue:20
Exploration of piperidine-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-Phenyl derivatives with broad potency against resistant mutant viruses.
AID586893Induction of ABCC3 mRNA expression in human LS180 cells at 0.1 to 1 umol/liter after 4 days by RT-PCR analysis2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1811048Aqueous solubility of compound in PBS at pH 7 by HPLC-UV analysis2021Journal of medicinal chemistry, 07-22, Volume: 64, Issue:14
Improving Druggability of Novel Diarylpyrimidine NNRTIs by a Fragment-Based Replacement Strategy: From Biphenyl-DAPYs to Heteroaromatic-Biphenyl-DAPYs.
AID508754Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase V179F, Y181C mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1141976Inhibition of HIV1 wild-type reverse transcriptase K103N mutant using [3H]dTTP by scintillation counting2014European journal of medicinal chemistry, Jun-10, Volume: 80New indolylarylsulfones as highly potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1741397Inhibition of reverse transcriptase E138K mutant in HIV-1 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Sep-15, Volume: 202Bioisosterism-based design and enantiomeric profiling of chiral hydroxyl-substituted biphenyl-diarylpyrimidine nonnucleoside HIV-1 reverse transcriptase inhibitors.
AID533676AUC (0 to 12 hrs) in healthy human plasma at 200 mg, po twice a day for 8 days by reverse-phase HPLC-MS method2008Antimicrobial agents and chemotherapy, Dec, Volume: 52, Issue:12
Minimal pharmacokinetic interaction between the human immunodeficiency virus nonnucleoside reverse transcriptase inhibitor etravirine and the integrase inhibitor raltegravir in healthy subjects.
AID1775802Antiviral activity against HIV-1 harboring RT F227L/V106A mutant infected in human MT4 cells assessed as protection against virus-induced cytopathogenicity after 5 days by MTT assay2021Journal of medicinal chemistry, 04-08, Volume: 64, Issue:7
2,4,5-Trisubstituted Pyrimidines as Potent HIV-1 NNRTIs: Rational Design, Synthesis, Activity Evaluation, and Crystallographic Studies.
AID738331Cytotoxicity against mock-infected human MT4 cells assessed as cell viability after 5 days by MTT assay2013Bioorganic & medicinal chemistry, Apr-01, Volume: 21, Issue:7
Synthesis and biological evaluation of pyridazine derivatives as novel HIV-1 NNRTIs.
AID1750719Inhibition of HIV-1 p66/p51 reverse transcriptase Y181C mutant incubated for 40 mins by picogreen dye-based spectrofluorometric analysis2021Journal of medicinal chemistry, 04-22, Volume: 64, Issue:8
Hydrophobic Pocket Occupation Design of Difluoro-Biphenyl-Diarylpyrimidines as Non-Nucleoside HIV-1 Reverse Transcriptase Inhibitors: from
AID1446830Solubility of the compound in 0.1 N HCl at pH 22017European journal of medicinal chemistry, Apr-21, Volume: 130Discovery of uracil-bearing DAPYs derivatives as novel HIV-1 NNRTIs via crystallographic overlay-based molecular hybridization.
AID1435504Antiviral activity against HIV1 expressing reverse transcriptase K103N mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay
AID1884487Cytotoxicity against human MT4 cells assessed as cell growth inhibition incubated for 5 days by MTT assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Expansion of the S-CN-DABO scaffold to exploit the impact on inhibitory activities against the non-nucleoside HIV-1 reverse transcriptase.
AID1249718Antiviral activity against HIV1 expressing reverse transcriptase L100I mutant2015European journal of medicinal chemistry, Sep-18, Volume: 102Pyrimidine sulfonylacetanilides with improved potency against key mutant viruses of HIV-1 by specific targeting of a highly conserved residue.
AID302621Cytotoxicity against human MT2 cells by MTT assay2007Journal of medicinal chemistry, Nov-01, Volume: 50, Issue:22
From docking false-positive to active anti-HIV agent.
AID583854Antiviral activity against HIV1 NL4-3 infected in human MT2 cells assessed as inhibition of virus-induced cell death at 0.5 multiplicities of infection after 6 days2010Antimicrobial agents and chemotherapy, Oct, Volume: 54, Issue:10
Lersivirine, a nonnucleoside reverse transcriptase inhibitor with activity against drug-resistant human immunodeficiency virus type 1.
AID1163233Antiviral activity against HIV1 infected in human TZM-bl cells assessed as viral inhibition pre-incubated for 30 mins prior to infection measured after 48 hrs by2014Bioorganic & medicinal chemistry, Oct-01, Volume: 22, Issue:19
From human immunodeficiency virus non-nucleoside reverse transcriptase inhibitors to potent and selective antitrypanosomal compounds.
AID1326646Inhibition of HIV-1 3B reverse transcriptase K103N mutant infected in human MT4 cells assessed as inhibition of viral replication after 5 days by cell titer glo based luciferase reporter gene assay2016European journal of medicinal chemistry, Oct-21, Volume: 122Novel (2,6-difluorophenyl)(2-(phenylamino)pyrimidin-4-yl)methanones with restricted conformation as potent non-nucleoside reverse transcriptase inhibitors against HIV-1.
AID464766Antiviral activity against NNRTI-resistant HIV1 harboring reverse transcriptase K103N mutant infected in human MT2 cells2010Bioorganic & medicinal chemistry letters, Mar-01, Volume: 20, Issue:5
N1-Heterocyclic pyrimidinediones as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1561715Resistance index, ratio of EC50 for antiviral activity against HIV1 expressing RT F227L + V106A mutant infected in human MT4 cells to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells
AID1888695Antiviral activity against HIV1 with RT K103N mutant infected in human MT4 cells cells assessed as protection against viral-induced cytopathogenecity by MTT assay
AID246300Effective concentration of the compound to inhibit HIV-1 mutant Y181C replication in HIV-infected MT-4 cells2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
From 4,5,6,7-tetrahydro-5-methylimidazo[4,5,1-jk](1,4)benzodiazepin-2(1H)-one (TIBO) to etravirine (TMC125): fifteen years of research on non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1197830Cytotoxicity against mock-infected human MT4 cells assessed as reduction in cell viability after 4 days by MTT assay2015European journal of medicinal chemistry, Mar-06, Volume: 92Fused heterocycles bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 3: optimization of [1,2,4]triazolo[1,5-a]pyrimidine core via structure-based and physicochemical property-driven approaches.
AID1565094Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV RES056
AID1379961Antiviral activity against HIV1 RES056 expressing reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathogenicity by measuring virus-induced syncytium formation by MTT assay2017European journal of medicinal chemistry, Nov-10, Volume: 140Design, synthesis and anti-HIV evaluation of novel diarylpyridine derivatives as potent HIV-1 NNRTIs.
AID1572067Selectivity ratio of CC50 for human MT4 cells to EC50 for HIV1 harboring reverse transcriptase E138K mutant infected in human MT4 cells2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1483290Fold resistance, ratio of EC50 for reverse transcriptase K103N/Y181C double mutant in human MT4 cells infected HIV1 RES056 to EC50 for wild type HIV1 3B harboring reverse transcriptase infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1637389Antiviral activity against human HIV-1 3B harboring RES056 mutant infected in human MT4 cells assessed as protection from virus-induced cytopathic effect measured 5 days post infection by MTT assay2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Arylazolyl(azinyl)thioacetanilides. Part 20: Discovery of novel purinylthioacetanilides derivatives as potent HIV-1 NNRTIs via a structure-based bioisosterism approach.
AID533681Tmax in healthy human plasma at 200 mg, po twice a day for 4 days coadministered with 400 mg of raltegravir, po twice a day for 4 days by reverse-phase HPLC-MS method2008Antimicrobial agents and chemotherapy, Dec, Volume: 52, Issue:12
Minimal pharmacokinetic interaction between the human immunodeficiency virus nonnucleoside reverse transcriptase inhibitor etravirine and the integrase inhibitor raltegravir in healthy subjects.
AID1880377Antiviral activity against HIV-1 IIIB harboring reverse transcriptase K103N mutant infected in human MT4 cells by MTT assay2022Journal of medicinal chemistry, 06-23, Volume: 65, Issue:12
Structure-Based Discovery of Novel NH
AID586905Inhibition of ABCB1 activity2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1609110Inhibition of recombinant HIV-1 reverse transcriptase p66/p51 incubated for 40 mins by picogreen dye based spectrofluorometric assay2019European journal of medicinal chemistry, Nov-15, Volume: 182Conformational restriction design of thiophene-biphenyl-DAPY HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1572077Aqueous solubility of the compound at pH at 2 by HPLC analysis2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID508751Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase Y181C, F227C mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1705190Resistance index, ratio of EC50 for antiviral activity against HIV1 harboring RT K103N mutant infected in human MT4 cells to EC50 for antiviral activity against HIV1 NL4-3 infected in human MT4 cells2020European journal of medicinal chemistry, Dec-15, Volume: 208New indolylarylsulfone non-nucleoside reverse transcriptase inhibitors show low nanomolar inhibition of single and double HIV-1 mutant strains.
AID1815385Antiviral activity against HIV1 harboring reverse transcriptase F227L/V106A mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1275550Antiviral activity against wild type HIV1 3B infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 5 days by MTT assay2016European journal of medicinal chemistry, Feb-15, Volume: 109Design, synthesis and anti-HIV evaluation of novel diarylpyridine derivatives targeting the entrance channel of NNRTI binding pocket.
AID1446812Antiviral activity against HIV-2 ROD infected in human MT4 cells assessed as protection against virus-induced cytopathic effect measured at 5 days post infection by MTT assay2017European journal of medicinal chemistry, Apr-21, Volume: 130Discovery of uracil-bearing DAPYs derivatives as novel HIV-1 NNRTIs via crystallographic overlay-based molecular hybridization.
AID1815404Water solubility of compound at pH 7 at 0.1 to 10 mg by HPLC-UV analysis2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1862566Antiviral activity against HIV-1 K103N mutant infected in human MT4 cells assessed as reduction in viral replication by MTT assay
AID1298248Inhibition of HIV1 reverse transcriptase p66/p51 L100I mutant using poly(rA) template and measured after 40 mins by picogreen based spectrofluorometry2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
1,6-Bis[(benzyloxy)methyl]uracil derivatives-Novel antivirals with activity against HIV-1 and influenza H1N1 virus.
AID767499Antiviral activity against wild type HIV1 3B infected in human MT2 cells assessed as protection against virus-induced effect by MTT assay2013Bioorganic & medicinal chemistry letters, Sep-15, Volume: 23, Issue:18
Optimization of diarylazines as anti-HIV agents with dramatically enhanced solubility.
AID1207401Inhibition of slow delayed inward rectifying potassium current (Iks) in Chinese Hamster Ovary (CHO) cells transfected with KCNQ1 / Kv1.7 / KvLQT1 and KCNE1/minK measured using IonWorks automated patch clamp platform
AID1773457Resistance factor, ratio of EC50 for HIV1 harboring L1001 mutant infected in human MT4 cells to EC50 for HIV1 3B infected in MT4 cells
AID586881Inhibition of MRP3 expressed in MDCK2 cells assessed as cell growth inhibition by calcein and pheophorbide A efflux assays2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1636149Ratio of IC50 for NNRTI resistant HIV1 harboring reverse transcriptase K103N/V179F/Y181C mutant infected in human TZM-bl cells to IC50 for wild type HIV1 NL4-3 infected in human TZM-bl cells2016Journal of medicinal chemistry, Aug-11, Volume: 59, Issue:15
Discovery and Structure-Based Optimization of 2-Ureidothiophene-3-carboxylic Acids as Dual Bacterial RNA Polymerase and Viral Reverse Transcriptase Inhibitors.
AID1069122Antiviral activity against HIV harboring reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as inhibition of viral infection in presence of 50% normal human serum2014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Discovery of MK-1439, an orally bioavailable non-nucleoside reverse transcriptase inhibitor potent against a wide range of resistant mutant HIV viruses.
AID1884224Antiviral activity against HIV-1 harboring L100I mutant infected in human MT4 cells assessed as reduction in virus induced cytotoxicity incubated for 5 to 7 days by MTT assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Chemical space exploration around indolylarylsulfone scaffold led to a novel class of highly active HIV-1 NNRTIs with spiro structural features.
AID586910Induction of BCRP activity2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1609106Antiviral activity against HIV-1 harboring reverse transcriptase L100I mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 4 days by MTT assay2019European journal of medicinal chemistry, Nov-15, Volume: 182Conformational restriction design of thiophene-biphenyl-DAPY HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1884493Antiviral activity against drug-resistant HIV-1 E138K mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Expansion of the S-CN-DABO scaffold to exploit the impact on inhibitory activities against the non-nucleoside HIV-1 reverse transcriptase.
AID1815399Inhibition of HIV1 reverse transcriptase Y181C mutant using oligo(dT)16 as primer measured after 40 mins by picogreen-dye based spectrofluorimetric analysis2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1163232Cytotoxicity against human MRC5 SV2 cells assessed as reduction in cell growth after 72 hrs2014Bioorganic & medicinal chemistry, Oct-01, Volume: 22, Issue:19
From human immunodeficiency virus non-nucleoside reverse transcriptase inhibitors to potent and selective antitrypanosomal compounds.
AID1572056Selectivity ratio of CC50 for human MT4 cells to EC50 for HIV1 RES056 infected in human MT4 cells2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1750724Selectivity index, ratio of CC50 for cytotoxicity against human MT-4 cells to EC50 for inhibition of HIV-1 p66/51 reverse transcriptase K103N mutant2021Journal of medicinal chemistry, 04-22, Volume: 64, Issue:8
Hydrophobic Pocket Occupation Design of Difluoro-Biphenyl-Diarylpyrimidines as Non-Nucleoside HIV-1 Reverse Transcriptase Inhibitors: from
AID1561722Resistance index, ratio of EC50 for antiviral activity against HIV1 3B infected in human MT4 cells grown under 30 passages in presence of 4-[[4-[[4-[4-[2-cyanovinyl]-2,6-dimethylphenoxy]thieno[3,2-d]pyrimidin-2-yl]amino]-1-piperidyl]methyl]benzenesulfonam
AID614139Selectivity index, ratio of CC50 for human MT4 cells to EC50 for wild type HIV1 IIIB infected in human MT4 cells2011Bioorganic & medicinal chemistry, Sep-01, Volume: 19, Issue:17
Synthesis and structure-activity relationship of novel diarylpyrimidines with hydromethyl linker (CH(OH)-DAPYs) as HIV-1 NNRTIs.
AID709868Antiviral activity against NNRTI-resistant HIV1 harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as inhibition of viral replication after 72 hrs by HeLa-CD4-LTR-beta-gal assay2012Journal of medicinal chemistry, Dec-13, Volume: 55, Issue:23
Rational design of potent non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1504697Selectivity index, ratio of CC50 for human MT4 cells to EC50 for wild type HIV1 3B infected in human MT4 cells2017ACS medicinal chemistry letters, Nov-09, Volume: 8, Issue:11
Discovery of Thiophene[3,2-
AID236429Area under the concentration time curve in human after 100 mg/kg oral dosage2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Concentration and pH dependent aggregation of hydrophobic drug molecules and relevance to oral bioavailability.
AID1421310Inhibition of HIV1 reverse transcriptase L100I/K103N double mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect by MTT assay2018European journal of medicinal chemistry, Oct-05, Volume: 158Advances in diarylpyrimidines and related analogues as HIV-1 nonnucleoside reverse transcriptase inhibitors.
AID1636929Antiviral activity against HIV1 3B harboring reverse transcriptase Y181C mutant infected in human MT2 cells assessed as protection against viral infection by MTT method2016Bioorganic & medicinal chemistry, 10-15, Volume: 24, Issue:20
Computer-aided discovery of anti-HIV agents.
AID1815395Resistance fold, ratio of EC50 for HIV1 harboring reverse transcriptase K103N/Y181C double mutant infected MV4 cells to EC50 for wild type HIV1 infected MV4 cells2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1561730Inhibition of HIV1 RT K103N/Y181C mutant in presence of reconstituted template and viral nucleotides [digoxigenin (DIG)-dUTP, biotin-dUTP and dTTP] incubated for 1 hr by ELISA method
AID1410488Inhibition of HIV1 reverse transcriptase E138K mutant infected in human MT4 cells assessed as protection against virus-induced cytotoxicity by MTT assay2018ACS medicinal chemistry letters, Apr-12, Volume: 9, Issue:4
Further Exploring Solvent-Exposed Tolerant Regions of Allosteric Binding Pocket for Novel HIV-1 NNRTIs Discovery.
AID1572047Aqueous solubility of the compound at pH at 7.4 by HPLC analysis2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID508753Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase V179F, Y181I mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1421301Inhibition of HIV1 reverse transcriptase Y181C mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect by MTT assay2018European journal of medicinal chemistry, Oct-05, Volume: 158Advances in diarylpyrimidines and related analogues as HIV-1 nonnucleoside reverse transcriptase inhibitors.
AID1357796Resistance factor, ratio of EC50 for HIV1 harboring reverse transcriptase E138K mutant to EC50 for wild-type HIV-1 3B2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID1888704Resistance index, ratio of EC50 for antiviral activity against HIV1 with RT F227L + V106A mutant infected in human MT4 cells to EC50 for antiviral activity against wild type HIV1 infected in human MT4 cells
AID1248224Selectivity index, ratio of CC50 for uninfected human MT4 cells to EC50 for wild type HIV 1 3B infected in human MT4 cells2015Bioorganic & medicinal chemistry, Oct-15, Volume: 23, Issue:20
A novel family of diarylpyrimidines (DAPYs) featuring a diatomic linker: Design, synthesis and anti-HIV activities.
AID106053Inhibitory activity against 100I strain of HIV-I in MT-4 cells2001Bioorganic & medicinal chemistry letters, Sep-03, Volume: 11, Issue:17
Evolution of anti-HIV drug candidates. Part 3: Diarylpyrimidine (DAPY) analogues.
AID1565113Sub-acute toxicity in Kunming mouse assessed as increase in alveolar interstitial thickening administered for 14 days measured on day 14 by HE-staining based assay
AID1705187Antiviral activity against HIV1 harboring RT Y181C mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect by MTT assay2020European journal of medicinal chemistry, Dec-15, Volume: 208New indolylarylsulfone non-nucleoside reverse transcriptase inhibitors show low nanomolar inhibition of single and double HIV-1 mutant strains.
AID1754645Antiviral activity against HIV1 harboring RT E138K mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect incubated for 5 days by MTT assay2021Bioorganic & medicinal chemistry, 07-15, Volume: 42Exploiting the hydrophobic channel of the NNIBP: Discovery of novel diarylpyrimidines as HIV-1 NNRTIs against wild-type and K103N mutant viruses.
AID1750722Inhibition of recombinant wild type p66/p51 HIV1 reverse transcriptase incubated for 40 mins by picogreen dye-based spectrofluorometric analysis2021Journal of medicinal chemistry, 04-22, Volume: 64, Issue:8
Hydrophobic Pocket Occupation Design of Difluoro-Biphenyl-Diarylpyrimidines as Non-Nucleoside HIV-1 Reverse Transcriptase Inhibitors: from
AID1391086Antiviral activity against HIV1 harboring reverse transcriptase F227L/V106A double mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay2018Bioorganic & medicinal chemistry, 05-01, Volume: 26, Issue:8
The discovery of novel diarylpyri(mi)dine derivatives with high level activity against a wide variety of HIV-1 strains as well as against HIV-2.
AID1292009Antiviral activity against wild type HIV1 3B infected in human MT4 cells assessed as cell viability after 4 days by MTT assay2016European journal of medicinal chemistry, Jun-10, Volume: 115Design, synthesis and evaluation of novel HIV-1 NNRTIs with dual structural conformations targeting the entrance channel of the NNRTI binding pocket.
AID1197837Solubility in water at pH 7.0 after 2 hrs by HPLC-UV method2015European journal of medicinal chemistry, Mar-06, Volume: 92Fused heterocycles bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 3: optimization of [1,2,4]triazolo[1,5-a]pyrimidine core via structure-based and physicochemical property-driven approaches.
AID1888701Resistance index, ratio of EC50 for antiviral activity against HIV1 with RT Y181C mutant infected in human MT4 cells to EC50 for antiviral activity against wild type HIV1 infected in human MT4 cells
AID1466764Inhibition of wild-type HIV-1 3B reverse transcriptase infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect after 5 days by MTT assay2017Bioorganic & medicinal chemistry letters, 06-15, Volume: 27, Issue:12
Drug-like property-driven optimization of 4-substituted 1,5-diarylanilines as potent HIV-1 non-nucleoside reverse transcriptase inhibitors against rilpivirine-resistant mutant virus.
AID619641Ratio of EC50 for HIV-1 3B harboring RT K103N mutant infected in human MT4 cells to EC50 for wild type HIV-1 3B infected in human MT4 cells2011European journal of medicinal chemistry, Oct, Volume: 46, Issue:10
Arylazolylthioacetanilide. Part 8: Design, synthesis and biological evaluation of novel 2-(2-(2,4-dichlorophenyl)-2H-1,2,4-triazol-3-ylthio)-N-arylacetamides as potent HIV-1 inhibitors.
AID586901Activity at MRP12011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1561737Solubility of the compound in pH 7.4 buffer by HPLC analysis
AID508651Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase L100I, V179I, Y181C mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID701097Antiviral activity against HIV1 harboring reverse transcriptase L100I/K103N mutant2012Journal of medicinal chemistry, Apr-26, Volume: 55, Issue:8
Strategies for the design of HIV-1 non-nucleoside reverse transcriptase inhibitors: lessons from the development of seven representative paradigms.
AID1862571Antiviral activity against HIV-1 K103N-Y181C mutant infected in human MT4 cells assessed as reduction in viral replication by MTT assay
AID1565115Sub-acute toxicity in Kunming mouse assessed as increase in proximal convoluted tubule edema administered for 14 days measured on day 14 by HE-staining based assay
AID518738Inhibition of DNA-dependent DNA polymerase activity of HIV1 subtype B reverse transcriptase M230L mutant by gel-based primer extension assay2010Antimicrobial agents and chemotherapy, Jun, Volume: 54, Issue:6
The M230L nonnucleoside reverse transcriptase inhibitor resistance mutation in HIV-1 reverse transcriptase impairs enzymatic function and viral replicative capacity.
AID1775799Antiviral activity against HIV-1 harboring RT Y181C mutant infected in human MT4 cells assessed as protection against virus-induced cytopathogenicity after 5 days by MTT assay2021Journal of medicinal chemistry, 04-08, Volume: 64, Issue:7
2,4,5-Trisubstituted Pyrimidines as Potent HIV-1 NNRTIs: Rational Design, Synthesis, Activity Evaluation, and Crystallographic Studies.
AID1884488Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against NNRTI resistant HIV-1 infected in human MT4 cells2022European journal of medicinal chemistry, Aug-05, Volume: 238Expansion of the S-CN-DABO scaffold to exploit the impact on inhibitory activities against the non-nucleoside HIV-1 reverse transcriptase.
AID1880369Inhibition of HIV-1 reverse transcriptase using oligo(dT)16 as primer measured after 40 mins by picogreen-dye based spectrofluorometric analysis2022Journal of medicinal chemistry, 06-23, Volume: 65, Issue:12
Structure-Based Discovery of Novel NH
AID1483280Selectivity index, ratio of CC50 for human MT4 cells to EC50 for reverse transcriptase L100I mutant in human MT4 cells infected HIV1 3B2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1483276Inhibition of HIV1 3B reverse transcriptase E138K mutant infected in human MT4 cells assessed as protection against virus induced cytotoxicity after 5 days by MTT assay2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1815377Anti-HIV activity against HIV-1 IIIB infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1157594Antiviral activity against TMC120-resistant HIV1 harboring RT 100I, 138G mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathogenicity after 4 days by MTT assay2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Synthesis of novel fluoro analogues of MKC442 as microbicides.
AID1815382Antiviral activity against HIV1 harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1815398Inhibition of HIV1 reverse transcriptase K103N mutant using oligo(dT)16 as primer measured after 40 mins by picogreen-dye based spectrofluorimetric analysis2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1456306Inhibition of HIV-1 3B wild type reverse transcriptase infected in human MT4 cells assessed as protection against virus-induced cytopathic effect measured at 5 days post infection by MTT assay2017Bioorganic & medicinal chemistry, 04-15, Volume: 25, Issue:8
Structural modifications of diarylpyrimidines (DAPYs) as HIV-1 NNRTIs: Synthesis, anti-HIV activities and SAR.
AID1357804Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 harboring reverse transcriptase Y188L mutant2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID1126506Antiviral activity against HIV1 3B infected in human MT4 cells assessed as inhibition of viral cytopathogenicity after 5 days by MTT assay2014Bioorganic & medicinal chemistry, Apr-15, Volume: 22, Issue:8
Structural modifications of CH(OH)-DAPYs as new HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1316337Antiviral activity against HIV1 3B harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as protection against virus induced cytotoxicity after 5 days by MTT assay2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID1561700Antiviral activity against HIV1 expressing RT Y181C mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1561716Antiviral activity against HIV1 3B infected in human MT4 cells grown under 30 passages in absence of test compound assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1457053Antiviral activity against HIV1 NL4-3 infected in human ML4 cells assessed as protection against virus-induced cytopathic effect measured on day 5 post infection by MTT assay2017Journal of medicinal chemistry, 08-10, Volume: 60, Issue:15
Chiral Indolylarylsulfone Non-Nucleoside Reverse Transcriptase Inhibitors as New Potent and Broad Spectrum Anti-HIV-1 Agents.
AID1435516Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 expressing reverse transcriptase E138K mutant
AID1126508Antiviral activity against HIV2 ROD infected in human MT4 cells assessed as inhibition of viral cytopathogenicity after 5 days by MTT assay2014Bioorganic & medicinal chemistry, Apr-15, Volume: 22, Issue:8
Structural modifications of CH(OH)-DAPYs as new HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1884225Antiviral activity against HIV-1 harboring K103N mutant infected in human MT4 cells assessed as reduction in virus induced cytotoxicity incubated for 5 to 7 days by MTT assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Chemical space exploration around indolylarylsulfone scaffold led to a novel class of highly active HIV-1 NNRTIs with spiro structural features.
AID1171413Antiviral activity against wild type HIV1 harboring reverse transcriptase K103N/Y181C mutant infected in human MT2 cells assessed as protection from virus-induced cytopathicity by MTT assay2014ACS medicinal chemistry letters, Nov-13, Volume: 5, Issue:11
Picomolar Inhibitors of HIV-1 Reverse Transcriptase: Design and Crystallography of Naphthyl Phenyl Ethers.
AID698145Inhibition of HIV1 reverse transcriptase K103N mutant RNA-dependent DNA polymerase activity using poly(rA)/oligo(dT)10:1 and [3H]-dTTP substrate2012Journal of medicinal chemistry, Jul-26, Volume: 55, Issue:14
New nitrogen containing substituents at the indole-2-carboxamide yield high potent and broad spectrum indolylarylsulfone HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID541250Antiviral activity against multidrug resistant HIV1 RTMDR containing reverse transcriptase L74V, M41L, V106A and T215Y mutant infected in human TZM-bl cells after 4 days by luciferase reporter gene assay2010Journal of medicinal chemistry, Dec-09, Volume: 53, Issue:23
Design, synthesis, and evaluation of diarylpyridines and diarylanilines as potent non-nucleoside HIV-1 reverse transcriptase inhibitors.
AID246298Effective concentration of the compound to inhibit HIV-1 mutant K103N replication in HIV-infected MT-4 cells2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
From 4,5,6,7-tetrahydro-5-methylimidazo[4,5,1-jk](1,4)benzodiazepin-2(1H)-one (TIBO) to etravirine (TMC125): fifteen years of research on non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID701096Antiviral activity against HIV1 harboring reverse transcriptase K103N/Y181C mutant2012Journal of medicinal chemistry, Apr-26, Volume: 55, Issue:8
Strategies for the design of HIV-1 non-nucleoside reverse transcriptase inhibitors: lessons from the development of seven representative paradigms.
AID1637398Antiviral activity against human HIV-1 3B harboring reverse transcriptase F227L/V106A double mutant infected in human MT4 cells assessed as protection from virus-induced cytopathic effect measured 5 days post infection by MTT assay2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Arylazolyl(azinyl)thioacetanilides. Part 20: Discovery of novel purinylthioacetanilides derivatives as potent HIV-1 NNRTIs via a structure-based bioisosterism approach.
AID83550Potency evaluated against NNRTI-Resistant HIV-1 strain Lys103Asn2004Journal of medicinal chemistry, May-06, Volume: 47, Issue:10
Roles of conformational and positional adaptability in structure-based design of TMC125-R165335 (etravirine) and related non-nucleoside reverse transcriptase inhibitors that are highly potent and effective against wild-type and drug-resistant HIV-1 varian
AID1316333Cytotoxicity against human mock-infected MT4 cells assessed as reduction in cell viability after 5 days by MTT assay2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID1687682Antiviral activity against HIV-1 Y181C mutant strain infected in human MT4 cells assessed as reduction in virus-induced cytopathogenicity by MTT assay2020European journal of medicinal chemistry, Jan-15, Volume: 186Indazolyl-substituted piperidin-4-yl-aminopyrimidines as HIV-1 NNRTIs: Design, synthesis and biological activities.
AID1819213Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV-1 IIIB harboring K103N/Y181C double mutant infected in human MT4 cells
AID1446818Antiviral activity against HIV1 harboring Y188L mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect measured at 5 days post infection by MTT assay2017European journal of medicinal chemistry, Apr-21, Volume: 130Discovery of uracil-bearing DAPYs derivatives as novel HIV-1 NNRTIs via crystallographic overlay-based molecular hybridization.
AID1520057Antiviral activity against HIV1 3B infected in human MT4 cells assessed as virus-induced cytopathic effect incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Fragment hopping-based discovery of novel sulfinylacetamide-diarylpyrimidines (DAPYs) as HIV-1 nonnucleoside reverse transcriptase inhibitors.
AID1775793Antiviral activity against HIV-1 ROD infected in human MT4 cells assessed as protection against virus-induced cytopathogenicity after 5 days by MTT assay2021Journal of medicinal chemistry, 04-08, Volume: 64, Issue:7
2,4,5-Trisubstituted Pyrimidines as Potent HIV-1 NNRTIs: Rational Design, Synthesis, Activity Evaluation, and Crystallographic Studies.
AID1609104Antiviral activity against HIV-1 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 4 days by MTT assay2019European journal of medicinal chemistry, Nov-15, Volume: 182Conformational restriction design of thiophene-biphenyl-DAPY HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1561693Antiviral activity against HIV1 RES056 infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1811071Oral bioavailability in Sprague-Dawley rat at 5 mg/kg by LCMS analysis2021Journal of medicinal chemistry, 07-22, Volume: 64, Issue:14
Improving Druggability of Novel Diarylpyrimidine NNRTIs by a Fragment-Based Replacement Strategy: From Biphenyl-DAPYs to Heteroaromatic-Biphenyl-DAPYs.
AID1410408Cytotoxicity against human MT4 cells assessed as decrease in cell viability after 5 days by MTT assay2018ACS medicinal chemistry letters, Apr-12, Volume: 9, Issue:4
Discovery of Novel Diarylpyrimidine Derivatives as Potent HIV-1 NNRTIs Targeting the "NNRTI Adjacent" Binding Site.
AID517492Antiviral activity against wild type HIV1 infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect by replication assay in presence of 10% FBS2010Bioorganic & medicinal chemistry letters, Oct-15, Volume: 20, Issue:20
Exploration of piperidine-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-Phenyl derivatives with broad potency against resistant mutant viruses.
AID1410482Inhibition of HIV1 RES056 reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as protection against virus-induced cytotoxicity by MTT assay2018ACS medicinal chemistry letters, Apr-12, Volume: 9, Issue:4
Further Exploring Solvent-Exposed Tolerant Regions of Allosteric Binding Pocket for Novel HIV-1 NNRTIs Discovery.
AID1811044Antiviral activity against HIV1 IIIB infected human MT4 cells assessed as protection against virus-induced cytopathic effect by MTT assay2021Journal of medicinal chemistry, 07-22, Volume: 64, Issue:14
Improving Druggability of Novel Diarylpyrimidine NNRTIs by a Fragment-Based Replacement Strategy: From Biphenyl-DAPYs to Heteroaromatic-Biphenyl-DAPYs.
AID1275548Antiviral activity against HIV1 3B expressing reverse transcriptase L100I mutant infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 5 days by MTT assay2016European journal of medicinal chemistry, Feb-15, Volume: 109Design, synthesis and anti-HIV evaluation of novel diarylpyridine derivatives targeting the entrance channel of NNRTI binding pocket.
AID508757Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase E138K, M230L mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1435509Antiviral activity against HIV1 RES056 expressing reverse transcriptase mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay
AID1357798Resistance factor, ratio of EC50 for HIV1 harboring reverse transcriptase Y188L mutant to EC50 for wild-type HIV-1 3B2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID476470Antiviral activity against HIV1 reverse transcriptase Y181C mutant infected in human MT2 cells assessed as inhibition of viral replication by MTT assay2010Bioorganic & medicinal chemistry letters, Apr-15, Volume: 20, Issue:8
Eastern extension of azoles as non-nucleoside inhibitors of HIV-1 reverse transcriptase; cyano group alternatives.
AID1435503Antiviral activity against HIV1 expressing reverse transcriptase L100I mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay
AID1761017Antiviral activity against HIV-1 harboring reverse transcriptase Y188L mutant infected in human MT4 cells assessed as inhibition of virus induced cytopathic effect measured after 5 days by MTT assay2021European journal of medicinal chemistry, Feb-05, Volume: 211Design, synthesis, and evaluation of "dual-site"-binding diarylpyrimidines targeting both NNIBP and the NNRTI adjacent site of the HIV-1 reverse transcriptase.
AID1504702Antiviral activity against HIV1 3B harboring reverse transcriptase E138K mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2017ACS medicinal chemistry letters, Nov-09, Volume: 8, Issue:11
Discovery of Thiophene[3,2-
AID1815405Water solubility of compound at pH 7.4 at 0.1 to 10 mg by HPLC-UV analysis2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1729160Antiviral activity against HIV1 harboring E138K mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect by MTT assay2021European journal of medicinal chemistry, Mar-05, Volume: 213Exploiting the tolerant region I of the non-nucleoside reverse transcriptase inhibitor (NNRTI) binding pocket. Part 2: Discovery of diarylpyrimidine derivatives as potent HIV-1 NNRTIs with high Fsp
AID1884226Antiviral activity against HIV-1 harboring Y181C mutant infected in human MT4 cells assessed as reduction in virus induced cytotoxicity incubated for 5 to 7 days by MTT assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Chemical space exploration around indolylarylsulfone scaffold led to a novel class of highly active HIV-1 NNRTIs with spiro structural features.
AID1822280Antiviral activity against HIV-1 harboring E138K mutant infected in human MT4 cells assessed as protection against virus-induced cytotoxicity incubated for 5 days by MTT assay2022Journal of medicinal chemistry, 02-10, Volume: 65, Issue:3
Discovery of Novel Pyridine-Dimethyl-Phenyl-DAPY Hybrids by Molecular Fusing of Methyl-Pyrimidine-DAPYs and Difluoro-Pyridinyl-DAPYs: Improving the Druggability toward High Inhibitory Activity, Solubility, Safety, and PK.
AID1636928Cytotoxicity against human MT2 cells assessed as growth inhibition by MTT method2016Bioorganic & medicinal chemistry, 10-15, Volume: 24, Issue:20
Computer-aided discovery of anti-HIV agents.
AID557066Antiviral activity against HIV1 isolate R8 harboring reverse transcriptase K101P mutant relative to drug-sensitive HIV1 CNDO2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID1687684Antiviral activity against HIV-1 RES056 mutant strain infected in human MT4 cells assessed as reduction in virus-induced cytopathogenicity by MTT assay2020European journal of medicinal chemistry, Jan-15, Volume: 186Indazolyl-substituted piperidin-4-yl-aminopyrimidines as HIV-1 NNRTIs: Design, synthesis and biological activities.
AID665272Antiviral activity against HIV1 RES056 harboring K103N/Y181C RT mutant infected in MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2012European journal of medicinal chemistry, Jul, Volume: 53Chiral resolution, absolute configuration assignment and biological activity of racemic diarylpyrimidine CH(OH)-DAPY as potent nonnucleoside HIV-1 reverse transcriptase inhibitors.
AID508635Antiviral activity against Human immunodeficiency virus 1 subtype (H) infected in human MT4 cells assessed as reduction in virus induced cytopathicity2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1741393Inhibition of reverse transcriptase L100I mutant in HIV-1 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Sep-15, Volume: 202Bioisosterism-based design and enantiomeric profiling of chiral hydroxyl-substituted biphenyl-diarylpyrimidine nonnucleoside HIV-1 reverse transcriptase inhibitors.
AID508759Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase K103N, Y181C mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1888705Resistance index, ratio of EC50 for antiviral activity against HIV1 with RT RES056 mutant infected in human MT4 cells to EC50 for antiviral activity against wild type HIV1 infected in human MT4 cells
AID1197833Resistance index, ratio of EC50 for HIV1 RES056 expressing reverse transcriptase K103N + Y181C double mutant to EC50 for wild type HIV1 3B2015European journal of medicinal chemistry, Mar-06, Volume: 92Fused heterocycles bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 3: optimization of [1,2,4]triazolo[1,5-a]pyrimidine core via structure-based and physicochemical property-driven approaches.
AID508833Antiviral activity against Human immunodeficiency virus 1 subtype C isolate 93MW959 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1743637Resistance index, ratio of EC50 for antiviral activity against HIV1 expressing RT Y188L mutant infected in human MT4 cells to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells2020European journal of medicinal chemistry, Nov-15, Volume: 206Targeting dual tolerant regions of binding pocket: Discovery of novel morpholine-substituted diarylpyrimidines as potent HIV-1 NNRTIs with significantly improved water solubility.
AID557036Antiviral activity against HIV1 isolate R8 harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as inhibition of viral replication after 72 hrs in presence of 50% human serum2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID1572059Antiviral activity against HIV1 harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect after 5 days by MTT assay2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1884539Fold resistance, ratio of EC50 for antiviral activity against drug-resistant HIV-1 K103 N mutant infected in human MT4 cells to EC50 for antiviral activity against NNRTI resistant wild type HIV-1 infected in human MT4 cells2022European journal of medicinal chemistry, Aug-05, Volume: 238Expansion of the S-CN-DABO scaffold to exploit the impact on inhibitory activities against the non-nucleoside HIV-1 reverse transcriptase.
AID1304403Cytotoxicity against mock-infected human MT4 cells2016Bioorganic & medicinal chemistry, 07-01, Volume: 24, Issue:13
Systematic evaluation of methyl ester bioisosteres in the context of developing alkenyldiarylmethanes (ADAMs) as non-nucleoside reverse transcriptase inhibitors (NNRTIs) for anti-HIV-1 chemotherapy.
AID1574386Inhibition of recombinant HIV-1 His-tagged reverse transcriptase p66/p51 Y181I mutant expressed in Escherichia coli assessed as reduction in [3H]dTTP incorporation using poly(rA)/oligo(dT) as template/primer after 20 mins by scintillation counting analysi2019Journal of medicinal chemistry, 01-24, Volume: 62, Issue:2
Effect of α-Methoxy Substitution on the Anti-HIV Activity of Dihydropyrimidin-4(3 H)-ones.
AID1773444Selectivity ratio of CC50 for human MT4 cells to IC50 for HIV1 RES056 infected in MT4 cells
AID1637393Antiviral activity against human HIV-1 3B harboring reverse transcriptase L100I mutant infected in human MT4 cells assessed as protection from virus-induced cytopathic effect measured 5 days post infection by MTT assay2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Arylazolyl(azinyl)thioacetanilides. Part 20: Discovery of novel purinylthioacetanilides derivatives as potent HIV-1 NNRTIs via a structure-based bioisosterism approach.
AID1561739Solubility of the compound in pH 2.0 buffer by HPLC analysis
AID1152372Antiviral activity against HIV1 RES056 harboring RT K103N,Y181C double mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathicity measured 5 days post viral infection by MTT assay2014Bioorganic & medicinal chemistry, Jun-15, Volume: 22, Issue:12
Synthesis and biological evaluation of CHX-DAPYs as HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID586879Inhibition of MRP1 expressed in MDCK2 cells assessed as cell growth inhibition by calcein and pheophorbide A efflux assays2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID83547Potency evaluated against NNRTI-Resistant HIV-1 strain Gly190Ala2004Journal of medicinal chemistry, May-06, Volume: 47, Issue:10
Roles of conformational and positional adaptability in structure-based design of TMC125-R165335 (etravirine) and related non-nucleoside reverse transcriptase inhibitors that are highly potent and effective against wild-type and drug-resistant HIV-1 varian
AID508639Antiviral activity against Human immunodeficiency virus 1 subtype C infected in human MT4 cells assessed as reduction in virus induced cytopathicity2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1171578Cytotoxic activity against human MT4 cells by MTT assay2014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1235378Antiviral activity against HIV2 ROD infected in human MT4 cells assessed as inhibition of virus-induced cytopathicity incubated for 4 days by MTT method2015Bioorganic & medicinal chemistry, Aug-01, Volume: 23, Issue:15
Hybrid chemistry. Part 4: Discovery of etravirine-VRX-480773 hybrids as potent HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1822276Antiviral activity against HIV-1 harboring RT L100I mutant infected in human MT4 cells assessed as protection against virus-induced cytotoxicity incubated for 5 days by MTT assay2022Journal of medicinal chemistry, 02-10, Volume: 65, Issue:3
Discovery of Novel Pyridine-Dimethyl-Phenyl-DAPY Hybrids by Molecular Fusing of Methyl-Pyrimidine-DAPYs and Difluoro-Pyridinyl-DAPYs: Improving the Druggability toward High Inhibitory Activity, Solubility, Safety, and PK.
AID1558849Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells incubated for 5 days by MTT assay2019Journal of medicinal chemistry, 12-26, Volume: 62, Issue:24
Ligand-Based Design of Nondimethylphenyl-Diarylpyrimidines with Improved Metabolic Stability, Safety, and Oral Pharmacokinetic Profiles.
AID1157587Antiviral activity against saquinavir-resistant HIV1 infected in human MT4 cells assessed as inhibition of virus-induced cytopathogenicity after 4 days by MTT assay2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Synthesis of novel fluoro analogues of MKC442 as microbicides.
AID496619Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase A98G/V35T, V60I, I135L, S162A, K173A, Q174K, D177E, I178M, T200A,Q207E, R211K, V245Q, D250E mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID632795Antiviral activity against Human immunodeficiency virus 1 NL4.3 infected in human MT2 cells assessed as inhibition of viral infection2011Journal of medicinal chemistry, Dec-22, Volume: 54, Issue:24
Computationally-guided optimization of a docking hit to yield catechol diethers as potent anti-HIV agents.
AID508781Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase E138R mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID522373Antiviral activity against recombinant HIV1 harboring reverse transcriptase V106I/V179D mutant clone infected in MAGIC-5 cells using 5-bromo4-chloro-3-indolyl-beta-D-galactopyranoside staining based light microscopy2010Antimicrobial agents and chemotherapy, Apr, Volume: 54, Issue:4
Combination of V106I and V179D polymorphic mutations in human immunodeficiency virus type 1 reverse transcriptase confers resistance to efavirenz and nevirapine but not etravirine.
AID1880380Antiviral activity against HIV-1 IIIB harboring reverse transcriptase E138K mutant infected in human MT4 cells by MTT assay2022Journal of medicinal chemistry, 06-23, Volume: 65, Issue:12
Structure-Based Discovery of Novel NH
AID1888694Antiviral activity against HIV1 with RT L100I mutant infected in human MT4 cells cells assessed as protection against viral-induced cytopathogenecity by MTT assay
AID1773830Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for HIV1 RES056 infected in human MT4 cells2021Journal of medicinal chemistry, 09-23, Volume: 64, Issue:18
Structure-Based Design and Discovery of Pyridyl-Bearing Fused Bicyclic HIV-1 Inhibitors: Synthesis, Biological Characterization, and Molecular Modeling Studies.
AID1352322Antiviral activity against HIV1 harboring reverse transcriptase F227L/V106A double mutant infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 4 days by MTT assay2018European journal of medicinal chemistry, Feb-10, Volume: 145Discovery of biphenyl-substituted diarylpyrimidines as non-nucleoside reverse transcriptase inhibitors with high potency against wild-type and mutant HIV-1.
AID508634Antiviral activity against Human immunodeficiency virus 2 ROD infected in human MT4 cells assessed as reduction in virus induced cytopathicity2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1811037Anti-viral activity against HIV1 harboring RT K103N mutant infected MV4 cells assessed as protection against virus-induced cytopathic effect by MTT assay2021Journal of medicinal chemistry, 07-22, Volume: 64, Issue:14
Improving Druggability of Novel Diarylpyrimidine NNRTIs by a Fragment-Based Replacement Strategy: From Biphenyl-DAPYs to Heteroaromatic-Biphenyl-DAPYs.
AID1157584Antiviral activity against A17-sensitive HIV1 harboring 103N, 181C mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathogenicity after 4 days by MTT assay2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Synthesis of novel fluoro analogues of MKC442 as microbicides.
AID1391085Antiviral activity against HIV1 harboring reverse transcriptase E138K mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay2018Bioorganic & medicinal chemistry, 05-01, Volume: 26, Issue:8
The discovery of novel diarylpyri(mi)dine derivatives with high level activity against a wide variety of HIV-1 strains as well as against HIV-2.
AID1320865Cytotoxicity against mock-infected human MT4 cells assessed as reduction in cell viability after 5 days by MTT assay2016European journal of medicinal chemistry, Oct-04, Volume: 121Structural optimization of pyridine-type DAPY derivatives to exploit the tolerant regions of the NNRTI binding pocket.
AID1637403Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for human HIV-1 3B harboring E138K mutant infected in human MT4 cells2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Arylazolyl(azinyl)thioacetanilides. Part 20: Discovery of novel purinylthioacetanilides derivatives as potent HIV-1 NNRTIs via a structure-based bioisosterism approach.
AID484395Colloidal aggregation in fed state simulated intestinal fluid assessed as colloid radius at 200 uM by dynamic light scattering assay in presence of 1% DMSO2010Journal of medicinal chemistry, May-27, Volume: 53, Issue:10
Colloid formation by drugs in simulated intestinal fluid.
AID465419Antiviral activity against HIV1 harboring reverse transcriptase Y181C mutant infected in human MT2 cells2010Bioorganic & medicinal chemistry letters, Mar-01, Volume: 20, Issue:5
N1-Alkyl pyrimidinediones as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1316336Antiviral activity against HIV1 3B harboring reverse transcriptase L100I mutant infected in human MT4 cells assessed as protection against virus induced cytotoxicity after 5 days by MTT assay2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID1326647Inhibition of reverse transcriptase Y181C mutant in HIV-1 3B infected in human MT4 cells assessed as inhibition of viral replication after 5 days by cell titer glo based luciferase reporter gene assay2016European journal of medicinal chemistry, Oct-21, Volume: 122Novel (2,6-difluorophenyl)(2-(phenylamino)pyrimidin-4-yl)methanones with restricted conformation as potent non-nucleoside reverse transcriptase inhibitors against HIV-1.
AID1171583Antiviral activity against HIV1 NL4-3 expressing reverse transcriptase K103N mutant infected in human MT4 cells assessed as reduction in virus-induced cell death by MTT assay2014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1819233Aqueous solubility of the compound at pH 7.0 by HPLC analysis
AID1561698Antiviral activity against HIV1 expressing RT L100I mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay
AID484275Inhibition of Trypanosoma cruzi cruzaine preincubated for 5 mins before substrate addition by fluorescence assay in presence of 0.01% Triton X-1002010Journal of medicinal chemistry, May-27, Volume: 53, Issue:10
Colloid formation by drugs in simulated intestinal fluid.
AID1815381Antiviral activity against HIV1 harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID557019Inhibition of HIV1 isolate R8 reverse transcriptase after 90 mins2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID586887Induction of ABCC4 mRNA expression in human LS180 cells at 0.1 to 1 umol/liter after 4 days by RT-PCR analysis2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1609105Selectivity index, ratio of CC50 for human MT4 cells to EC50 of antiviral activity against HIV-1 IIIB infected in human MT4 cells2019European journal of medicinal chemistry, Nov-15, Volume: 182Conformational restriction design of thiophene-biphenyl-DAPY HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1729157Antiviral activity against HIV1 harboring K103N mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect by MTT assay2021European journal of medicinal chemistry, Mar-05, Volume: 213Exploiting the tolerant region I of the non-nucleoside reverse transcriptase inhibitor (NNRTI) binding pocket. Part 2: Discovery of diarylpyrimidine derivatives as potent HIV-1 NNRTIs with high Fsp
AID1637400Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for human HIV-1 3B harboring K103N mutant infected in human MT4 cells2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Arylazolyl(azinyl)thioacetanilides. Part 20: Discovery of novel purinylthioacetanilides derivatives as potent HIV-1 NNRTIs via a structure-based bioisosterism approach.
AID508767Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase M230L mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1572522Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 RES056 harboring reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells
AID508762Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase K101E, K103N mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1457061Ratio of EC50 for HIV-1 harboring reverse transcriptase Y188L mutant infected in human MT4 cells to EC50 for HIV1 NL4-3 infected in human MT4 cells2017Journal of medicinal chemistry, 08-10, Volume: 60, Issue:15
Chiral Indolylarylsulfone Non-Nucleoside Reverse Transcriptase Inhibitors as New Potent and Broad Spectrum Anti-HIV-1 Agents.
AID1888690Antiviral activity against HIV1 3B infected in human MT4 cells cells assessed as protection against viral-induced cytopathogenecity by MTT assay
AID738333Antiviral activity against HIV2 ROD infected in human MT4 cells assessed as protection against virus-induced cytopathicity after 5 days by MTT assay2013Bioorganic & medicinal chemistry, Apr-01, Volume: 21, Issue:7
Synthesis and biological evaluation of pyridazine derivatives as novel HIV-1 NNRTIs.
AID1750721Inhibition of HIV-1 p66/p51 reverse transcriptase E138K mutant incubated for 40 mins by picogreen dye-based spectrofluorometric analysis2021Journal of medicinal chemistry, 04-22, Volume: 64, Issue:8
Hydrophobic Pocket Occupation Design of Difluoro-Biphenyl-Diarylpyrimidines as Non-Nucleoside HIV-1 Reverse Transcriptase Inhibitors: from
AID557067Plasma protein binding in HIV1 infected patient2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID1884222Cytotoxicity against human MT4 cells assessed as reduction in cell viability incubated for 5 to 7 days by MTT assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Chemical space exploration around indolylarylsulfone scaffold led to a novel class of highly active HIV-1 NNRTIs with spiro structural features.
AID1561726Inhibition of HIV1 RT Y181C mutant in presence of reconstituted template and viral nucleotides [digoxigenin (DIG)-dUTP, biotin-dUTP and dTTP] incubated for 1 hr by ELISA method
AID1693799Antiviral activity against HIV-3B infected in human MT-4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2021Bioorganic & medicinal chemistry, 01-15, Volume: 30Novel indolylarylsulfone derivatives as covalent HIV-1 reverse transcriptase inhibitors specifically targeting the drug-resistant mutant Y181C.
AID1741399Inhibition of reverse transcriptase RES056 mutant in HIV-1 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Sep-15, Volume: 202Bioisosterism-based design and enantiomeric profiling of chiral hydroxyl-substituted biphenyl-diarylpyrimidine nonnucleoside HIV-1 reverse transcriptase inhibitors.
AID1705188Antiviral activity against HIV1 harboring RT Y188L mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect by MTT assay2020European journal of medicinal chemistry, Dec-15, Volume: 208New indolylarylsulfone non-nucleoside reverse transcriptase inhibitors show low nanomolar inhibition of single and double HIV-1 mutant strains.
AID1731755Solubility in water at pH 7 at 0.1 to 10 mg
AID586883fCmax in human plasma at 200 mg bid after 8 days2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID632801Antiviral activity against Human immunodeficiency virus 1 NL4.3 reverse transcriptase K103N and Y181C double mutant infected in human MT4 cells assessed as inhibition of viral infection2011Journal of medicinal chemistry, Dec-22, Volume: 54, Issue:24
Computationally-guided optimization of a docking hit to yield catechol diethers as potent anti-HIV agents.
AID1761009Antiviral activity against wild type HIV-1 3B infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect measured after 5 days by MTT assay2021European journal of medicinal chemistry, Feb-05, Volume: 211Design, synthesis, and evaluation of "dual-site"-binding diarylpyrimidines targeting both NNIBP and the NNRTI adjacent site of the HIV-1 reverse transcriptase.
AID465418Antiviral activity against wild type HIV1 infected in human MT2 cells2010Bioorganic & medicinal chemistry letters, Mar-01, Volume: 20, Issue:5
N1-Alkyl pyrimidinediones as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1457056Antiviral activity against HIV1 harboring reverse transcriptase Y181C mutant infected in human ML4 cells assessed as protection against virus-induced cytopathic effect measured on day 5 post infection by MTT assay2017Journal of medicinal chemistry, 08-10, Volume: 60, Issue:15
Chiral Indolylarylsulfone Non-Nucleoside Reverse Transcriptase Inhibitors as New Potent and Broad Spectrum Anti-HIV-1 Agents.
AID1691438Selectivity index, ratio of CC50 for cytotoxicity against human MT4 cells to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells2020European journal of medicinal chemistry, May-01, Volume: 193In situ click chemistry-based rapid discovery of novel HIV-1 NNRTIs by exploiting the hydrophobic channel and tolerant regions of NNIBP.
AID490368Antiviral activity against HIV1 expressing reverse transcriptase Y188L mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect2010Bioorganic & medicinal chemistry letters, Jul-15, Volume: 20, Issue:14
Discovery of piperidin-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-benzyl derivatives with broad potency against resistant mutant viruses.
AID586886Induction of ABCC2 mRNA expression in human LS180 cells at 0.1 to 1 umol/liter after 4 days by RT-PCR analysis2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1558855Antiviral activity against HIV1 harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2019Journal of medicinal chemistry, 12-26, Volume: 62, Issue:24
Ligand-Based Design of Nondimethylphenyl-Diarylpyrimidines with Improved Metabolic Stability, Safety, and Oral Pharmacokinetic Profiles.
AID1705192Resistance index, ratio of EC50 for antiviral activity against HIV1 harboring RT Y188L mutant infected in human MT4 cells to EC50 for antiviral activity against HIV1 NL4-3 infected in human MT4 cells2020European journal of medicinal chemistry, Dec-15, Volume: 208New indolylarylsulfone non-nucleoside reverse transcriptase inhibitors show low nanomolar inhibition of single and double HIV-1 mutant strains.
AID1609109Antiviral activity against HIV-1 harboring reverse transcriptase E138K mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 4 days by MTT assay2019European journal of medicinal chemistry, Nov-15, Volume: 182Conformational restriction design of thiophene-biphenyl-DAPY HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1197835Inhibition of HIV1 reverse transcriptase assessed as reduction in biotin-labeled dUTP incorporation into DNA after 1 hr using poly(A) x oligo(dT)15 template/primer hybrid, digoxigenin and biotin-labeled nucleotides2015European journal of medicinal chemistry, Mar-06, Volume: 92Fused heterocycles bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 3: optimization of [1,2,4]triazolo[1,5-a]pyrimidine core via structure-based and physicochemical property-driven approaches.
AID1457066Inhibition of HIV1 reverse transcriptase p66 K103N/Y1881C double mutant associated RNA dependent DNA polymerase activity expressed in Escherichia coli assessed as reduction in [3H]dTTP incorporation using poly(rA)/oligo(dT) as template/primer by scintilla2017Journal of medicinal chemistry, 08-10, Volume: 60, Issue:15
Chiral Indolylarylsulfone Non-Nucleoside Reverse Transcriptase Inhibitors as New Potent and Broad Spectrum Anti-HIV-1 Agents.
AID259755Cytotoxicity against MT2 cells2006Bioorganic & medicinal chemistry letters, Feb, Volume: 16, Issue:3
Computer-aided design of non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1483293Fold resistance, ratio of EC50 for reverse transcriptase F227L/V106A double mutant in human MT4 cells infected HIV1 3B to EC50 for wild type HIV1 3B harboring reverse transcriptase infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID476473Cytotoxicity against human MT2 cells infected with HIV1 harboring reverse transcriptase K103N/Y181C mutation by MTT assay2010Bioorganic & medicinal chemistry letters, Apr-15, Volume: 20, Issue:8
Eastern extension of azoles as non-nucleoside inhibitors of HIV-1 reverse transcriptase; cyano group alternatives.
AID1583024Antiviral activity against HIV1 containing reverse transcriptase L100I mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2020Journal of medicinal chemistry, 02-13, Volume: 63, Issue:3
Discovery and Characterization of Fluorine-Substituted Diarylpyrimidine Derivatives as Novel HIV-1 NNRTIs with Highly Improved Resistance Profiles and Low Activity for the hERG Ion Channel.
AID586898Induction of ABCB1 protein expression in human LS180 cells at 1 umol/liter after 7 days by RT-PCR analysis relative to control2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID508797Ratio of EC50 for HIV1 in presence of 45 mg/ml HSA to EC50 for HIV1 in absence of serum proteins by GFP assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1435510Cytotoxicity against human MT4 cells assessed as decrease in cell viability after 5 days by MTT assay
AID1504705Inhibition of recombinant wild type HIV1 reverse transcriptase using DIG-labeled dUTP/biotin-labeled dUTP and dTTP as template and viral nucleotides assessed as decrease in biotin-dUTP incorporation after 1 hr by ELISA2017ACS medicinal chemistry letters, Nov-09, Volume: 8, Issue:11
Discovery of Thiophene[3,2-
AID1731747Inhibition of reverse transcriptase K103N mutant in HIV-1 infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay
AID464772Oral bioavailability in Beagle dog at 4 mg/kg, po2010Bioorganic & medicinal chemistry letters, Mar-01, Volume: 20, Issue:5
N1-Heterocyclic pyrimidinediones as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1705196Inhibition of RNA-dependent DNA polymerase activity of recombinant HIV-1 p66/p51 reverse transcriptase Y188L mutant assessed as inhibition of [3H]dTTP incorporation using poly(rA)/oligo(dT) as templates incubated for 15 mins by MicroBeta scintillation cou2020European journal of medicinal chemistry, Dec-15, Volume: 208New indolylarylsulfone non-nucleoside reverse transcriptase inhibitors show low nanomolar inhibition of single and double HIV-1 mutant strains.
AID1072807Antiviral activity against HIV1 harboring reverse transcriptase K101N mutant infected in human MT4 cells2014Bioorganic & medicinal chemistry, Mar-15, Volume: 22, Issue:6
Discovery of 2-pyridone derivatives as potent HIV-1 NNRTIs using molecular hybridization based on crystallographic overlays.
AID1152371Antiviral activity against HIV2 ROD infected in human MT4 cells assessed as inhibition of virus-induced cytopathicity measured 5 days post viral infection by MTT assay2014Bioorganic & medicinal chemistry, Jun-15, Volume: 22, Issue:12
Synthesis and biological evaluation of CHX-DAPYs as HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1773826Antiviral activity against HIV1 3B infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2021Journal of medicinal chemistry, 09-23, Volume: 64, Issue:18
Structure-Based Design and Discovery of Pyridyl-Bearing Fused Bicyclic HIV-1 Inhibitors: Synthesis, Biological Characterization, and Molecular Modeling Studies.
AID508771Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase G190A mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1197834Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for HIV1 RES056 expressing reverse transcriptase K103N + Y181C double mutant2015European journal of medicinal chemistry, Mar-06, Volume: 92Fused heterocycles bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 3: optimization of [1,2,4]triazolo[1,5-a]pyrimidine core via structure-based and physicochemical property-driven approaches.
AID508778Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase V179E mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID586892Induction of ABCB1 mRNA expression in human LS180 cells at 0.1 to 1 umol/liter after 4 days by RT-PCR analysis2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1561736Aqueous solubility of the compound in pH 7 buffer
AID1572052Antiviral activity against HIV1 RES056 infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect after 5 days by MTT assay2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1446831Lipophilicity, log P of the compound2017European journal of medicinal chemistry, Apr-21, Volume: 130Discovery of uracil-bearing DAPYs derivatives as novel HIV-1 NNRTIs via crystallographic overlay-based molecular hybridization.
AID1815403Water solubility of compound at pH 2 at 0.1 to 10 mg by HPLC-UV analysis2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID476471Cytotoxicity against human MT2 cells infected with HIV1 harboring reverse transcriptase Y181C mutation by MTT assay2010Bioorganic & medicinal chemistry letters, Apr-15, Volume: 20, Issue:8
Eastern extension of azoles as non-nucleoside inhibitors of HIV-1 reverse transcriptase; cyano group alternatives.
AID1292011Cytotoxicity against mock infected human MT4 cells after 4 days by MTT assay2016European journal of medicinal chemistry, Jun-10, Volume: 115Design, synthesis and evaluation of novel HIV-1 NNRTIs with dual structural conformations targeting the entrance channel of the NNRTI binding pocket.
AID1379959Antiviral activity against HIV1 3B infected in human MT4 cells assessed as inhibition of virus-induced cytopathogenicity by measuring virus-induced syncytium formation by MTT assay2017European journal of medicinal chemistry, Nov-10, Volume: 140Design, synthesis and anti-HIV evaluation of novel diarylpyridine derivatives as potent HIV-1 NNRTIs.
AID522369Antiviral activity against recombinant HIV1 harboring reverse transcriptase V106A mutant clone infected in MAGIC-5 cells using 5-bromo4-chloro-3-indolyl-beta-D-galactopyranoside staining based light microscopy2010Antimicrobial agents and chemotherapy, Apr, Volume: 54, Issue:4
Combination of V106I and V179D polymorphic mutations in human immunodeficiency virus type 1 reverse transcriptase confers resistance to efavirenz and nevirapine but not etravirine.
AID1609107Antiviral activity against HIV-1 harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 4 days by MTT assay2019European journal of medicinal chemistry, Nov-15, Volume: 182Conformational restriction design of thiophene-biphenyl-DAPY HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1888708Solubility in water at pH 7
AID1171584Antiviral activity against HIV1 NL4-3 expressing reverse transcriptase Y181C mutant infected in human MT4 cells assessed as reduction in virus-induced cell death by MTT assay2014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1637402Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for human HIV-1 3B harboring Y188L mutant infected in human MT4 cells2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Arylazolyl(azinyl)thioacetanilides. Part 20: Discovery of novel purinylthioacetanilides derivatives as potent HIV-1 NNRTIs via a structure-based bioisosterism approach.
AID522577Resistance index, ratio of EC50 for recombinant HIV1 harboring reverse transcriptase V106I mutant clone to EC50 for wild type HIV12010Antimicrobial agents and chemotherapy, Apr, Volume: 54, Issue:4
Combination of V106I and V179D polymorphic mutations in human immunodeficiency virus type 1 reverse transcriptase confers resistance to efavirenz and nevirapine but not etravirine.
AID1391080Selectivity index, ratio of CC50 for human MT4 cells to EC50 for wild type HIV1 3B infected in human MT4 cells2018Bioorganic & medicinal chemistry, 05-01, Volume: 26, Issue:8
The discovery of novel diarylpyri(mi)dine derivatives with high level activity against a wide variety of HIV-1 strains as well as against HIV-2.
AID583853Antiviral activity against HIV1 NL4-3 infected in human MT2 cells assessed as inhibition of virus-induced cell death at 0.05 multiplicities of infection after 6 days2010Antimicrobial agents and chemotherapy, Oct, Volume: 54, Issue:10
Lersivirine, a nonnucleoside reverse transcriptase inhibitor with activity against drug-resistant human immunodeficiency virus type 1.
AID1124991Cytotoxicity against human MT4 cells assessed as cell viability by MTT assay2014Bioorganic & medicinal chemistry, Apr-01, Volume: 22, Issue:7
Fused heterocyclic compounds bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 1: design, synthesis and biological evaluation of novel 5,7-disubstituted pyrazolo[1,5-a]pyrimidine derivatives.
AID1888700Resistance index, ratio of EC50 for antiviral activity against HIV1 with RT K103N mutant infected in human MT4 cells to EC50 for antiviral activity against wild type HIV1 infected in human MT4 cells
AID1163254Antileishmanial activity against Leishmania infantum MHOM/MA (BE)/67 infected in primary peritoneal mouse macrophages assessed as reduction in parasite burdun2014Bioorganic & medicinal chemistry, Oct-01, Volume: 22, Issue:19
From human immunodeficiency virus non-nucleoside reverse transcriptase inhibitors to potent and selective antitrypanosomal compounds.
AID1572524Antiviral activity against HIV1 harboring reverse transcriptase L100I mutant infected in human MT4 cells assessed as inhibition of virus-induced cytotoxicity after 5 days by MTT assay
AID1815401Inhibition of HIV1 reverse transcriptase E138K mutant using oligo(dT)16 as primer measured after 40 mins by picogreen-dye based spectrofluorimetric analysis2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1207464Inhibition of rapid delayed inward rectifying potassium current (IKr) in Chinese hamster ovary (CHO) K1 cells stably expressing hERG measured using IonWorks Quattro automated patch clamp platform
AID635302Antiviral activity against HIV1 harboring reverse transcriptase K103N/Y181C mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathogenicity after 5 days by MTT assay2011Bioorganic & medicinal chemistry, Dec-01, Volume: 19, Issue:23
Design, synthesis and biological evaluation of cycloalkyl arylpyrimidines (CAPYs) as HIV-1 NNRTIs.
AID1754647Antiviral activity against HIV1 harboring RT RES056 mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect incubated for 5 days by MTT assay2021Bioorganic & medicinal chemistry, 07-15, Volume: 42Exploiting the hydrophobic channel of the NNIBP: Discovery of novel diarylpyrimidines as HIV-1 NNRTIs against wild-type and K103N mutant viruses.
AID1763898Antiviral activity against HIV-1 IIIB harboring RT L100I mutant infected in human MT4 cells assessed as protection against virus-induced cytotoxicity incubated for 5 days by MTT assay2021Bioorganic & medicinal chemistry, 06-15, Volume: 40Design, synthesis and anti-HIV evaluation of novel 5-substituted diarylpyrimidine derivatives as potent HIV-1 NNRTIs.
AID1249720Antiviral activity against HIV1 expressing reverse transcriptase E138K mutant2015European journal of medicinal chemistry, Sep-18, Volume: 102Pyrimidine sulfonylacetanilides with improved potency against key mutant viruses of HIV-1 by specific targeting of a highly conserved residue.
AID1773454Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 harboring Y188L mutant infected in human MT4 cells
AID1565114Sub-acute toxicity in Kunming mouse assessed as increase in alveolar hemorrhage administered for 14 days measured on day 14 by HE-staining based assay
AID1320868Solubility of the compound2016European journal of medicinal chemistry, Oct-04, Volume: 121Structural optimization of pyridine-type DAPY derivatives to exploit the tolerant regions of the NNRTI binding pocket.
AID1275552Selectivity index, ratio of CC50 for human MT4 cells to EC50 for wild type HIV1 3B infected in human MT4 cells2016European journal of medicinal chemistry, Feb-15, Volume: 109Design, synthesis and anti-HIV evaluation of novel diarylpyridine derivatives targeting the entrance channel of NNRTI binding pocket.
AID1298243Antiviral activity against HIV-1 3B infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 5 days by MTT assay2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
1,6-Bis[(benzyloxy)methyl]uracil derivatives-Novel antivirals with activity against HIV-1 and influenza H1N1 virus.
AID106074Inhibitory activity against LAI strain of HIV-I in MT-4 cells2001Bioorganic & medicinal chemistry letters, Sep-03, Volume: 11, Issue:17
Evolution of anti-HIV drug candidates. Part 3: Diarylpyrimidine (DAPY) analogues.
AID698956Antiviral activity against HIV-1 NL4-3 infected in human TZM-bl cells assessed as inhibition of viral replication after 2 days by luciferase reporter gene based luminescence assay2012Journal of medicinal chemistry, Aug-23, Volume: 55, Issue:16
Design, synthesis, and preclinical evaluations of novel 4-substituted 1,5-diarylanilines as potent HIV-1 non-nucleoside reverse transcriptase inhibitor (NNRTI) drug candidates.
AID1729158Antiviral activity against HIV1 harboring Y181C mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect by MTT assay2021European journal of medicinal chemistry, Mar-05, Volume: 213Exploiting the tolerant region I of the non-nucleoside reverse transcriptase inhibitor (NNRTI) binding pocket. Part 2: Discovery of diarylpyrimidine derivatives as potent HIV-1 NNRTIs with high Fsp
AID698146Inhibition of wild type HIV1 reverse transcriptase RNA-dependent DNA polymerase activity using poly(rA)/oligo(dT)10:1 and [3H]-dTTP substrate2012Journal of medicinal chemistry, Jul-26, Volume: 55, Issue:14
New nitrogen containing substituents at the indole-2-carboxamide yield high potent and broad spectrum indolylarylsulfone HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID508621Antiviral activity against Human immunodeficiency virus 1 subtype D isolate 92UG001 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1141978Inhibition of HIV1 wild-type reverse transcriptase L1001 mutant using [3H]dTTP by scintillation counting2014European journal of medicinal chemistry, Jun-10, Volume: 80New indolylarylsulfones as highly potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1316340Antiviral activity against HIV1 3B harboring reverse transcriptase E138K mutant infected in human MT4 cells assessed as protection against virus induced cytotoxicity after 5 days by MTT assay2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID1410481Inhibition of wild-type HIV1 3B reverse transcriptase infected in human MT4 cells assessed as protection against virus-induced cytotoxicity by MTT assay2018ACS medicinal chemistry letters, Apr-12, Volume: 9, Issue:4
Further Exploring Solvent-Exposed Tolerant Regions of Allosteric Binding Pocket for Novel HIV-1 NNRTIs Discovery.
AID484383Colloidal aggregation in fed state simulated intestinal fluid assessed as colloid radius at 10 uM by dynamic light scattering assay in presence of 1% DMSO2010Journal of medicinal chemistry, May-27, Volume: 53, Issue:10
Colloid formation by drugs in simulated intestinal fluid.
AID709836Ratio of EC50 for HIV1 expressing wild type reverse transcriptase in presence of human serum albumin and human alpha-1 acid glycoprotein to EC50 for HIV1 expressing wild type reverse transcriptase2012Journal of medicinal chemistry, Dec-13, Volume: 55, Issue:23
Rational design of potent non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1574374Antiviral activity against HIV-1 IRLL98 harboring reverse transcriptase M41L, D67N, Y181C, M184V, R211K, and T215Y mutants infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2019Journal of medicinal chemistry, 01-24, Volume: 62, Issue:2
Effect of α-Methoxy Substitution on the Anti-HIV Activity of Dihydropyrimidin-4(3 H)-ones.
AID508641Antiviral activity against Human immunodeficiency virus 1 subtype AG infected in human MT4 cells assessed as reduction in virus induced cytopathicity2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID557038Antiviral activity against HIV1 isolate R8 harboring wild type reverse transcriptase infected in human MT4 cells assessed as inhibition of viral replication after 72 hrs in presence of 50% human serum2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID1574372Antiviral activity against wild type HIV 1 NL4-3 infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2019Journal of medicinal chemistry, 01-24, Volume: 62, Issue:2
Effect of α-Methoxy Substitution on the Anti-HIV Activity of Dihydropyrimidin-4(3 H)-ones.
AID541251Resistance index, ratio of EC50 for multidrug resistant HIV1 RTMDR to EC50 for wild type HIV1 NL4-32010Journal of medicinal chemistry, Dec-09, Volume: 53, Issue:23
Design, synthesis, and evaluation of diarylpyridines and diarylanilines as potent non-nucleoside HIV-1 reverse transcriptase inhibitors.
AID449195Antimalarial activity against Plasmodium falciparum W2mef at 10 uM after 48 hrs by hoechst 33342-thiazole orange stain based flow cytometry assay2009Bioorganic & medicinal chemistry letters, Sep-15, Volume: 19, Issue:18
Addressing the malaria drug resistance challenge using flow cytometry to discover new antimalarials.
AID1815389Antiviral activity against HIV1 harboring reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1882474Antiviral activity against HIV-1 harboring E138K mutant infected in human MT4 cells assessed as protection against virus-induced cytopathicity by MTT assay2022Journal of medicinal chemistry, 03-10, Volume: 65, Issue:5
Contemporary Medicinal Chemistry Strategies for the Discovery and Development of Novel HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors.
AID1743639Resistance index, ratio of EC50 for antiviral activity against HIV1 expressing RT F227L/V106A mutant infected in human MT4 cells to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells2020European journal of medicinal chemistry, Nov-15, Volume: 206Targeting dual tolerant regions of binding pocket: Discovery of novel morpholine-substituted diarylpyrimidines as potent HIV-1 NNRTIs with significantly improved water solubility.
AID1763904Antiviral activity against HIV-1 RES056 infected in human MT4 cells assessed as protection against virus-induced cytotoxicity incubated for 5 days by MTT assay2021Bioorganic & medicinal chemistry, 06-15, Volume: 40Design, synthesis and anti-HIV evaluation of novel 5-substituted diarylpyrimidine derivatives as potent HIV-1 NNRTIs.
AID1457052Cytotoxicity against human MT4 cells assessed as cell viability by MTT assay2017Journal of medicinal chemistry, 08-10, Volume: 60, Issue:15
Chiral Indolylarylsulfone Non-Nucleoside Reverse Transcriptase Inhibitors as New Potent and Broad Spectrum Anti-HIV-1 Agents.
AID1811035Anti-viral activity against HIV1 harboring RT Y188L mutant infected MV4 cells assessed as protection against virus-induced cytopathic effect by MTT assay2021Journal of medicinal chemistry, 07-22, Volume: 64, Issue:14
Improving Druggability of Novel Diarylpyrimidine NNRTIs by a Fragment-Based Replacement Strategy: From Biphenyl-DAPYs to Heteroaromatic-Biphenyl-DAPYs.
AID347627Antiviral activity against NNRTI-resistant HIV HXB2 isolate 16 with reverse transcriptase K103, P225 mutation in human MT4 cells assessed as protection against virus-induced cell death after 5 days by MTT assay2008Journal of medicinal chemistry, Dec-11, Volume: 51, Issue:23
Design of annulated pyrazoles as inhibitors of HIV-1 reverse transcriptase.
AID586894Induction of CYP3A5 mRNA expression in human LS180 cells at 0.1 to 1 umol/liter after 4 days by RT-PCR analysis2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1231486Antiviral activity against HIV1 harboring K103N/Y181C double mutant infected in human MT4 cells assessed as protection of cells from virus-induced cytopathic after 5 days by MTT assay2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Anti-HIV diarylpyrimidine-quinolone hybrids and their mode of action.
AID1773440Antiviral activity against HIV1 RES056 infected in human MT4 cells assessed as reduction in virus-induced cytopathogenic effect by MTT assay
AID619642Ratio of EC50 for HIV-1 3B harboring RT E138K mutant infected in human MT4 cells to EC50 for wild type HIV-1 3B infected in human MT4 cells2011European journal of medicinal chemistry, Oct, Volume: 46, Issue:10
Arylazolylthioacetanilide. Part 8: Design, synthesis and biological evaluation of novel 2-(2-(2,4-dichlorophenyl)-2H-1,2,4-triazol-3-ylthio)-N-arylacetamides as potent HIV-1 inhibitors.
AID1357793Antiviral activity against HIV1 harboring reverse transcriptase F227L/V106A double mutant infected in human MT4 cells assessed as inhibition of viral replication after 5 days by MTT assay2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID1298247Inhibition of HIV1 wild type reverse transcriptase p66/p51 using poly(rA) template and measured after 40 mins by picogreen based spectrofluorometry2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
1,6-Bis[(benzyloxy)methyl]uracil derivatives-Novel antivirals with activity against HIV-1 and influenza H1N1 virus.
AID1773828Cytotoxicity against mock-infected human MT4 cells assessed as reduction in cell viability measured after 5 days by MTT assay2021Journal of medicinal chemistry, 09-23, Volume: 64, Issue:18
Structure-Based Design and Discovery of Pyridyl-Bearing Fused Bicyclic HIV-1 Inhibitors: Synthesis, Biological Characterization, and Molecular Modeling Studies.
AID1249714Antiviral activity against HIV1 3B expressing reverse transcriptase K103N + Y181C mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathogenicity incubated for 4 days by MTT method2015European journal of medicinal chemistry, Sep-18, Volume: 102Pyrimidine sulfonylacetanilides with improved potency against key mutant viruses of HIV-1 by specific targeting of a highly conserved residue.
AID1775792Antiviral activity against NNRTI-resistant HIV-1 RES056 harboring RT K103N/Y181C mutant infected in human MT4 cells assessed as protection against virus-induced cytopathogenicity after 5 days by MTT assay2021Journal of medicinal chemistry, 04-08, Volume: 64, Issue:7
2,4,5-Trisubstituted Pyrimidines as Potent HIV-1 NNRTIs: Rational Design, Synthesis, Activity Evaluation, and Crystallographic Studies.
AID236424Area under the concentration time curve in rat after 40 mg/kg oral dosage2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Concentration and pH dependent aggregation of hydrophobic drug molecules and relevance to oral bioavailability.
AID1609108Antiviral activity against HIV-1 harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 4 days by MTT assay2019European journal of medicinal chemistry, Nov-15, Volume: 182Conformational restriction design of thiophene-biphenyl-DAPY HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID709867Antiviral activity against NNRTI-resistant HIV1 harboring reverse transcriptase Y188L mutant infected in human MT4 cells assessed as inhibition of viral replication after 72 hrs by HeLa-CD4-LTR-beta-gal assay2012Journal of medicinal chemistry, Dec-13, Volume: 55, Issue:23
Rational design of potent non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1561711Resistance index, ratio of EC50 for antiviral activity against HIV1 expressing RT Y188L mutant infected in human MT4 cells to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells
AID1691436Antiviral activity against HIV1 RES056 infected in human MT4 cells assessed as reduction in virus-induced cell death incubated for 5 days by MTT assay2020European journal of medicinal chemistry, May-01, Volume: 193In situ click chemistry-based rapid discovery of novel HIV-1 NNRTIs by exploiting the hydrophobic channel and tolerant regions of NNIBP.
AID1483288Fold resistance, ratio of EC50 for reverse transcriptase E138K mutant in human MT4 cells infected HIV1 3B to EC50 for wild type HIV1 3B harboring reverse transcriptase infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1637395Antiviral activity against human HIV-1 3B harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as protection from virus-induced cytopathic effect measured 5 days post infection by MTT assay2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Arylazolyl(azinyl)thioacetanilides. Part 20: Discovery of novel purinylthioacetanilides derivatives as potent HIV-1 NNRTIs via a structure-based bioisosterism approach.
AID619634Antiviral activity against HIV-1 3B harboring RT E138K mutant infected in human MT4 cells assessed as inhibition of virus-induced syncytium formation after 5 days by MTT assay2011European journal of medicinal chemistry, Oct, Volume: 46, Issue:10
Arylazolylthioacetanilide. Part 8: Design, synthesis and biological evaluation of novel 2-(2-(2,4-dichlorophenyl)-2H-1,2,4-triazol-3-ylthio)-N-arylacetamides as potent HIV-1 inhibitors.
AID557063Antiviral activity against HIV1 isolate R8 harboring reverse transcriptase Y181I mutant relative to drug-sensitive HIV1 CNDO2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID1636930Antiviral activity against HIV1 3B harboring reverse transcriptase K103N/Y181C double mutant infected in human MT2 cells assessed as protection against viral infection by MTT method2016Bioorganic & medicinal chemistry, 10-15, Volume: 24, Issue:20
Computer-aided discovery of anti-HIV agents.
AID522377Resistance index, ratio of EC50 for recombinant HIV1 harboring reverse transcriptase V106A/V179D mutant clone to EC50 for wild type HIV12010Antimicrobial agents and chemotherapy, Apr, Volume: 54, Issue:4
Combination of V106I and V179D polymorphic mutations in human immunodeficiency virus type 1 reverse transcriptase confers resistance to efavirenz and nevirapine but not etravirine.
AID1565098Antiviral activity against drug resistant HIV-1 harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1754638Cytotoxicity against mock-infected human MT4 cells incubated for 5 days by MTT assay2021Bioorganic & medicinal chemistry, 07-15, Volume: 42Exploiting the hydrophobic channel of the NNIBP: Discovery of novel diarylpyrimidines as HIV-1 NNRTIs against wild-type and K103N mutant viruses.
AID246343Effective concentration of the compound to inhibit HIV-1 mutant K103N+Y181C replication in HIV-infected MT-4 cells2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
From 4,5,6,7-tetrahydro-5-methylimidazo[4,5,1-jk](1,4)benzodiazepin-2(1H)-one (TIBO) to etravirine (TMC125): fifteen years of research on non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1691437Cytotoxicity against human MT4 cells assessed as reduction in cell viability after 5 days by MTT assay2020European journal of medicinal chemistry, May-01, Volume: 193In situ click chemistry-based rapid discovery of novel HIV-1 NNRTIs by exploiting the hydrophobic channel and tolerant regions of NNIBP.
AID1292010Antiviral activity against HIV1 RES056 harboring reverse transcriptase K103N/YI81C double mutant infected in human MT4 cells assessed as cell viability after 4 days by MTT assay2016European journal of medicinal chemistry, Jun-10, Volume: 115Design, synthesis and evaluation of novel HIV-1 NNRTIs with dual structural conformations targeting the entrance channel of the NNRTI binding pocket.
AID1191402Cytotoxicity against human MT4 cells after 5 days MTT assay2015Bioorganic & medicinal chemistry, Feb-01, Volume: 23, Issue:3
Synthesis and biological evaluation of DAPY-DPEs hybrids as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID347626Antiviral activity against NNRTI-resistant HIV HXB2 isolate 14 with reverse transcriptase A98, K103, V108, M230 mutation in human MT4 cells assessed as protection against virus-induced cell death after 5 days by MTT assay2008Journal of medicinal chemistry, Dec-11, Volume: 51, Issue:23
Design of annulated pyrazoles as inhibitors of HIV-1 reverse transcriptase.
AID1815386Resistance fold, ratio of EC50 for HIV1 harboring reverse transcriptase Y181C mutant infected MV4 cells to EC50 for wild type HIV1 infected MV4 cells2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1206938Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 3B infected in human MT4 cells2015European journal of medicinal chemistry, Jun-05, Volume: 97Discovery of piperidin-4-yl-aminopyrimidine derivatives as potent non-nucleoside HIV-1 reverse transcriptase inhibitors.
AID709869Antiviral activity against NNRTI-resistant HIV1 harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as inhibition of viral replication after 72 hrs by HeLa-CD4-LTR-beta-gal assay2012Journal of medicinal chemistry, Dec-13, Volume: 55, Issue:23
Rational design of potent non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1729156Antiviral activity against HIV1 harboring L100I mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect by MTT assay2021European journal of medicinal chemistry, Mar-05, Volume: 213Exploiting the tolerant region I of the non-nucleoside reverse transcriptase inhibitor (NNRTI) binding pocket. Part 2: Discovery of diarylpyrimidine derivatives as potent HIV-1 NNRTIs with high Fsp
AID518734Inhibition of RNA-dependent DNA polymerase activity of wild type HIV1 subtype B reverse transcriptase by filter-based filtration assay2010Antimicrobial agents and chemotherapy, Jun, Volume: 54, Issue:6
The M230L nonnucleoside reverse transcriptase inhibitor resistance mutation in HIV-1 reverse transcriptase impairs enzymatic function and viral replicative capacity.
AID1572521Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 3B infected in human MT4 cells
AID1165074Antiviral activity against HIV1 3B infected in human MT4 cells assessed as inhibition of virus-induced syncytium formation after 5 days by MTT assay2014European journal of medicinal chemistry, Nov-24, Volume: 87Design, synthesis and anti-HIV evaluation of novel diarylnicotinamide derivatives (DANAs) targeting the entrance channel of the NNRTI binding pocket through structure-guided molecular hybridization.
AID1743621Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells2020European journal of medicinal chemistry, Nov-15, Volume: 206Targeting dual tolerant regions of binding pocket: Discovery of novel morpholine-substituted diarylpyrimidines as potent HIV-1 NNRTIs with significantly improved water solubility.
AID1456308Inhibition of HIV1 RES056 reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect measured at 5 days post infection by MTT assay2017Bioorganic & medicinal chemistry, 04-15, Volume: 25, Issue:8
Structural modifications of diarylpyrimidines (DAPYs) as HIV-1 NNRTIs: Synthesis, anti-HIV activities and SAR.
AID573476Antiviral activity of wild-type Human immunodeficiency virus 1 isolate 5512 by cell based assay2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID1298249Inhibition of HIV1 reverse transcriptase p66/p51 K103N mutant using poly(rA) template and measured after 40 mins by picogreen based spectrofluorometry2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
1,6-Bis[(benzyloxy)methyl]uracil derivatives-Novel antivirals with activity against HIV-1 and influenza H1N1 virus.
AID709870Antiviral activity against HIV1 expressing wild type reverse transcriptase infected in human MT4 cells assessed as inhibition of viral replication after 72 hrs by HeLa-CD4-LTR-beta-gal assay2012Journal of medicinal chemistry, Dec-13, Volume: 55, Issue:23
Rational design of potent non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID508795Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase V090I mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID508647Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase L100I, K103N, E138G mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1171596Inhibition of wild type HIV1 reverse transcriptase L100I mutant assessed as reduction in enzyme activity2014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID383498Partition coefficient, log P of the compound2008European journal of medicinal chemistry, Mar, Volume: 43, Issue:3
SAR and QSAR studies: modelling of new DAPY derivatives.
AID1249721Antiviral activity against HIV1 expressing reverse transcriptase Y181C mutant2015European journal of medicinal chemistry, Sep-18, Volume: 102Pyrimidine sulfonylacetanilides with improved potency against key mutant viruses of HIV-1 by specific targeting of a highly conserved residue.
AID1249715Antiviral activity against HIV2 ROD infected in human MT4 cells assessed as reduction in virus-induced cytopathogenicity incubated for 4 days by MTT method2015European journal of medicinal chemistry, Sep-18, Volume: 102Pyrimidine sulfonylacetanilides with improved potency against key mutant viruses of HIV-1 by specific targeting of a highly conserved residue.
AID508632Antiviral activity against Human immunodeficiency virus 1 subtype G isolate RU132 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1888702Resistance index, ratio of EC50 for antiviral activity against HIV1 with RT Y188L mutant infected in human MT4 cells to EC50 for antiviral activity against wild type HIV1 infected in human MT4 cells
AID1750714Antiviral activity against HIV-1 infected in human MT-4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2021Journal of medicinal chemistry, 04-22, Volume: 64, Issue:8
Hydrophobic Pocket Occupation Design of Difluoro-Biphenyl-Diarylpyrimidines as Non-Nucleoside HIV-1 Reverse Transcriptase Inhibitors: from
AID693583Antiviral activity against wild type HIV1 3B infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2012European journal of medicinal chemistry, Dec, Volume: 58Synthesis, biological evaluation and molecular modeling of 4,6-diarylpyrimidines and diarylbenzenes as novel non-nucleosides HIV-1 reverse transcriptase inhibitors.
AID464770Half life in Beagle dog at 0.5 mg/kg, iv or 0.5 mg/kg, po2010Bioorganic & medicinal chemistry letters, Mar-01, Volume: 20, Issue:5
N1-Heterocyclic pyrimidinediones as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID573481Fold resistance, ratio of EC50 for Human immunodeficiency virus 1 isolate 8117 harboring A98S, G190A mutation in reverse transcriptase to wild-type2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID1249722Antiviral activity against HIV1 expressing reverse transcriptase Y188L mutant2015European journal of medicinal chemistry, Sep-18, Volume: 102Pyrimidine sulfonylacetanilides with improved potency against key mutant viruses of HIV-1 by specific targeting of a highly conserved residue.
AID1775795Selectivity index, ratio of CC50 for cytotoxicity against human MT4 cells reduction in cell viability for 5 days by MTT assay to EC50 for antiviral activity against wild type HIV-1 strain IIIB infected in human MT4 cells assessed as reduction in virus-ind2021Journal of medicinal chemistry, 04-08, Volume: 64, Issue:7
2,4,5-Trisubstituted Pyrimidines as Potent HIV-1 NNRTIs: Rational Design, Synthesis, Activity Evaluation, and Crystallographic Studies.
AID573469Inhibition of DNA-dependent DNA polymerase activity of wild-type Human immunodeficiency virus 1 subtype B reverse transcriptase G190A mutant by filter-based filtration assay2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID1888689Antiviral activity against HIV1 with RT F227L + V106A mutant infected in human MT4 cells cells assessed as protection against viral-induced cytopathogenecity by MTT assay
AID449273Antimalarial activity against Plasmodium falciparum W2mef trophozoite form by hoechst 33342-thiazole orange stain based flow cytometry assay2009Bioorganic & medicinal chemistry letters, Sep-15, Volume: 19, Issue:18
Addressing the malaria drug resistance challenge using flow cytometry to discover new antimalarials.
AID508827Antiviral activity against Human immunodeficiency virus 1 subtype A isolate 92UG037 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID417446Inhibition of HIV1 reverse transcriptase L100I mutant2009European journal of medicinal chemistry, Feb, Volume: 44, Issue:2
QSAR studies for diarylpyrimidines against HIV-1 reverse transcriptase wild-type and mutant strains.
AID1572070Resistance factor, ratio of EC50 for HIV1 harboring reverse transcriptase K103N mutant to EC50 for wild type HIV12019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID557068Plasma protein binding in healthy human at 200 mg/kg2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID1171582Antiviral activity against HIV2 ROD infected in human CEM cells assessed as protection against virus-induced cytopathicity by giant cell formation assay2014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1862561Antiviral activity against HIV-1 IIIB infected in human MT4 cells assessed as reduction in viral replication by MTT assay
AID1207341Inhibition of fast sodium current (INa) in HEK293 cells transfected with human Nav1.5 measured using IonWorks Quattro automated patch clamp platform
AID1691434Inhibition of HIV-1 reverse transcriptase assessed as reduction in biotin-dUTP incorporation in to cDNA chain at 0.267 uM in presence of DIG-dUTP, biotin-dUTP and dTTP measured after 1 hr by ELISA relative to control2020European journal of medicinal chemistry, May-01, Volume: 193In situ click chemistry-based rapid discovery of novel HIV-1 NNRTIs by exploiting the hydrophobic channel and tolerant regions of NNIBP.
AID557037Antiviral activity against HIV1 isolate R8 harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as inhibition of viral replication after 72 hrs in presence of 50% human serum2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID635301Antiviral activity against wild type HIV1 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathogenicity after 5 days by MTT assay2011Bioorganic & medicinal chemistry, Dec-01, Volume: 19, Issue:23
Design, synthesis and biological evaluation of cycloalkyl arylpyrimidines (CAPYs) as HIV-1 NNRTIs.
AID586907Induction of ABCB1 activity2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1773442Cytotoxicity against human MT4 cells assessed as reduction in cell viability by MTT assay
AID1357800Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 harboring reverse transcriptase L100I mutant2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID1352321Antiviral activity against HIV1 RES056 harboring reverse transcriptase K103 N/Y181C double mutant infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 4 days by MTT assay2018European journal of medicinal chemistry, Feb-10, Volume: 145Discovery of biphenyl-substituted diarylpyrimidines as non-nucleoside reverse transcriptase inhibitors with high potency against wild-type and mutant HIV-1.
AID508784Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase E138G mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1819212Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV-1 IIIB infected in human MT4 cells
AID693587Selectivity index, ratio of CC50 for human MT4 cells to EC50 for wild type HIV1 3B infected in human MT4 cells2012European journal of medicinal chemistry, Dec, Volume: 58Synthesis, biological evaluation and molecular modeling of 4,6-diarylpyrimidines and diarylbenzenes as novel non-nucleosides HIV-1 reverse transcriptase inhibitors.
AID1693800Cytotoxicity against human MT4 cells assessed as reduction in cell viability incubated for 5 days by MTT assay2021Bioorganic & medicinal chemistry, 01-15, Volume: 30Novel indolylarylsulfone derivatives as covalent HIV-1 reverse transcriptase inhibitors specifically targeting the drug-resistant mutant Y181C.
AID490372Antiviral activity against HIV1 expressing reverse transcriptase V106A mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect2010Bioorganic & medicinal chemistry letters, Jul-15, Volume: 20, Issue:14
Discovery of piperidin-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-benzyl derivatives with broad potency against resistant mutant viruses.
AID1124989Antiviral activity against HIV1 3B infected in human MT4 cells assessed as virus-induced cytopathic effect by MTT assay2014Bioorganic & medicinal chemistry, Apr-01, Volume: 22, Issue:7
Fused heterocyclic compounds bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 1: design, synthesis and biological evaluation of novel 5,7-disubstituted pyrazolo[1,5-a]pyrimidine derivatives.
AID619643Ratio of EC50 for HIV-1 3B harboring RT L100I mutant infected in human MT4 cells to EC50 for wild type HIV-1 3B infected in human MT4 cells2011European journal of medicinal chemistry, Oct, Volume: 46, Issue:10
Arylazolylthioacetanilide. Part 8: Design, synthesis and biological evaluation of novel 2-(2-(2,4-dichlorophenyl)-2H-1,2,4-triazol-3-ylthio)-N-arylacetamides as potent HIV-1 inhibitors.
AID347631Antiviral activity against NNRTI-resistant HIV HXB2 isolate 29 with reverse transcriptase K103, Y181 mutation in human MT4 cells assessed as protection against virus-induced cell death after 5 days by MTT assay2008Journal of medicinal chemistry, Dec-11, Volume: 51, Issue:23
Design of annulated pyrazoles as inhibitors of HIV-1 reverse transcriptase.
AID693585Antiviral activity against HIV2 ROD infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2012European journal of medicinal chemistry, Dec, Volume: 58Synthesis, biological evaluation and molecular modeling of 4,6-diarylpyrimidines and diarylbenzenes as novel non-nucleosides HIV-1 reverse transcriptase inhibitors.
AID1815394Resistance fold, ratio of EC50 for HIV1 harboring reverse transcriptase F227L/V106A double mutant infected MV4 cells to EC50 for wild type HIV1 infected MV4 cells2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID246344Effective concentration of the compound to inhibit HIV-1 mutant L100I+K103N replication in HIV-infected MT-4 cells2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
From 4,5,6,7-tetrahydro-5-methylimidazo[4,5,1-jk](1,4)benzodiazepin-2(1H)-one (TIBO) to etravirine (TMC125): fifteen years of research on non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1811036Anti-viral activity against HIV1 harboring RT Y181C mutant infected MV4 cells assessed as protection against virus-induced cytopathic effect by MTT assay2021Journal of medicinal chemistry, 07-22, Volume: 64, Issue:14
Improving Druggability of Novel Diarylpyrimidine NNRTIs by a Fragment-Based Replacement Strategy: From Biphenyl-DAPYs to Heteroaromatic-Biphenyl-DAPYs.
AID1882471Cytotoxicity against human MT2 cells assessed as cell viability by MTT assay2022Journal of medicinal chemistry, 03-10, Volume: 65, Issue:5
Contemporary Medicinal Chemistry Strategies for the Discovery and Development of Novel HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors.
AID508644Antiviral activity against Human immunodeficiency virus 1 3B infected in human MT4 cells assessed as reduction in virus induced cytopathicity2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1773447Antiviral activity against wild type HIV1 harboring Y181C mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathogenic effect by MTT assay
AID767498Antiviral activity against HIV1 3B harboring reverse transcriptase K103N/Y181C double mutant infected in human MT2 cells assessed as protection against virus-induced effect by MTT assay2013Bioorganic & medicinal chemistry letters, Sep-15, Volume: 23, Issue:18
Optimization of diarylazines as anti-HIV agents with dramatically enhanced solubility.
AID1687680Antiviral activity against wild type HIV-1 strain 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathogenicity by MTT assay2020European journal of medicinal chemistry, Jan-15, Volume: 186Indazolyl-substituted piperidin-4-yl-aminopyrimidines as HIV-1 NNRTIs: Design, synthesis and biological activities.
AID1731746Inhibition of reverse transcriptase L100I mutant in HIV-1 infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay
AID518739Selectivity ratio of IC50 for DNA-dependent DNA polymerase activity of HIV1 subtype B reverse transcriptase M230L mutant to IC50 for DNA-dependent DNA polymerase activity of wild type HIV1 subtype B reverse transcriptase2010Antimicrobial agents and chemotherapy, Jun, Volume: 54, Issue:6
The M230L nonnucleoside reverse transcriptase inhibitor resistance mutation in HIV-1 reverse transcriptase impairs enzymatic function and viral replicative capacity.
AID1171589Resistance index, ratio of EC50 for HIV1 NL4-3 expressing reverse transcriptase Y181C mutant to EC50 for wild type HIV1 NL4-32014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1880372Antiviral activity against wild type HIV-1 IIIB infected in human MT4 cells assessed as protection against virus-induced cytopathic effect by MTT assay2022Journal of medicinal chemistry, 06-23, Volume: 65, Issue:12
Structure-Based Discovery of Novel NH
AID508793Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase K101E mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID635345Antiviral activity against HIV2 ROD infected in human MT4 cells assessed as reduction in virus-induced cytopathogenicity after 5 days by MTT assay2011Bioorganic & medicinal chemistry, Dec-01, Volume: 19, Issue:23
Design, synthesis and biological evaluation of cycloalkyl arylpyrimidines (CAPYs) as HIV-1 NNRTIs.
AID259754Antiviral activity against wild-type HIV1 in MT2 cells by MTT assay2006Bioorganic & medicinal chemistry letters, Feb, Volume: 16, Issue:3
Computer-aided design of non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID508625Antiviral activity against Human immunodeficiency virus 1 subtype E isolate 93TH073 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID619635Antiviral activity against HIV-1 3B harboring RT K103N mutant infected in human MT4 cells assessed as inhibition of virus-induced syncytium formation after 5 days by MTT assay2011European journal of medicinal chemistry, Oct, Volume: 46, Issue:10
Arylazolylthioacetanilide. Part 8: Design, synthesis and biological evaluation of novel 2-(2-(2,4-dichlorophenyl)-2H-1,2,4-triazol-3-ylthio)-N-arylacetamides as potent HIV-1 inhibitors.
AID1729159Antiviral activity against HIV1 harboring Y188L mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect by MTT assay2021European journal of medicinal chemistry, Mar-05, Volume: 213Exploiting the tolerant region I of the non-nucleoside reverse transcriptase inhibitor (NNRTI) binding pocket. Part 2: Discovery of diarylpyrimidine derivatives as potent HIV-1 NNRTIs with high Fsp
AID1483279Selectivity index, ratio of CC50 for human MT4 cells to EC50 for wild type HIV1 3B harboring reverse transcriptase infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1316335Selectivity index, ratio of CC50 for human mock-infected MT4 cells to EC50 for HIV1 RES056 harboring reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID632804Cytotoxicity against human MT2 cells infected with HIV1 NL4.3 by MTT assay2011Journal of medicinal chemistry, Dec-22, Volume: 54, Issue:24
Computationally-guided optimization of a docking hit to yield catechol diethers as potent anti-HIV agents.
AID573479Antiviral activity of Human immunodeficiency virus 1 isolate 9225 harboring A98S, G190A mutation in reverse transcriptase by cell based assay2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID586880Inhibition of MRP2 expressed in MDCK2 cells assessed as cell growth inhibition by calcein and pheophorbide A efflux assays2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1888691Toxicity in mock-infected human MT4 cells assessed as reduction in cell viability by MTT assay
AID1731744Cytotoxicity against human MT4 cells assessed as reduction in cell viability incubated for 5 days by MTT assay
AID1483287Fold resistance, ratio of EC50 for reverse transcriptase K103N mutant in human MT4 cells infected HIV1 3B to EC50 for wild type HIV1 3B harboring reverse transcriptase infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID726437Antiviral activity against multidrug resistant HIV1 IIIB containing reverse transcriptase K103N and Y181C mutation infected in human MT2 cells assessed as cytoprotection from infection by MTT colorimetric method2013Bioorganic & medicinal chemistry letters, Feb-15, Volume: 23, Issue:4
Optimization of benzyloxazoles as non-nucleoside inhibitors of HIV-1 reverse transcriptase to enhance Y181C potency.
AID1171580Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 NL4-3 infected in human MT4 cells2014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1815388Resistance fold, ratio of EC50 for HIV1 harboring reverse transcriptase K103N mutant infected MV4 cells to EC50 for wild type HIV1 infected MV4 cells2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1574387Inhibition of recombinant HIV-1 His-tagged reverse transcriptase p66/p51 L100I mutant expressed in Escherichia coli assessed as reduction in [3H]dTTP incorporation using poly(rA)/oligo(dT) as template/primer after 20 mins by scintillation counting analysi2019Journal of medicinal chemistry, 01-24, Volume: 62, Issue:2
Effect of α-Methoxy Substitution on the Anti-HIV Activity of Dihydropyrimidin-4(3 H)-ones.
AID1200845Cytotoxicity against mock-infected human MT4 cells assessed as reduction in cell viability after 4 days by MTT assay2015European journal of medicinal chemistry, Mar-26, Volume: 93Fused heterocycles bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 4: design, synthesis and biological evaluation of novel imidazo[1,2-a]pyrazines.
AID1811045Cytotoxicity against mock infected human MT4 cells by MTT assay2021Journal of medicinal chemistry, 07-22, Volume: 64, Issue:14
Improving Druggability of Novel Diarylpyrimidine NNRTIs by a Fragment-Based Replacement Strategy: From Biphenyl-DAPYs to Heteroaromatic-Biphenyl-DAPYs.
AID1609103Cytotoxicity against human MT4 cells assessed as reduction in cell viability incubated for 4 days by MTT assay2019European journal of medicinal chemistry, Nov-15, Volume: 182Conformational restriction design of thiophene-biphenyl-DAPY HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1761007Cytotoxicity against human TZM-bl cells assessed as reduction in cell viability measured after 1 day by CytoTox-Glo assay2021European journal of medicinal chemistry, Feb-05, Volume: 211Design, synthesis, and evaluation of "dual-site"-binding diarylpyrimidines targeting both NNIBP and the NNRTI adjacent site of the HIV-1 reverse transcriptase.
AID632796Antiviral activity against Human immunodeficiency virus 1 NL4.3 reverse transcriptase Y181C mutant infected in human MT2 cells assessed as inhibition of viral infection2011Journal of medicinal chemistry, Dec-22, Volume: 54, Issue:24
Computationally-guided optimization of a docking hit to yield catechol diethers as potent anti-HIV agents.
AID1520061Selectivity index, ratio of CC50 for cytotoxicity against human MT4 cells to EC50 for antiviral activity against HIV1 3B harbouring NNRTI K103N mutant infected in human MT4 cells2020European journal of medicinal chemistry, Jan-01, Volume: 185Fragment hopping-based discovery of novel sulfinylacetamide-diarylpyrimidines (DAPYs) as HIV-1 nonnucleoside reverse transcriptase inhibitors.
AID1421298Inhibition of HIV1 LAI reverse transcriptase infected in human MT4 cells assessed as protection against virus-induced cytopathic effect by MTT assay2018European journal of medicinal chemistry, Oct-05, Volume: 158Advances in diarylpyrimidines and related analogues as HIV-1 nonnucleoside reverse transcriptase inhibitors.
AID1561697Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV1 RES056 infected in human MT4 cells
AID1572525Antiviral activity against HIV1 harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as inhibition of virus-induced cytotoxicity after 5 days by MTT assay
AID1574371Therapeutic index, ratio of CC50 for human MT4 cells to EC50 for HIV-1 NL4-3 infected in human MT4 cells2019Journal of medicinal chemistry, 01-24, Volume: 62, Issue:2
Effect of α-Methoxy Substitution on the Anti-HIV Activity of Dihydropyrimidin-4(3 H)-ones.
AID1763902Antiviral activity against HIV-1 IIIB harboring RT E138K mutant infected in human MT4 cells assessed as protection against virus-induced cytotoxicity incubated for 5 days by MTT assay2021Bioorganic & medicinal chemistry, 06-15, Volume: 40Design, synthesis and anti-HIV evaluation of novel 5-substituted diarylpyrimidine derivatives as potent HIV-1 NNRTIs.
AID1773439Antiviral activity against HIV1 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathogenic effect by MTT assay
AID1882469Antiviral activity against HIV-1 harboring Y181C mutant infected in human MT2 cells assessed as protection against virus-induced cytopathicity by MTT assay2022Journal of medicinal chemistry, 03-10, Volume: 65, Issue:5
Contemporary Medicinal Chemistry Strategies for the Discovery and Development of Novel HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors.
AID632797Antiviral activity against Human immunodeficiency virus 1 NL4.3 reverse transcriptase K103N and Y181C double mutant infected in human MT2 cells assessed as inhibition of viral infection2011Journal of medicinal chemistry, Dec-22, Volume: 54, Issue:24
Computationally-guided optimization of a docking hit to yield catechol diethers as potent anti-HIV agents.
AID665273Antiviral activity against HIV2 ROD mutant infected in MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2012European journal of medicinal chemistry, Jul, Volume: 53Chiral resolution, absolute configuration assignment and biological activity of racemic diarylpyrimidine CH(OH)-DAPY as potent nonnucleoside HIV-1 reverse transcriptase inhibitors.
AID557064Antiviral activity against HIV1 isolate R8 harboring reverse transcriptase F227C mutant relative to drug-sensitive HIV1 CNDO2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID1487263Selectivity index, ratio of CC50 for human MT4 cells to EC50 for wild type HIV1 3B infected in human MT4 cells2017Bioorganic & medicinal chemistry, 08-15, Volume: 25, Issue:16
Discovery of novel DAPY-IAS hybrid derivatives as potential HIV-1 inhibitors using molecular hybridization based on crystallographic overlays.
AID586877Inhibition of MDR1 expressed in MDCK2 cells assessed as inhibition of cell proliferation by calcein and pheophorbide A efflux assays2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1487261Antiviral activity against wild type HIV1 3B infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2017Bioorganic & medicinal chemistry, 08-15, Volume: 25, Issue:16
Discovery of novel DAPY-IAS hybrid derivatives as potential HIV-1 inhibitors using molecular hybridization based on crystallographic overlays.
AID508622Antiviral activity against Human immunodeficiency virus 1 subtype D isolate 92UG024 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1483289Fold resistance, ratio of EC50 for reverse transcriptase L100I mutant in human MT4 cells infected HIV1 3B to EC50 for wild type HIV1 3B harboring reverse transcriptase infected in human MT4 cells2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1558857Antiviral activity against HIV1 harboring reverse transcriptase E138K mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2019Journal of medicinal chemistry, 12-26, Volume: 62, Issue:24
Ligand-Based Design of Nondimethylphenyl-Diarylpyrimidines with Improved Metabolic Stability, Safety, and Oral Pharmacokinetic Profiles.
AID1561694Antiviral activity against HIV1 3B infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1811047Aqueous solubility of compound in PBS at pH 2 by HPLC-UV analysis2021Journal of medicinal chemistry, 07-22, Volume: 64, Issue:14
Improving Druggability of Novel Diarylpyrimidine NNRTIs by a Fragment-Based Replacement Strategy: From Biphenyl-DAPYs to Heteroaromatic-Biphenyl-DAPYs.
AID1391076Inhibition of wild type HIV1 reverse transcriptase using poly (A)/oligo (dT)15 as template/primer assessed as decrease in biotin-dUTP incorporation after 1 hr by ELISA2018Bioorganic & medicinal chemistry, 05-01, Volume: 26, Issue:8
The discovery of novel diarylpyri(mi)dine derivatives with high level activity against a wide variety of HIV-1 strains as well as against HIV-2.
AID1731754Solubility in water at pH 2 at 0.1 to 10 mg
AID635347Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 3B2011Bioorganic & medicinal chemistry, Dec-01, Volume: 19, Issue:23
Design, synthesis and biological evaluation of cycloalkyl arylpyrimidines (CAPYs) as HIV-1 NNRTIs.
AID1457054Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 NL4-3 infected in human MT4 cells2017Journal of medicinal chemistry, 08-10, Volume: 60, Issue:15
Chiral Indolylarylsulfone Non-Nucleoside Reverse Transcriptase Inhibitors as New Potent and Broad Spectrum Anti-HIV-1 Agents.
AID1357805Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 harboring reverse transcriptase F227L/V106A mutant2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID1773838Antiviral activity against HIV1 harboring reverse transcriptase Y188L mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2021Journal of medicinal chemistry, 09-23, Volume: 64, Issue:18
Structure-Based Design and Discovery of Pyridyl-Bearing Fused Bicyclic HIV-1 Inhibitors: Synthesis, Biological Characterization, and Molecular Modeling Studies.
AID726436Cytotoxicity in human MT2 cells assessed as inhibition of cell growth2013Bioorganic & medicinal chemistry letters, Feb-15, Volume: 23, Issue:4
Optimization of benzyloxazoles as non-nucleoside inhibitors of HIV-1 reverse transcriptase to enhance Y181C potency.
AID1880373Cytotoxicity against human MT4 cells assessed as reduction in cell viability by MTT assay2022Journal of medicinal chemistry, 06-23, Volume: 65, Issue:12
Structure-Based Discovery of Novel NH
AID1320864Antiviral activity against HIV1 RES056 expressing reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay2016European journal of medicinal chemistry, Oct-04, Volume: 121Structural optimization of pyridine-type DAPY derivatives to exploit the tolerant regions of the NNRTI binding pocket.
AID1773445Antiviral activity against HIV1 harboring L100I mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathogenic effect by MTT assay
AID1379963Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells2017European journal of medicinal chemistry, Nov-10, Volume: 140Design, synthesis and anti-HIV evaluation of novel diarylpyridine derivatives as potent HIV-1 NNRTIs.
AID1572531Solubility of the compound in deionized water at pH 7
AID1474167Liver toxicity in human assessed as induction of drug-induced liver injury by measuring verified drug-induced liver injury concern status2016Drug discovery today, Apr, Volume: 21, Issue:4
DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans.
AID1822283Inhibition of recombinant wild type p66/p51 HIV1 reverse transcriptase incubated for 40 mins by picogreen dye-based spectrofluorometric analysis2022Journal of medicinal chemistry, 02-10, Volume: 65, Issue:3
Discovery of Novel Pyridine-Dimethyl-Phenyl-DAPY Hybrids by Molecular Fusing of Methyl-Pyrimidine-DAPYs and Difluoro-Pyridinyl-DAPYs: Improving the Druggability toward High Inhibitory Activity, Solubility, Safety, and PK.
AID573482Fold resistance, ratio of EC50 for Human immunodeficiency virus 1 isolate 9225 harboring A98S, G190A mutation in reverse transcriptase to wild-type2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID83551Potency evaluated against NNRTI-Resistant HIV-1 strain Tyr181Cys2004Journal of medicinal chemistry, May-06, Volume: 47, Issue:10
Roles of conformational and positional adaptability in structure-based design of TMC125-R165335 (etravirine) and related non-nucleoside reverse transcriptase inhibitors that are highly potent and effective against wild-type and drug-resistant HIV-1 varian
AID1124990Antiviral activity against HIV2 ROD infected in human MT4 cells assessed as virus-induced cytopathic effect by MTT assay2014Bioorganic & medicinal chemistry, Apr-01, Volume: 22, Issue:7
Fused heterocyclic compounds bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 1: design, synthesis and biological evaluation of novel 5,7-disubstituted pyrazolo[1,5-a]pyrimidine derivatives.
AID1591876Solubility of the compound in water at pH 72019Bioorganic & medicinal chemistry letters, 08-15, Volume: 29, Issue:16
Molecular and cellular studies evaluating a potent 2-cyanoindolizine catechol diether NNRTI targeting wildtype and Y181C mutant HIV-1 reverse transcriptase.
AID573477Antiviral activity of Human immunodeficiency virus 1 isolate 8116 harboring A98S, G190A mutation in reverse transcriptase by cell based assay2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID1583029Antiviral activity against HIV1 containing reverse transcriptase F227L+V106A mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2020Journal of medicinal chemistry, 02-13, Volume: 63, Issue:3
Discovery and Characterization of Fluorine-Substituted Diarylpyrimidine Derivatives as Novel HIV-1 NNRTIs with Highly Improved Resistance Profiles and Low Activity for the hERG Ion Channel.
AID1457065Inhibition of HIV1 reverse transcriptase p66 Y181I mutant associated RNA dependent DNA polymerase activity expressed in Escherichia coli assessed as reduction in [3H]dTTP incorporation using poly(rA)/oligo(dT) as template/primer by scintillation counting 2017Journal of medicinal chemistry, 08-10, Volume: 60, Issue:15
Chiral Indolylarylsulfone Non-Nucleoside Reverse Transcriptase Inhibitors as New Potent and Broad Spectrum Anti-HIV-1 Agents.
AID1558853Antiviral activity against HIV1 harboring reverse transcriptase L100I mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2019Journal of medicinal chemistry, 12-26, Volume: 62, Issue:24
Ligand-Based Design of Nondimethylphenyl-Diarylpyrimidines with Improved Metabolic Stability, Safety, and Oral Pharmacokinetic Profiles.
AID1862563Cytotoxicity against human MT4 cells assessed as reduction on cell growth by MTT assay
AID584239Antiviral activity against HIV1 NL4-3 harboring reverse transcriptase Y181C, V179V/D mutant derived from 17 days viral passages with lersivirine infected in human SupT1 cells assessed as inhibition of viral replication after 21 days relative to wild type 2010Antimicrobial agents and chemotherapy, Oct, Volume: 54, Issue:10
Lersivirine, a nonnucleoside reverse transcriptase inhibitor with activity against drug-resistant human immunodeficiency virus type 1.
AID1637404Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for human HIV-1 3B harboring F227L/V106A double mutant infected in human MT4 cells2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Arylazolyl(azinyl)thioacetanilides. Part 20: Discovery of novel purinylthioacetanilides derivatives as potent HIV-1 NNRTIs via a structure-based bioisosterism approach.
AID1572068Selectivity ratio of CC50 for human MT4 cells to EC50 for HIV1 harboring reverse transcriptase F227L/V106A double mutant infected in human MT4 cells2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID106049Inhibitory activity against 100I strain and 103N strain of HIV-I in MT-4 cells2001Bioorganic & medicinal chemistry letters, Sep-03, Volume: 11, Issue:17
Evolution of anti-HIV drug candidates. Part 3: Diarylpyrimidine (DAPY) analogues.
AID573473Antiviral activity of wild-type Human immunodeficiency virus 1 harboring Y181C mutation in reverse transcriptase infected in human TZM-bl cells human assessed as inhibition of viral replication by luciferase reporter gene assay2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID1191401Antiviral activity against wild type HIV-2 ROD infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 5 days by MTT assay2015Bioorganic & medicinal chemistry, Feb-01, Volume: 23, Issue:3
Synthesis and biological evaluation of DAPY-DPEs hybrids as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1880379Antiviral activity against HIV-1 IIIB harboring reverse transcriptase Y188L mutant infected in human MT4 cells by MTT assay2022Journal of medicinal chemistry, 06-23, Volume: 65, Issue:12
Structure-Based Discovery of Novel NH
AID449275Cytotoxicity against human erythrocytes assessed as intact cells by propidium iodide stain based flow cytometry2009Bioorganic & medicinal chemistry letters, Sep-15, Volume: 19, Issue:18
Addressing the malaria drug resistance challenge using flow cytometry to discover new antimalarials.
AID508649Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase L100I, K103N, V179L mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1200846Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for HIV1 3B infected in human MT4 cells2015European journal of medicinal chemistry, Mar-26, Volume: 93Fused heterocycles bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 4: design, synthesis and biological evaluation of novel imidazo[1,2-a]pyrazines.
AID533675Trough drug concentration at 12 hrs in healthy human plasma at 200 mg, po twice a day for 4 days coadministered with 400 mg of raltegravir, po twice a day for 4 days by reverse-phase HPLC-MS method2008Antimicrobial agents and chemotherapy, Dec, Volume: 52, Issue:12
Minimal pharmacokinetic interaction between the human immunodeficiency virus nonnucleoside reverse transcriptase inhibitor etravirine and the integrase inhibitor raltegravir in healthy subjects.
AID1754643Antiviral activity against HIV1 harboring RT Y181C mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect incubated for 5 days by MTT assay2021Bioorganic & medicinal chemistry, 07-15, Volume: 42Exploiting the hydrophobic channel of the NNIBP: Discovery of novel diarylpyrimidines as HIV-1 NNRTIs against wild-type and K103N mutant viruses.
AID1572063Selectivity ratio of CC50 for human MT4 cells to EC50 for HIV1 harboring reverse transcriptase L100I mutant infected in human MT4 cells2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1637388Antiviral activity against human HIV-1 3B infected in human MT4 cells assessed as protection from virus-induced cytopathic effect measured 5 days post infection by MTT assay2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Arylazolyl(azinyl)thioacetanilides. Part 20: Discovery of novel purinylthioacetanilides derivatives as potent HIV-1 NNRTIs via a structure-based bioisosterism approach.
AID464767Binding affinity to human serum albumin2010Bioorganic & medicinal chemistry letters, Mar-01, Volume: 20, Issue:5
N1-Heterocyclic pyrimidinediones as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID508752Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase V179I, Y181C mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1410485Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 RES056 reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells2018ACS medicinal chemistry letters, Apr-12, Volume: 9, Issue:4
Further Exploring Solvent-Exposed Tolerant Regions of Allosteric Binding Pocket for Novel HIV-1 NNRTIs Discovery.
AID1561703Antiviral activity against HIV1 expressing RT F227L + V106A mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay
AID701102Antiviral activity against wild type HIV12012Journal of medicinal chemistry, Apr-26, Volume: 55, Issue:8
Strategies for the design of HIV-1 non-nucleoside reverse transcriptase inhibitors: lessons from the development of seven representative paradigms.
AID1743627Antiviral activity against HIV1 harboring RT K103N mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Nov-15, Volume: 206Targeting dual tolerant regions of binding pocket: Discovery of novel morpholine-substituted diarylpyrimidines as potent HIV-1 NNRTIs with significantly improved water solubility.
AID586888Induction of ABCC5 mRNA expression in human LS180 cells at 0.1 to 1 umol/liter after 4 days by RT-PCR analysis2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID586902Activity at MRP22011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1357802Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 harboring reverse transcriptase E138K mutant2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID1163234Cytotoxicity against human TZM-bl cells assessed as cell viability after 48 hrs by WST1 assay2014Bioorganic & medicinal chemistry, Oct-01, Volume: 22, Issue:19
From human immunodeficiency virus non-nucleoside reverse transcriptase inhibitors to potent and selective antitrypanosomal compounds.
AID1410411Antiviral activity against HIV1 harboring reverse transcriptase E138K mutant infected in human MT4 cells assessed as inhibition of viral replication after 5 days by MTT assay2018ACS medicinal chemistry letters, Apr-12, Volume: 9, Issue:4
Discovery of Novel Diarylpyrimidine Derivatives as Potent HIV-1 NNRTIs Targeting the "NNRTI Adjacent" Binding Site.
AID1637391Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for human HIV-1 3B infected in human MT4 cells2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Arylazolyl(azinyl)thioacetanilides. Part 20: Discovery of novel purinylthioacetanilides derivatives as potent HIV-1 NNRTIs via a structure-based bioisosterism approach.
AID496632Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase V90I/V35T, T39L, K122E, D123N, S162A, E169T, K173E, Q174E, D177E,T200A, I202V, Q207E mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID738332Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for wild type HIV1 3B2013Bioorganic & medicinal chemistry, Apr-01, Volume: 21, Issue:7
Synthesis and biological evaluation of pyridazine derivatives as novel HIV-1 NNRTIs.
AID484274Inhibition of Trypanosoma cruzi cruzaine preincubated for 5 mins before substrate addition by fluorescence assay in absence of Triton X-1002010Journal of medicinal chemistry, May-27, Volume: 53, Issue:10
Colloid formation by drugs in simulated intestinal fluid.
AID1565090Antiviral activity against HIV-1 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1741394Inhibition of reverse transcriptase K103N mutant in HIV-1 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Sep-15, Volume: 202Bioisosterism-based design and enantiomeric profiling of chiral hydroxyl-substituted biphenyl-diarylpyrimidine nonnucleoside HIV-1 reverse transcriptase inhibitors.
AID1446815Antiviral activity against HIV1 harboring L100I mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect measured at 5 days post infection by MTT assay2017European journal of medicinal chemistry, Apr-21, Volume: 130Discovery of uracil-bearing DAPYs derivatives as novel HIV-1 NNRTIs via crystallographic overlay-based molecular hybridization.
AID1249713Antiviral activity against HIV1 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathogenicity incubated for 4 days by MTT method2015European journal of medicinal chemistry, Sep-18, Volume: 102Pyrimidine sulfonylacetanilides with improved potency against key mutant viruses of HIV-1 by specific targeting of a highly conserved residue.
AID496621Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase K101E/V35T, E36D, K49R, V60I, K122P, I135R, S162A, E169R, K173T, Q174E,D177E, V189I, T200A, I202V, Q207E, R211K, V245Q mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID1882470Antiviral activity against HIV-1 harboring K103N/Y181C mutant infected in human MT2 cells assessed as protection against virus-induced cytopathicity by MTT assay2022Journal of medicinal chemistry, 03-10, Volume: 65, Issue:5
Contemporary Medicinal Chemistry Strategies for the Discovery and Development of Novel HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors.
AID1822279Antiviral activity against HIV-1 harboring Y188L mutant infected in human MT4 cells assessed as protection against virus-induced cytotoxicity incubated for 5 days by MTT assay2022Journal of medicinal chemistry, 02-10, Volume: 65, Issue:3
Discovery of Novel Pyridine-Dimethyl-Phenyl-DAPY Hybrids by Molecular Fusing of Methyl-Pyrimidine-DAPYs and Difluoro-Pyridinyl-DAPYs: Improving the Druggability toward High Inhibitory Activity, Solubility, Safety, and PK.
AID1572061Antiviral activity against HIV1 harboring reverse transcriptase E138K mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect after 5 days by MTT assay2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID496627Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase E138A/K32R, V35E, T39S, S48T, V60I, D121Y, K122E, I135T, S162A, K173A, Q174K, D177E, V179I, T200E, Q207D, R211K mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID1572057Antiviral activity against HIV1 harboring reverse transcriptase L100I mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect after 5 days by MTT assay2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1391081Antiviral activity against HIV1 harboring reverse transcriptase L100I mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay2018Bioorganic & medicinal chemistry, 05-01, Volume: 26, Issue:8
The discovery of novel diarylpyri(mi)dine derivatives with high level activity against a wide variety of HIV-1 strains as well as against HIV-2.
AID1157593Antiviral activity against MC1220-resistant HIV1 harboring RT 100I, 179D, 181C mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathogenicity after 4 days by MTT assay2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Synthesis of novel fluoro analogues of MKC442 as microbicides.
AID83552Potency evaluated against NNRTI-Resistant HIV-1 strain Tyr188Leu2004Journal of medicinal chemistry, May-06, Volume: 47, Issue:10
Roles of conformational and positional adaptability in structure-based design of TMC125-R165335 (etravirine) and related non-nucleoside reverse transcriptase inhibitors that are highly potent and effective against wild-type and drug-resistant HIV-1 varian
AID1594856Half life in human serum at 200 mg, bid2019Journal of medicinal chemistry, 05-23, Volume: 62, Issue:10
The Journey of HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors (NNRTIs) from Lab to Clinic.
AID1773452Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 harboring K103N mutant infected in human MT4 cells
AID464768Half life in human microsomes2010Bioorganic & medicinal chemistry letters, Mar-01, Volume: 20, Issue:5
N1-Heterocyclic pyrimidinediones as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1418466Cytotoxicity against human MT4 cells assessed as reduction in cell viability after 4 days by MTT assay2018Bioorganic & medicinal chemistry letters, 12-01, Volume: 28, Issue:22
Design and synthesis of a novel series of non-nucleoside HIV-1 inhibitors bearing pyrimidine and N-substituted aromatic piperazine.
AID1171588Resistance index, ratio of EC50 for HIV1 NL4-3 expressing reverse transcriptase K103N mutant to EC50 for wild type HIV1 NL4-32014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1880376Antiviral activity against HIV-1 IIIB harboring reverse transcriptase L1001 mutant infected in human MT4 cells by MTT assay2022Journal of medicinal chemistry, 06-23, Volume: 65, Issue:12
Structure-Based Discovery of Novel NH
AID1504696Cytotoxicity against human MT4 cells assessed as reduction in cell viability after 5 days by MTT assay2017ACS medicinal chemistry letters, Nov-09, Volume: 8, Issue:11
Discovery of Thiophene[3,2-
AID557029Ratio of EC95 for HIV1 isolate R8 harboring reverse transcriptase K103N mutant in presence of 50% human serum to EC95 for HIV1 isolate R8 harboring wild type reverse transcriptase in presence of 50% human serum2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID1750716Selectivity index, ratio of CC50 for human MT-4 cells to EC50 for HIV-1 infected in MT-4 cells2021Journal of medicinal chemistry, 04-22, Volume: 64, Issue:8
Hydrophobic Pocket Occupation Design of Difluoro-Biphenyl-Diarylpyrimidines as Non-Nucleoside HIV-1 Reverse Transcriptase Inhibitors: from
AID1171579Antiviral activity against HIV1 NL4-3 infected in human MT4 cells assessed as reduction in virus-induced cell death by MTT assay2014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1197832Antiviral activity against HIV1 RES056 expressing reverse transcriptase K103N + Y181C double mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect after 5 days by MTT assay2015European journal of medicinal chemistry, Mar-06, Volume: 92Fused heterocycles bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 3: optimization of [1,2,4]triazolo[1,5-a]pyrimidine core via structure-based and physicochemical property-driven approaches.
AID1819210Antiviral activity against HIV-1 IIIB harboring K103N/Y181C double mutant infected in human MT4 cells assessed as protection against virus-induced cytotoxicity incubated for 5 days by MTT assay
AID508773Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase Y181V mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1884486Antiviral activity against NNRTI resistant wild type HIV-1 infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Expansion of the S-CN-DABO scaffold to exploit the impact on inhibitory activities against the non-nucleoside HIV-1 reverse transcriptase.
AID490369Antiviral activity against wild type HIV1 infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect in presence of 40 % human serum2010Bioorganic & medicinal chemistry letters, Jul-15, Volume: 20, Issue:14
Discovery of piperidin-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-benzyl derivatives with broad potency against resistant mutant viruses.
AID1152373Cytotoxicity against human MT4 cells by MTT assay2014Bioorganic & medicinal chemistry, Jun-15, Volume: 22, Issue:12
Synthesis and biological evaluation of CHX-DAPYs as HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1565100Antiviral activity against drug resistant HIV-1 harboring reverse transcriptase F227L/V106A double mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1743638Resistance index, ratio of EC50 for antiviral activity against HIV1 expressing RT E138K mutant infected in human MT4 cells to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells2020European journal of medicinal chemistry, Nov-15, Volume: 206Targeting dual tolerant regions of binding pocket: Discovery of novel morpholine-substituted diarylpyrimidines as potent HIV-1 NNRTIs with significantly improved water solubility.
AID1069126Antiviral activity against wild-type HIV infected in human MT4 cells assessed as inhibition of viral infection in presence of 50% normal human serum2014Bioorganic & medicinal chemistry letters, Feb-01, Volume: 24, Issue:3
Discovery of MK-1439, an orally bioavailable non-nucleoside reverse transcriptase inhibitor potent against a wide range of resistant mutant HIV viruses.
AID1352316Antiviral activity against HIV1 harboring reverse transcriptase L100I mutant infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 4 days by MTT assay2018European journal of medicinal chemistry, Feb-10, Volume: 145Discovery of biphenyl-substituted diarylpyrimidines as non-nucleoside reverse transcriptase inhibitors with high potency against wild-type and mutant HIV-1.
AID1819218Antiviral activity against HIV-1 harboring reverse transcriptase E138K mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1741400Inhibition of recombinant HIV-1 reverse transcriptase p66/p51 incubated for 40 mins by picogreen dye based fluorescence assay2020European journal of medicinal chemistry, Sep-15, Volume: 202Bioisosterism-based design and enantiomeric profiling of chiral hydroxyl-substituted biphenyl-diarylpyrimidine nonnucleoside HIV-1 reverse transcriptase inhibitors.
AID1249716Cytotoxicity against human MT4 cells assessed as reduction in cell viability incubated for 4 days by MTT assay2015European journal of medicinal chemistry, Sep-18, Volume: 102Pyrimidine sulfonylacetanilides with improved potency against key mutant viruses of HIV-1 by specific targeting of a highly conserved residue.
AID1483275Inhibition of HIV1 RES056 reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as protection against virus induced cytotoxicity after 5 days by MTT assay2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1561710Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV1 expressing RT Y188L mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect
AID508789Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase K103S mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1729149Antiviral activity against HIV1 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect by MTT assay2021European journal of medicinal chemistry, Mar-05, Volume: 213Exploiting the tolerant region I of the non-nucleoside reverse transcriptase inhibitor (NNRTI) binding pocket. Part 2: Discovery of diarylpyrimidine derivatives as potent HIV-1 NNRTIs with high Fsp
AID1636146Antiviral activity against wild type HIV1 NL4-32016Journal of medicinal chemistry, Aug-11, Volume: 59, Issue:15
Discovery and Structure-Based Optimization of 2-Ureidothiophene-3-carboxylic Acids as Dual Bacterial RNA Polymerase and Viral Reverse Transcriptase Inhibitors.
AID1862567Antiviral activity against HIV-1 Y181C mutant infected in human MT4 cells assessed as reduction in viral replication by MTT assay
AID1741395Inhibition of reverse transcriptase Y181C mutant in HIV-1 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Sep-15, Volume: 202Bioisosterism-based design and enantiomeric profiling of chiral hydroxyl-substituted biphenyl-diarylpyrimidine nonnucleoside HIV-1 reverse transcriptase inhibitors.
AID1741392Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV IIIB2020European journal of medicinal chemistry, Sep-15, Volume: 202Bioisosterism-based design and enantiomeric profiling of chiral hydroxyl-substituted biphenyl-diarylpyrimidine nonnucleoside HIV-1 reverse transcriptase inhibitors.
AID709858Antiviral activity against NNRTI-resistant HIV1 harboring reverse transcriptase Y181I/V179F double mutant infected in human MT4 cells assessed as inhibition of viral replication after 72 hrs by HeLa-CD4-LTR-beta-gal assay2012Journal of medicinal chemistry, Dec-13, Volume: 55, Issue:23
Rational design of potent non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID518740Antiviral activity against HIV 1 subtype B harboring wild type reverse transcriptase infected in human TZM-bl cells assessed as inhibition of viral growth by luciferase reporter gene assay2010Antimicrobial agents and chemotherapy, Jun, Volume: 54, Issue:6
The M230L nonnucleoside reverse transcriptase inhibitor resistance mutation in HIV-1 reverse transcriptase impairs enzymatic function and viral replicative capacity.
AID1357797Resistance factor, ratio of EC50 for HIV1 harboring reverse transcriptase Y181C mutant to EC50 for wild-type HIV-1 3B2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID1574376Antiviral activity against HIV-1 harboring reverse transcriptase Y188L mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2019Journal of medicinal chemistry, 01-24, Volume: 62, Issue:2
Effect of α-Methoxy Substitution on the Anti-HIV Activity of Dihydropyrimidin-4(3 H)-ones.
AID1743622Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV1 RES056 infected in human MT4 cells2020European journal of medicinal chemistry, Nov-15, Volume: 206Targeting dual tolerant regions of binding pocket: Discovery of novel morpholine-substituted diarylpyrimidines as potent HIV-1 NNRTIs with significantly improved water solubility.
AID508755Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase V179E, Y181C mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID709865Antiviral activity against NNRTI-resistant HIV1 harboring reverse transcriptase Y181C/F227C double mutant infected in human MT4 cells assessed as inhibition of viral replication after 72 hrs by HeLa-CD4-LTR-beta-gal assay2012Journal of medicinal chemistry, Dec-13, Volume: 55, Issue:23
Rational design of potent non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1275547Antiviral activity against HIV1 3B expressing reverse transcriptase K103N mutant infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect after 5 days by MTT assay2016European journal of medicinal chemistry, Feb-15, Volume: 109Design, synthesis and anti-HIV evaluation of novel diarylpyridine derivatives targeting the entrance channel of NNRTI binding pocket.
AID1504694Antiviral activity against wild type HIV1 3B infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2017ACS medicinal chemistry letters, Nov-09, Volume: 8, Issue:11
Discovery of Thiophene[3,2-
AID573467Inhibition of Human immunodeficiency virus 1 subtype B reverse transcriptase G190A-81C mutant by filter-based filtration assay2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID1457064Inhibition of HIV1 reverse transcriptase p66 K103N mutant associated RNA dependent DNA polymerase activity expressed in Escherichia coli assessed as reduction in [3H]dTTP incorporation using poly(rA)/oligo(dT) as template/primer by scintillation counting 2017Journal of medicinal chemistry, 08-10, Volume: 60, Issue:15
Chiral Indolylarylsulfone Non-Nucleoside Reverse Transcriptase Inhibitors as New Potent and Broad Spectrum Anti-HIV-1 Agents.
AID1443667Antiviral activity against HIV1 harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as inhibition of viral replication after 5 days by MTT assay2017Bioorganic & medicinal chemistry letters, 04-15, Volume: 27, Issue:8
Design and synthesis of hybrids of diarylpyrimidines and diketo acids as HIV-1 inhibitors.
AID508798Ratio of EC50 for HIV1 in presence of 50% human serum to EC50 for HIV1 in absence of serum proteins by GFP assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1443664Selectivity index, ratio of CC50 for human MT4 cells to EC50 for wild type HIV1 3B2017Bioorganic & medicinal chemistry letters, 04-15, Volume: 27, Issue:8
Design and synthesis of hybrids of diarylpyrimidines and diketo acids as HIV-1 inhibitors.
AID508787Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase V106M mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1561696Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells
AID236595pKa value was determined2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Concentration and pH dependent aggregation of hydrophobic drug molecules and relevance to oral bioavailability.
AID1572078Aqueous solubility of the compound in FASSIF by HPLC analysis2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1596751Antiviral activity against wild type HIV-1 3B infected in MT4 cells measured after 5 days by MTT assay2019European journal of medicinal chemistry, Aug-15, Volume: 176Synthesis and biological evaluation of dihydroquinazoline-2-amines as potent non-nucleoside reverse transcriptase inhibitors of wild-type and mutant HIV-1 strains.
AID1888692Selectivity index, ratio of CC50 for toxicity in mock-infected human MT4 cells assessed as reduction in cell viability by MTT assay to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells cells assessed as protection against viral-induc
AID496630Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase Y181C/V35T, V60I, I135V, S162A, K173T, Q174K, N175Y, D177E, T200A, Q207E, R211K, H221Y mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID517499Antiviral activity against HIV1 harboring reverse transcriptase K103N/L100I double mutant infected in human MT4 cells assessed as reduction of virus-induced cytopathic effect by replication assay in presence of 10% FBS2010Bioorganic & medicinal chemistry letters, Oct-15, Volume: 20, Issue:20
Exploration of piperidine-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-Phenyl derivatives with broad potency against resistant mutant viruses.
AID1884538Fold resistance, ratio of EC50 for antiviral activity against drug-resistant HIV-1 L100I mutant infected in human MT4 cells to EC50 for antiviral activity against NNRTI resistant wild type HIV-1 infected in human MT4 cells2022European journal of medicinal chemistry, Aug-05, Volume: 238Expansion of the S-CN-DABO scaffold to exploit the impact on inhibitory activities against the non-nucleoside HIV-1 reverse transcriptase.
AID1729151Cytotoxicity against human MT4 cells assessed as reduction in cell viability by MTT assay2021European journal of medicinal chemistry, Mar-05, Volume: 213Exploiting the tolerant region I of the non-nucleoside reverse transcriptase inhibitor (NNRTI) binding pocket. Part 2: Discovery of diarylpyrimidine derivatives as potent HIV-1 NNRTIs with high Fsp
AID1171590Resistance index, ratio of EC50 for HIV1 NL4-3 expressing reverse transcriptase Y188L mutant to EC50 for wild type HIV1 NL4-32014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID767497Cytotoxicity against human MT2 cells assessed as growth inhibition2013Bioorganic & medicinal chemistry letters, Sep-15, Volume: 23, Issue:18
Optimization of diarylazines as anti-HIV agents with dramatically enhanced solubility.
AID508760Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase K103N, V108I mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1705191Resistance index, ratio of EC50 for antiviral activity against HIV1 harboring RT Y181C mutant infected in human MT4 cells to EC50 for antiviral activity against HIV1 NL4-3 infected in human MT4 cells2020European journal of medicinal chemistry, Dec-15, Volume: 208New indolylarylsulfone non-nucleoside reverse transcriptase inhibitors show low nanomolar inhibition of single and double HIV-1 mutant strains.
AID1574384Inhibition of recombinant wild type HIV-1 His-tagged reverse transcriptase p66/p51 expressed in Escherichia coli assessed as reduction in [3H]dTTP incorporation using poly(rA)/oligo(dT) as template/primer after 20 mins by scintillation counting analysis2019Journal of medicinal chemistry, 01-24, Volume: 62, Issue:2
Effect of α-Methoxy Substitution on the Anti-HIV Activity of Dihydropyrimidin-4(3 H)-ones.
AID573478Antiviral activity of Human immunodeficiency virus 1 isolate 8117 harboring A98S, G190A mutation in reverse transcriptase by cell based assay2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID1773446Antiviral activity against HIV1 harboring K103N mutant infected in human MT4 cells assessed as reduction in virus-induced cytopathogenic effect by MTT assay
AID1761012Selectivity index, ratio of CC50 for human MT4 cells to EC50 for wild type HIV-1 3B infected in human MT4 cells2021European journal of medicinal chemistry, Feb-05, Volume: 211Design, synthesis, and evaluation of "dual-site"-binding diarylpyrimidines targeting both NNIBP and the NNRTI adjacent site of the HIV-1 reverse transcriptase.
AID1316338Antiviral activity against HIV1 3B harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as protection against virus induced cytotoxicity after 5 days by MTT assay2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Design, Synthesis, and Evaluation of Thiophene[3,2-d]pyrimidine Derivatives as HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Significantly Improved Drug Resistance Profiles.
AID557028Ratio of EC95 for HIV1 isolate R8 harboring reverse transcriptase K103N and Y181C mutant in presence of 50% human serum to EC95 for HIV1 isolate R8 harboring wild type reverse transcriptase in presence of 50% human serum2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID1815391Inhibition of HIV1 reverse transcriptase K103N/Y181C double mutant using oligo(dT)16 as primer measured after 40 mins by picogreen-dye based spectrofluorimetric analysis2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1583025Antiviral activity against HIV1 containing reverse transcriptase K103N mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay2020Journal of medicinal chemistry, 02-13, Volume: 63, Issue:3
Discovery and Characterization of Fluorine-Substituted Diarylpyrimidine Derivatives as Novel HIV-1 NNRTIs with Highly Improved Resistance Profiles and Low Activity for the hERG Ion Channel.
AID1815397Inhibition of HIV1 reverse transcriptase L100I mutant using oligo(dT)16 as primer measured after 40 mins by picogreen-dye based spectrofluorimetric analysis2021European journal of medicinal chemistry, Dec-15, Volume: 226Design of the naphthyl-diarylpyrimidines as potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) via structure-based extension into the entrance channel.
AID1775800Antiviral activity against HIV-1 harboring RT Y188L mutant infected in human MT4 cells assessed as protection against virus-induced cytopathogenicity after 5 days by MTT assay2021Journal of medicinal chemistry, 04-08, Volume: 64, Issue:7
2,4,5-Trisubstituted Pyrimidines as Potent HIV-1 NNRTIs: Rational Design, Synthesis, Activity Evaluation, and Crystallographic Studies.
AID1391088Inhibition of HIV1 reverse transcriptase K103N/Y181C double mutant using poly (A)/oligo (dT)15 as template/primer assessed as decrease in biotin-dUTP incorporation after 1 hr by ELISA2018Bioorganic & medicinal chemistry, 05-01, Volume: 26, Issue:8
The discovery of novel diarylpyri(mi)dine derivatives with high level activity against a wide variety of HIV-1 strains as well as against HIV-2.
AID1741390Inhibition of reverse transcriptase in HIV-1 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Sep-15, Volume: 202Bioisosterism-based design and enantiomeric profiling of chiral hydroxyl-substituted biphenyl-diarylpyrimidine nonnucleoside HIV-1 reverse transcriptase inhibitors.
AID573472Antiviral activity of wild-type Human immunodeficiency virus 1 infected in human TZM-bl cells human assessed as inhibition of viral replication by luciferase reporter gene assay2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID619640Antiviral activity against HIV-1 3B harboring RT K103N and Y181C mutant infected in human MT4 cells assessed as inhibition of virus-induced syncytium formation after 5 days by MTT assay2011European journal of medicinal chemistry, Oct, Volume: 46, Issue:10
Arylazolylthioacetanilide. Part 8: Design, synthesis and biological evaluation of novel 2-(2-(2,4-dichlorophenyl)-2H-1,2,4-triazol-3-ylthio)-N-arylacetamides as potent HIV-1 inhibitors.
AID1558858Inhibition of recombinant wild type HIV1 reverse transcriptase p66/p51 incubated for 40 mins by picogreen dye-based spectrofluorometric analysis2019Journal of medicinal chemistry, 12-26, Volume: 62, Issue:24
Ligand-Based Design of Nondimethylphenyl-Diarylpyrimidines with Improved Metabolic Stability, Safety, and Oral Pharmacokinetic Profiles.
AID508766Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase M230V mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID508770Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase G190S mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID614140Cytotoxicity against human MT4 cells after 4 days by MTT assay2011Bioorganic & medicinal chemistry, Sep-01, Volume: 19, Issue:17
Synthesis and structure-activity relationship of novel diarylpyrimidines with hydromethyl linker (CH(OH)-DAPYs) as HIV-1 NNRTIs.
AID1504700Antiviral activity against HIV1 3B harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2017ACS medicinal chemistry letters, Nov-09, Volume: 8, Issue:11
Discovery of Thiophene[3,2-
AID1773451Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 harboring L1001 mutant infected in human MT4 cells
AID1197829Antiviral activity against HIV1 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect after 5 days by MTT assay2015European journal of medicinal chemistry, Mar-06, Volume: 92Fused heterocycles bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 3: optimization of [1,2,4]triazolo[1,5-a]pyrimidine core via structure-based and physicochemical property-driven approaches.
AID1773843Inhibition of wild type HIV1 p66/p51 reverse transcriptase using poly(rA) as template, oligo(dT)16 as primer and RNA/DNA as substrate measured after 40 mins by PICOGreen-dye based spectrofluorimetry analysis2021Journal of medicinal chemistry, 09-23, Volume: 64, Issue:18
Structure-Based Design and Discovery of Pyridyl-Bearing Fused Bicyclic HIV-1 Inhibitors: Synthesis, Biological Characterization, and Molecular Modeling Studies.
AID665274Cytotoxicity against human MT4 cells after 5 days by MTT assay2012European journal of medicinal chemistry, Jul, Volume: 53Chiral resolution, absolute configuration assignment and biological activity of racemic diarylpyrimidine CH(OH)-DAPY as potent nonnucleoside HIV-1 reverse transcriptase inhibitors.
AID1379964Inhibition of recombinant wild type HIV1 reverse transcriptase p66/p51 assessed as reduction in biotin-dUTP incorporation incubated for 40 mins using poly(rA) template and oligo(dT)16 primer by picogreen dye based spectrofluorometry2017European journal of medicinal chemistry, Nov-10, Volume: 140Design, synthesis and anti-HIV evaluation of novel diarylpyridine derivatives as potent HIV-1 NNRTIs.
AID1763903Antiviral activity against HIV-1 IIIB harboring RT F227L/V106A mutant infected in human MT4 cells assessed as protection against virus-induced cytotoxicity incubated for 5 days by MTT assay2021Bioorganic & medicinal chemistry, 06-15, Volume: 40Design, synthesis and anti-HIV evaluation of novel 5-substituted diarylpyrimidine derivatives as potent HIV-1 NNRTIs.
AID1819216Antiviral activity against HIV-1 harboring reverse transcriptase Y181C mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay
AID665271Antiviral activity against HIV1 3B harboring wild type RT infected in MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2012European journal of medicinal chemistry, Jul, Volume: 53Chiral resolution, absolute configuration assignment and biological activity of racemic diarylpyrimidine CH(OH)-DAPY as potent nonnucleoside HIV-1 reverse transcriptase inhibitors.
AID1565093Selectivity index, ratio of CC50 for human MT4 cells to EC50 for antiviral activity against HIV IIIB
AID276231Antiviral activity against HIV1 3B in MT2 cells2006Bioorganic & medicinal chemistry letters, Nov-01, Volume: 16, Issue:21
Optimization of pyrimidinyl- and triazinyl-amines as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1504703Antiviral activity against HIV1 3B harboring reverse transcriptase F227L/V106A double mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2017ACS medicinal chemistry letters, Nov-09, Volume: 8, Issue:11
Discovery of Thiophene[3,2-
AID1487264Inhibition of recombinant wild type HIV1 reverse transcriptase assessed as reduction in biotin-dUTP incorporation at 50 uM using poly(A)/oligo(dT)15 as template/primer by ELISA relative to control2017Bioorganic & medicinal chemistry, 08-15, Volume: 25, Issue:16
Discovery of novel DAPY-IAS hybrid derivatives as potential HIV-1 inhibitors using molecular hybridization based on crystallographic overlays.
AID1636148Ratio of IC50 for NNRTI resistant HIV1 harboring reverse transcriptase A98G/K101E/Y181C/G190A mutant infected in human TZM-bl cells to IC50 for wild type HIV1 NL4-3 infected in human TZM-bl cells2016Journal of medicinal chemistry, Aug-11, Volume: 59, Issue:15
Discovery and Structure-Based Optimization of 2-Ureidothiophene-3-carboxylic Acids as Dual Bacterial RNA Polymerase and Viral Reverse Transcriptase Inhibitors.
AID1457057Antiviral activity against HIV1 harboring reverse transcriptase Y188L mutant infected in human ML4 cells assessed as protection against virus-induced cytopathic effect measured on day 5 post infection by MTT assay2017Journal of medicinal chemistry, 08-10, Volume: 60, Issue:15
Chiral Indolylarylsulfone Non-Nucleoside Reverse Transcriptase Inhibitors as New Potent and Broad Spectrum Anti-HIV-1 Agents.
AID648421Cytotoxicity against human MT4 cells after 5 days by MTT assay2012European journal of medicinal chemistry, May, Volume: 51Synthesis and biological evaluation of piperidine-substituted triazine derivatives as HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID586896Induction of ABCG2 mRNA expression in human LS180 cells at 1 umol/liter after 4 days by RT-PCR analysis relative to beta-glucuronidase gene expression2011Antimicrobial agents and chemotherapy, Mar, Volume: 55, Issue:3
Interaction potential of etravirine with drug transporters assessed in vitro.
AID1292013Selectivity index, ratio of CC50 for mock infected human MT4 cells to EC50 for HIV1 RES056 harboring reverse transcriptase K103N/YI81C double mutant infected in human MT4 cells2016European journal of medicinal chemistry, Jun-10, Volume: 115Design, synthesis and evaluation of novel HIV-1 NNRTIs with dual structural conformations targeting the entrance channel of the NNRTI binding pocket.
AID1504698Antiviral activity against HIV1 3B harboring reverse transcriptase L100I mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2017ACS medicinal chemistry letters, Nov-09, Volume: 8, Issue:11
Discovery of Thiophene[3,2-
AID347632Antiviral activity against NNRTI-resistant HIV HXB2 isolate 31 with reverse transcriptase A98, V179, Y181 mutation in human MT4 cells assessed as protection against virus-induced cell death after 5 days by MTT assay2008Journal of medicinal chemistry, Dec-11, Volume: 51, Issue:23
Design of annulated pyrazoles as inhibitors of HIV-1 reverse transcriptase.
AID1705195Inhibition of RNA-dependent DNA polymerase activity of recombinant HIV-1 p66/p51 reverse transcriptase K103N mutant assessed as inhibition of [3H]dTTP incorporation using poly(rA)/oligo(dT) as templates incubated for 15 mins by MicroBeta scintillation cou2020European journal of medicinal chemistry, Dec-15, Volume: 208New indolylarylsulfone non-nucleoside reverse transcriptase inhibitors show low nanomolar inhibition of single and double HIV-1 mutant strains.
AID1410484Selectivity index, ratio of CC50 for human MT4 cells to EC50 for wild-type HIV1 3B reverse transcriptase infected in human MT4 cells2018ACS medicinal chemistry letters, Apr-12, Volume: 9, Issue:4
Further Exploring Solvent-Exposed Tolerant Regions of Allosteric Binding Pocket for Novel HIV-1 NNRTIs Discovery.
AID1163230Antitrypanocidal activity against suramin-sensitive Trypanosoma brucei brucei Squib427 assessed as reduction in parasite growth after 72 hrs2014Bioorganic & medicinal chemistry, Oct-01, Volume: 22, Issue:19
From human immunodeficiency virus non-nucleoside reverse transcriptase inhibitors to potent and selective antitrypanosomal compounds.
AID1248223Cytotoxicity against uninfected human MT4 cells assessed as decrease in cell viability after 5 days by MTT assay2015Bioorganic & medicinal chemistry, Oct-15, Volume: 23, Issue:20
A novel family of diarylpyrimidines (DAPYs) featuring a diatomic linker: Design, synthesis and anti-HIV activities.
AID1754649Water solubility of compound at pH 22021Bioorganic & medicinal chemistry, 07-15, Volume: 42Exploiting the hydrophobic channel of the NNIBP: Discovery of novel diarylpyrimidines as HIV-1 NNRTIs against wild-type and K103N mutant viruses.
AID619637Antiviral activity against HIV-1 3B harboring RT Y181C mutant infected in human MT4 cells assessed as inhibition of virus-induced syncytium formation after 5 days by MTT assay2011European journal of medicinal chemistry, Oct, Volume: 46, Issue:10
Arylazolylthioacetanilide. Part 8: Design, synthesis and biological evaluation of novel 2-(2-(2,4-dichlorophenyl)-2H-1,2,4-triazol-3-ylthio)-N-arylacetamides as potent HIV-1 inhibitors.
AID508779Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase V179D mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID464764Antiviral activity against wild type HIV1 infected in human MT2 cells2010Bioorganic & medicinal chemistry letters, Mar-01, Volume: 20, Issue:5
N1-Heterocyclic pyrimidinediones as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
AID1072797Inhibition of HIV1 reverse transcriptase assessed as inhibition of biotinylated dUTP incorporation after 1 hr by ELISA2014Bioorganic & medicinal chemistry, Mar-15, Volume: 22, Issue:6
Discovery of 2-pyridone derivatives as potent HIV-1 NNRTIs using molecular hybridization based on crystallographic overlays.
AID1572055Selectivity ratio of CC50 for human MT4 cells to EC50 for HIV1 3B infected in human MT4 cells2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1157586Antiviral activity against MDR-resistant HIV1 harboring 41L, 74V, 106A, 215Y mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathogenicity after 4 days by MTT assay2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Synthesis of novel fluoro analogues of MKC442 as microbicides.
AID1171598Inhibition of wild type HIV1 reverse transcriptase Y181I mutant assessed as reduction in enzyme activity2014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID557041Antiviral activity against HIV1 isolate R8 harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as inhibition of viral replication after 72 hrs in presence of 10% FBS2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID449277Cytotoxicity against human erythrocytes assessed as change in cell shape by flow cytometry2009Bioorganic & medicinal chemistry letters, Sep-15, Volume: 19, Issue:18
Addressing the malaria drug resistance challenge using flow cytometry to discover new antimalarials.
AID1561699Antiviral activity against HIV1 expressing RT K103N mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1572073Resistance factor, ratio of EC50 for HIV1 harboring reverse transcriptase E138K mutant to EC50 for wild type HIV12019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID347634Antiviral activity against NNRTI-resistant HIV HXB2 isolate 34 with reverse transcriptase K101, V106, Y181 mutation in human MT4 cells assessed as protection against virus-induced cell death after 5 days by MTT assay2008Journal of medicinal chemistry, Dec-11, Volume: 51, Issue:23
Design of annulated pyrazoles as inhibitors of HIV-1 reverse transcriptase.
AID1163231Antitrypanocidal activity against suramin-sensitive Trypanosoma brucei rhodesiense STIB-900 assessed as reduction in parasite growth after 72 hrs2014Bioorganic & medicinal chemistry, Oct-01, Volume: 22, Issue:19
From human immunodeficiency virus non-nucleoside reverse transcriptase inhibitors to potent and selective antitrypanosomal compounds.
AID1171586Antiviral activity against HIV1 NL4-3 expressing reverse transcriptase L100I mutant infected in human MT4 cells assessed as reduction in virus-induced cell death by MTT assay2014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1743635Resistance index, ratio of EC50 for antiviral activity against HIV1 expressing RT K103N mutant infected in human MT4 cells to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells2020European journal of medicinal chemistry, Nov-15, Volume: 206Targeting dual tolerant regions of binding pocket: Discovery of novel morpholine-substituted diarylpyrimidines as potent HIV-1 NNRTIs with significantly improved water solubility.
AID1446811Antiviral activity against HIV1 RES056 harboring K103N/Y181C double mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect measured at 5 days post infection by MTT assay2017European journal of medicinal chemistry, Apr-21, Volume: 130Discovery of uracil-bearing DAPYs derivatives as novel HIV-1 NNRTIs via crystallographic overlay-based molecular hybridization.
AID508761Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase K103N, F227L mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID508645Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase Y181C, G190S mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1572076Aqueous solubility of the compound at pH at 7 by HPLC analysis2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Identification of Dihydrofuro[3,4- d]pyrimidine Derivatives as Novel HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors with Promising Antiviral Activities and Desirable Physicochemical Properties.
AID1483281Selectivity index, ratio of CC50 for human MT4 cells to EC50 for reverse transcriptase K103N mutant in human MT4 cells infected HIV1 3B2017Journal of medicinal chemistry, 05-25, Volume: 60, Issue:10
Structure-Based Optimization of Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-nucleoside Reverse Transcriptase Inhibitors with Improved Potency against Resistance-Associated Variants.
AID1822303Oral bioavailability in Sprague-Dawley rat at 5 mg/kg administered via gavage measured up to 24 hrs by LCMS analysis2022Journal of medicinal chemistry, 02-10, Volume: 65, Issue:3
Discovery of Novel Pyridine-Dimethyl-Phenyl-DAPY Hybrids by Molecular Fusing of Methyl-Pyrimidine-DAPYs and Difluoro-Pyridinyl-DAPYs: Improving the Druggability toward High Inhibitory Activity, Solubility, Safety, and PK.
AID449274Antimalarial activity against Plasmodium falciparum W2mef schizont form by hoechst 33342-thiazole orange stain based flow cytometry assay2009Bioorganic & medicinal chemistry letters, Sep-15, Volume: 19, Issue:18
Addressing the malaria drug resistance challenge using flow cytometry to discover new antimalarials.
AID1741396Inhibition of reverse transcriptase Y188L mutant in HIV-1 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay2020European journal of medicinal chemistry, Sep-15, Volume: 202Bioisosterism-based design and enantiomeric profiling of chiral hydroxyl-substituted biphenyl-diarylpyrimidine nonnucleoside HIV-1 reverse transcriptase inhibitors.
AID1200847Selectivity index, ratio of CC50 for mock-infected human MT4 cells to EC50 for HIV1 RES056 expressing reverse transcriptase K103N/Y181C double mutant2015European journal of medicinal chemistry, Mar-26, Volume: 93Fused heterocycles bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 4: design, synthesis and biological evaluation of novel imidazo[1,2-a]pyrazines.
AID1705193Resistance index, ratio of EC50 for antiviral activity against HIV1 harboring RT K103N/Y181C double mutant infected in human MT4 cells to EC50 for antiviral activity against HIV1 NL4-3 infected in human MT4 cells2020European journal of medicinal chemistry, Dec-15, Volume: 208New indolylarylsulfone non-nucleoside reverse transcriptase inhibitors show low nanomolar inhibition of single and double HIV-1 mutant strains.
AID1390713Selectivity index, ratio of CC50 for human MT4 cells to EC50 for HIV1 3B expressing wild type reverse transcriptase infected in human MT4 cells2018Bioorganic & medicinal chemistry letters, 05-01, Volume: 28, Issue:8
First discovery of a potential carbonate prodrug of NNRTI drug candidate RDEA427 with submicromolar inhibitory activity against HIV-1 K103N/Y181C double mutant strain.
AID557065Antiviral activity against HIV1 isolate R8 harboring reverse transcriptase M230L mutant relative to drug-sensitive HIV1 CNDO2009Antimicrobial agents and chemotherapy, Jun, Volume: 53, Issue:6
Antiviral activity of MK-4965, a novel nonnucleoside reverse transcriptase inhibitor.
AID1391082Antiviral activity against HIV1 harboring reverse transcriptase K103N mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay2018Bioorganic & medicinal chemistry, 05-01, Volume: 26, Issue:8
The discovery of novel diarylpyri(mi)dine derivatives with high level activity against a wide variety of HIV-1 strains as well as against HIV-2.
AID1705194Inhibition of RNA-dependent DNA polymerase activity of wild type recombinant HIV-1 His-tagged p66/p51 reverse transcriptase assessed as inhibition of [3H]dTTP incorporation using poly(rA)/oligo(dT) as templates incubated for 15 mins by MicroBeta scintilla2020European journal of medicinal chemistry, Dec-15, Volume: 208New indolylarylsulfone non-nucleoside reverse transcriptase inhibitors show low nanomolar inhibition of single and double HIV-1 mutant strains.
AID1171597Inhibition of wild type HIV1 reverse transcriptase V106A mutant assessed as reduction in enzyme activity2014Journal of medicinal chemistry, Dec-11, Volume: 57, Issue:23
Indolylarylsulfones carrying a heterocyclic tail as very potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors.
AID1888698Antiviral activity against HIV1 with RT E138K mutant infected in human MT4 cells cells assessed as protection against viral-induced cytopathogenecity by MTT assay
AID1561713Resistance index, ratio of EC50 for antiviral activity against HIV1 expressing RT E138K mutant infected in human MT4 cells to EC50 for antiviral activity against HIV1 3B infected in human MT4 cells
AID1391087Antiviral activity against HIV1 harboring reverse transcriptase RES056 double mutant infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 5 days by MTT assay2018Bioorganic & medicinal chemistry, 05-01, Volume: 26, Issue:8
The discovery of novel diarylpyri(mi)dine derivatives with high level activity against a wide variety of HIV-1 strains as well as against HIV-2.
AID1357799Resistance factor, ratio of EC50 for HIV1 harboring reverse transcriptase F227L/V106A mutant to EC50 for wild-type HIV-1 3B2018European journal of medicinal chemistry, May-10, Volume: 151Targeting the entrance channel of NNIBP: Discovery of diarylnicotinamide 1,4-disubstituted 1,2,3-triazoles as novel HIV-1 NNRTIs with high potency against wild-type and E138K mutant virus.
AID701095Half life in plasma2012Journal of medicinal chemistry, Apr-26, Volume: 55, Issue:8
Strategies for the design of HIV-1 non-nucleoside reverse transcriptase inhibitors: lessons from the development of seven representative paradigms.
AID508783Antiviral activity against Human immunodeficiency virus 1 HXB2 containing reverse transcriptase E138K mutant relative to Human immunodeficiency virus 1 3B2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID1572528Antiviral activity against HIV1 harboring reverse transcriptase F227L/V106A double mutant infected in human MT4 cells assessed as inhibition of virus-induced cytotoxicity after 5 days by MTT assay
AID1889295Aqueous solubility of the compound2022Journal of medicinal chemistry, 02-10, Volume: 65, Issue:3
Dose Number as a Tool to Guide Lead Optimization for Orally Bioavailable Compounds in Drug Discovery.
AID573475Antiviral activity of wild-type Human immunodeficiency virus 1 isolate 5331 by cell based assay2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID1754644Antiviral activity against HIV1 harboring RT Y188L mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect incubated for 5 days by MTT assay2021Bioorganic & medicinal chemistry, 07-15, Volume: 42Exploiting the hydrophobic channel of the NNIBP: Discovery of novel diarylpyrimidines as HIV-1 NNRTIs against wild-type and K103N mutant viruses.
AID1731743Antiviral activity against HIV-1 3B infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay
AID496618Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase V90I/V35T, E36D, V60I, K122E, D123S, I135V, E138D, S162A, K173T,Q174S, D177E, I178M, V189I, T200A, I202V, Q207E, F214L, V245Q mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID1574378Antiviral activity against HIV-1 harboring reverse transcriptase K103N/Y181C double mutant infected in human MT4 cells assessed as inhibition of virus-induced cytopathic effect after 5 days by MTT assay2019Journal of medicinal chemistry, 01-24, Volume: 62, Issue:2
Effect of α-Methoxy Substitution on the Anti-HIV Activity of Dihydropyrimidin-4(3 H)-ones.
AID508826Antiviral activity against Human immunodeficiency virus 1 subtype B isolate 93BR021 infected in human PBMC cells by cell based assay2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1.
AID573470Inhibition of DNA-dependent DNA polymerase activity of wild-type Human immunodeficiency virus 1 subtype B reverse transcriptase Y181C mutant by filter-based filtration assay2009Antimicrobial agents and chemotherapy, Nov, Volume: 53, Issue:11
Human immunodeficiency virus type 1 recombinant reverse transcriptase enzymes containing the G190A and Y181C resistance mutations remain sensitive to etravirine.
AID1731752Inhibition of reverse transcriptase RES056 mutant in HIV-1 infected in human MT4 cells assessed as reduction in virus-induced cytopathic effect incubated for 5 days by MTT assay
AID1418463Antiviral activity against wild-type HIV1 3B infected in human MT4 cells assessed as protection against virus-induced cytopathic effect after 4 days by MTT assay2018Bioorganic & medicinal chemistry letters, 12-01, Volume: 28, Issue:22
Design and synthesis of a novel series of non-nucleoside HIV-1 inhibitors bearing pyrimidine and N-substituted aromatic piperazine.
AID496623Antiviral activity against HIV-1 subtype CRF02_AG containing reverse transcriptase E138A/G18V, Q23P, V35T, E40D, V60I, S68G, D123E, I135V, S162A, K173T, Q174N, T200A, Q207E, R211K, F214L mutant relative to control2010Antimicrobial agents and chemotherapy, Feb, Volume: 54, Issue:2
Resistance-associated mutations to etravirine (TMC-125) in antiretroviral-naïve patients infected with non-B HIV-1 subtypes.
AID1801807HIV-1 RT Inhibition Assay from Article 10.1111/cbdd.12751: \\Arylazolyl(azinyl)thioacetanilides: Part 19: Discovery of Novel Substituted Imidazo[4,5-b]pyridin-2-ylthioacetanilides as Potent HIV NNRTIs Via a Structure-based Bioisosterism Approach.\\2016Chemical biology & drug design, 08, Volume: 88, Issue:2
Arylazolyl(azinyl)thioacetanilides: Part 19: Discovery of Novel Substituted Imidazo[4,5-b]pyridin-2-ylthioacetanilides as Potent HIV NNRTIs Via a Structure-based Bioisosterism Approach.
AID1801345In vitro Anti-HIV RT Assay from Article 10.1111/cbdd.12497: \\Design, Synthesis, and Biological Evaluation of Novel Benzoyl Diarylamine/ether Derivatives as Potential Anti-HIV-1 Agents.\\2015Chemical biology & drug design, Sep, Volume: 86, Issue:3
Design, Synthesis, and Biological Evaluation of Novel Benzoyl Diarylamine/ether Derivatives as Potential Anti-HIV-1 Agents.
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 (543)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's0 (0.00)18.2507
2000's144 (26.52)29.6817
2010's342 (62.98)24.3611
2020's57 (10.50)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 48.12

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 strong demand-to-supply ratio for research on this compound.

MetricThis Compound (vs All)
Research Demand Index48.12 (24.57)
Research Supply Index6.50 (2.92)
Research Growth Index4.63 (4.65)
Search Engine Demand Index78.29 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (48.12)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials90 (15.73%)5.53%
Reviews63 (11.01%)6.00%
Case Studies27 (4.72%)4.05%
Observational10 (1.75%)0.25%
Other382 (66.78%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (60)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
An Open-Label Trial With TMC125 in HIV-1 Infected Subjects, Who Were Randomized to a TMC125 Treatment Arm in a Sponsor-Selected TMC125 Trial and Were Treated for at Least 48 Weeks [NCT00128830]Phase 2211 participants (Actual)Interventional2005-06-30Completed
An Open-label Trial With TMC125 as Part of an ART Including TMC114/Rtv and an Investigator-selected OBR in HIV-1 Infected Subjects Who Participated in a DUET Phase III Trial (TMC125-C206 or TMC125-C216). [NCT00359021]Phase 3503 participants (Actual)Interventional2006-06-30Completed
A Randomised, Open Label, Prospective Study to Assess Two Different Therapeutic Strategies Following First Treatment Failure in HIV-1 Infected Subjects [NCT01118871]Phase 43 participants (Actual)Interventional2010-05-31Terminated
Open-Label, Single-Sequence Study to Evaluate the Pharmacokinetic Interaction of BMS-663068 With Darunavir/Ritonavir and/or Etravirine in Healthy Subjects [NCT02063360]Phase 142 participants (Actual)Interventional2014-02-01Completed
A Phase III Randomized, Double-blinded, Placebo-controlled Trial to Investigate the Efficacy, Tolerability and Safety of TMC125 as Part of an ART Regimen, Including TMC114/RTV and an Investigator-selected OBR, in HIV-1 Infected Patients With Limited Treat [NCT00255099]Phase 3593 participants (Actual)Interventional2005-12-31Completed
Continued Access to Etravirine (ETR) in Treatment Experienced HIV-1 Infected Subjects [NCT00980538]Phase 3180 participants (Actual)Interventional2009-12-09Active, not recruiting
Pharmacokinetic Properties of Antiretroviral and Related Drugs During Pregnancy and Postpartum [NCT00042289]1,578 participants (Actual)Observational2003-06-09Completed
Plasma and Intracellular Concentrations of Raltegravir and Etravirine Administered Once Daily (800 mg and 400 mg, Respectively) Compared With Standard Dosing (400 mg and 200 mg/12 h) in Patients With HIV Infection [NCT01121809]Phase 416 participants (Actual)Interventional2010-04-30Completed
A Phase I, Open-label Trial to Investigate the Pharmacokinetic Effect of Multiple-dose TMC125 on Buprenorphine and Norbuprenorphine Administered in HIV-negative Subjects on Stable Buprenorphine/Naloxone Maintenance Therapy. [NCT00828815]Phase 122 participants (Actual)Interventional2009-03-31Completed
A Phase 1, Open-label, Fixed-sequence, 2-Panel Study to Assess the Effects of Itraconazole and Etravirine on the Single-dose Pharmacokinetics of JNJ-64417184 in Healthy Adult Subjects [NCT04208373]Phase 132 participants (Actual)Interventional2019-12-20Completed
A Phase I, Open-label Trial to Investigate the Pharmacokinetic Interaction Between TMC125 and Two Antifungal Agents (Fluconazole and Voriconazole), All at Steady-state in Healthy Subjects. [NCT00740389]Phase 118 participants (Actual)Interventional2008-09-30Completed
Antiretroviral Activity and Tolerability of Once Daily Etravirine in Treatment-Naïve Adults With HIV-1 Infection [NCT00959894]Phase 280 participants (Actual)Interventional2009-09-30Completed
A Phase I, Open Label Crossover Study to Evaluate the Effect of Etravirine on GSK1265744 Pharmacokinetics in Healthy Adult Subjects [NCT00920296]Phase 112 participants (Actual)Interventional2009-07-31Completed
Evaluation of the Capacity of a Weekly Strategy of 4 Consecutive Days on Treatment Followed by 3 Days Off Treatment, in HIV-1 Infected Patients With Undetectable Viral Load for at Least 12 Months, to Maintain a Virological Success With This Intermittent M [NCT02157311]Phase 3100 participants (Actual)Interventional2014-07-31Completed
IMPAACT P1058A: Intensive Pharmacokinetic Studies of New Classes of Antiretroviral Drug Combinations in Children, Adolescents and Young Adults [NCT00977756]168 participants (Actual)Observational2002-08-31Completed
A Phase I, Open-label, Randomized Cross-over, 2-period, 2-way Interaction Trial to Investigate the Pharmacokinetic Interaction Between Lopinavir/Ritonavir and TMC125 Both at Steady-state in Healthy Subjects. [NCT00767117]Phase 116 participants (Actual)Interventional2008-09-30Completed
A Prospective Longitudinal Pilot Study to Measure the Effect of Intensification With Raltegravir +/- a Protease Inhibitor (PI) or Non-Nucleoside Reverse Transcriptase Inhibitor (NNRTI) on HIV-1 Levels in the Gut [NCT00884793]8 participants (Actual)Interventional2008-09-30Completed
TMC125-TiDP2-C238: A Randomized, Exploratory, Open-label 48-week Trial to Investigate the Pharmacokinetics, Safety, Tolerability and Antiviral Activity of Etravirine (ETR) in Combination With Ritonavir-boosted Atazanavir (ATV/Rtv) and 1 NRTI in Treatment- [NCT00896051]Phase 250 participants (Actual)Interventional2009-08-31Completed
Open Label Phase 3b, 48 wk Pilot Study of the Antiviral Efficacy and Tolerability of Combination of PREZISTA/r and TMC125 When Substituted for Enfuvirtide, Current Protease Inhibitor(s) and NNRTI(s) in Antiretroviral Resistant Patients With Viral Suppress [NCT00460746]Phase 310 participants (Actual)Interventional2007-05-31Completed
A Phase 2b Multicenter, Randomized, Comparative Trial Of Uk-453,061 Versus Etravirine In Combination With Darunavir/Ritonavir And A Nucleotide/Nucleoside Reverse Transcriptase Inhibitor For The Treatment Of Antiretroviral Experienced Hiv-1 Infected Subjec [NCT00823979]Phase 2105 participants (Actual)Interventional2009-03-25Terminated(stopped due to See termination reason in detailed description.)
A Single Arm, Open Label Study to Assess the Pharmacokinetics of Darunavir and Ritonavir, Darunavir and Cobicistat, Etravirine, and Rilpivirine in HIV-1 Infected Pregnant Women [NCT00855335]Phase 377 participants (Actual)Interventional2009-04-09Completed
A Multicenter, Single Arm, Open-Label Study of the Once Daily Combination of Etravirine and Darunavir/Ritonavir As Dual Therapy in Early Treatment-Experienced Patients [NCT01199939]Phase 254 participants (Actual)Interventional2010-05-31Completed
A Multicenter, Randomized, Open, Comparative Trial Of Maraviroc Versus Etravirine Each In Combination With Darunavir/Ritonavir And Raltegravir For The Treatment Of Antiretroviral-Experienced HIV-1 Subjects Co-Infected With Hepatitis C And/Or Hepatitis B [NCT00782301]Phase 40 participants (Actual)Interventional2009-03-31Withdrawn
A Phase II, Open-label Trial, to Evaluate the Safety, Tolerability and Antiviral Activity of TMC125 in Antiretroviral Experienced HIV-1 Infected Children and Adolescents [NCT00665847]Phase 2103 participants (Actual)Interventional2008-11-30Completed
CID 0708 - Sex, Aging and Antiretroviral Pharmacokinetics [NCT00666055]11 participants (Actual)Observational2008-03-31Completed
Pilot Study to Assess the Efficacy and Safety of Switching Protease Inhibitor to Etravirine in HIV-1-infected Subjects With Viremia Suppression [NCT01034917]Phase 343 participants (Actual)Interventional2009-12-31Completed
Clinical Trial to Evaluate Drug-drug Interactions Between Darunavir/Cobicistat and Etravirine in Hiv- Infected Patients [NCT02818348]Phase 130 participants (Actual)Interventional2016-06-30Completed
A Phase IV 48 Week, Open Label, Pilot Study of Kaletra and Intelence Tablets in Naive Subjects [NCT01045369]Phase 430 participants (Anticipated)Interventional2010-01-31Completed
A Phase I, Open Label, Randomized, Three Period, One-way, Two Cohort, Adaptive Crossover Study to Evaluate the Effect of Darunavir/Ritonavir Plus Etravirine and Lopinavir/Ritonavir Plus Etravirine on GSK1349572 Pharmacokinetics in Healthy Adult Subjects ( [NCT00867152]Phase 117 participants (Actual)Interventional2009-04-30Completed
Effects of Etravirine (INTELENCETM) on Endothelial Function in HIV-uninfected Adults: A Pilot Study [NCT00871234]Phase 128 participants (Actual)Interventional2009-04-30Completed
A Phase III, Double Blind, Mulit-centre, Randomised Placebo Controlled, Pilot Study to Assess the Feasibility of Switching Individuals Receiving Efavirez With Continuing Central Nervous System (CNS) Toxicity to TMC125. [NCT00792324]Phase 324 participants (Actual)Interventional2008-06-30Completed
The Optimized Treatment That Includes or Omits NRTIs Trial: A Randomized Strategy Study for HIV-1-Infected Treatment-Experienced Subjects Using the cPSS to Select an Effective Regimen [NCT00537394]Phase 3517 participants (Actual)Interventional2008-01-31Completed
A Multicenter Study to Evaluate the Pharmacokinetic Profile and Safety of TMC125 Plus Tenofovir DF/Emtricitabine All Dosed Once Daily With and Without Darunavir (PREZISTA™)/ Ritonavir Once Daily in Antiretroviral naïve HIV-1 Infected Subjects [NCT00534352]Phase 223 participants (Actual)Interventional2008-01-31Completed
Patient Preference, Sleep Quality, and Anxiety/Depression: A Randomized Comparison of Etravirine and Efavirenz [NCT00792584]50 participants (Anticipated)Interventional2008-11-30Completed
A Phase 2 Clinical Trial to Test the Safety and Efficacy of Etravirine in Friedreich Ataxia Patients [NCT04273165]Phase 230 participants (Actual)Interventional2020-09-17Completed
A Study to Survey the Swallowability of Uncoated 200-mg Tablets of Etravirine in HIV-1 Infected Subjects [NCT01090648]Phase 149 participants (Actual)Interventional2010-03-31Completed
A Phase I, Partially Randomized, Open Label, Two-way, Two Period Cross-over Study to Investigate the Pharmacokinetic Interaction Between Etravirine or Darunavir/Rtv and Artemether/Lumefantrine at Steady-state in Healthy HIV-negative Subjects [NCT01876966]Phase 133 participants (Actual)Interventional2011-03-31Completed
A Randomized, Placebo-controlled Phase II Trial in HIV-1-infected, NRTI-, PI and NNRTI-experienced Subjects to Evaluate the Safety, Tolerability and Efficacy of Different Doses of TMC125 b.i.d. on Top of an Individually Optimized Antiretroviral Therapy by [NCT00412646]Phase 2260 participants (Actual)Interventional2002-06-30Completed
A Phase 2b Study to Select a Once Daily Oral Dose of GSK2248761 in HIV-1 Infected Antiretroviral Therapy Experienced Adults With Non-nucleoside Reverse Transcriptase Inhibitor (NNRTI) Resistance [NCT01199731]Phase 230 participants (Actual)Interventional2010-10-05Terminated(stopped due to The study was stopped due to safety concerns)
A Phase IV, Open Label Study in Healthy Male Subjects to Investigate the Extent of Darunavir/Ritonavir and Etravirine Exposure in Blood, Seminal Fluid, and Rectal Mucosal Tissue Following Single and Multiple Dosing of Darunavir/Ritonavir and Etravirine [NCT00855088]Phase 113 participants (Actual)Interventional2009-07-31Completed
A Phase IIb, Multi-centre, Randomised, Double-blind, Active-controlled Trial Comparing the Neuropsychiatric Adverse Event Profile of Etravirine 400mg qd Versus Efavirenz 600mg qd in Combination With 2 Nucleoside/Nucleotide Reverse Transcriptase Inhibitors [NCT00903682]Phase 2157 participants (Actual)Interventional2009-06-30Completed
A Long Term Open-Label Extension Study Of Lersivirine For The Treatment Of HIV-1 Infection [NCT01254656]Phase 2108 participants (Actual)Interventional2011-02-28Terminated(stopped due to See termination reason in detailed description.)
An Open-Label Study to Evaluate the Safety, Tolerability and Pharmacokinetics of Etravirine (ETR) in Combination With Other Antiretrovirals (ARVs) in Antiretroviral Treatment-Experienced HIV-1 Infected Subjects [NCT01422330]Phase 4211 participants (Actual)Interventional2011-09-30Completed
Evaluation of the Pharmacokinetics and Safety of TMC125 Administered Once and Twice-Daily and Following a 2-Week-Treatment Period With Efavirenz in Male and Female Healthy Volunteers. [NCT00531323]Phase 124 participants (Anticipated)Interventional2007-09-30Completed
Open-Label, Single-Sequence Study to Evaluate the Effects of Darunavir/Ritonavir and/or Etravirine on the Pharmacokinetics of GSK3640254 and the Effects of GSK3640254 on the Pharmacokinetics of Darunavir/Ritonavir and/or Etravirine in Heathy Adults [NCT04630002]Phase 154 participants (Actual)Interventional2020-10-28Completed
Antiretroviral Drug Concentrations and HIV Viral Load in Breast Milk and Plasma in HIV+ Women Receiving HAART (Highly Active Antiretroviral Therapy) Therapy: Etravirine (ETR) Pharmacokinetics (PK) in Breast Milk and Plasma [NCT01625169]9 participants (Actual)Interventional2010-04-30Completed
A Phase II, Randomized, Active Controlled, Open Label Trial to Investigate the Efficacy and Tolerability of TMC125 in HIV-1 Infected Subjects, Who Are PI-na�ve and With Documented Genotypic Evidence of NNRTI Resistance From Previous NNRTI Use. [NCT00225303]Phase 2116 participants (Actual)Interventional2005-03-31Completed
A Phase III Randomized, Double-blinded, Placebo-controlled Trial to Investigate the Efficacy, Tolerability and Safety of TMC125 as Part of an ART Regimen, Including TMC114/RTV and an Investigator-selected OBR, in HIV-1 Infected Patients With Limited Treat [NCT00254046]Phase 3616 participants (Actual)Interventional2005-11-30Completed
Prospective Clinical Trial to Assess Safety and Efficacy of DRV/r(TMC 114/r), ETV(TMC 125) and MK-0518 in Addition to OBT in HIV-1 Infected Patients With Limited to No Treatment Options ANRS 139 TRIO [NCT00460382]Phase 2103 participants (Actual)Interventional2007-05-31Completed
HIV-1 Infected Subjects Who Were Randomized in Any Sponsor-selected TMC125 Trial to an Active Control Arm and Either Virologically Failed or Completed the Entire Treatment Period, or to Placebo Arm and Were Treated for at Least 48 Weeks. [NCT00111280]Phase 248 participants (Actual)Interventional2004-09-30Completed
A Randomized, Controlled, Partially Blinded Phase IIb Dose-Finding Trial of TMC125 in HIV-1 Infected Subjects With Documented Genotypic Evidence of Resistance to Currently Available NNRTIs and With at Least Three Primary PI Mutations [NCT00081978]Phase 2211 participants (Actual)Interventional2004-03-31Completed
Early Access of TMC125 in Combination With Other Antiretrovirals in Treatment-experienced HIV-1 Infected Subjects With Limited Treatment Options [NCT00354627]Phase 35,178 participants (Actual)Interventional2006-01-31Completed
CID 0821 - Pilot Study to Evaluate HIV Viremia and Persistence in Acutely HIV-Infected Antiretroviral Naïve Patients Treated With Darunavir/Ritonavir and Etravirine [NCT00855413]Phase 415 participants (Actual)Interventional2009-03-31Terminated(stopped due to Study halted by sponsor due to slow enrollment.)
A Bioequivalence Study to Evaluate the Potential for Drug-drug Interactions Between Boceprevir 800mg Thrice Daily and the HIV Non-nucleoside Reverse Transcriptase Inhibitor Etravirine 200mg Twice Daily in HIV/(Hepatitis C Virus) (HCV) Negative Volunteers [NCT01427504]26 participants (Actual)Interventional2011-08-31Completed
A Phase I/II, Open-Label Trial to Evaluate the Safety, Tolerability, Pharmacokinetics and Antiviral Activity of Etravirine (ETR) in Antiretroviral (ARV) Treatment-Experienced HIV-1 Infected Infants and Children, Aged ≥ 2 Months to < 6 Years [NCT01504841]Phase 1/Phase 226 participants (Actual)Interventional2013-03-14Completed
PREZISTA or INTELENCE Switch Evaluation in Virologically Suppressed Patients Naïve to Darunavir or Etravirine and Who Are Intolerant of Their Current or Prior Combination Antiretroviral Therapy Regimen: A Phase IV, Open-label, Multicentre Observational Tr [NCT01615601]77 participants (Actual)Observational2011-10-31Completed
Management Using the Latest Technologies in Resource-limited Settings to Optimize Combination Therapy After Viral Failure (MULTI-OCTAVE) [NCT01641367]Phase 4545 participants (Actual)Interventional2013-02-22Completed
HIV Neurocognitive Disorders: A Randomized Clinical Trial of CNS-Targeted HAART [NCT00624195]Phase 2/Phase 359 participants (Actual)Interventional2007-03-31Completed
Randomized Clinical Trial to Evaluate the Interest of a Down-scaled Treatment Strategy Using Dual Therapy (Nucleoside Analogs) in HIV Infected Patients Already Being Treated Using Triple Therapy, Who Present With a Successful Virological Control and for W [NCT02302547]Phase 3224 participants (Actual)Interventional2014-12-31Completed
Dual Therapy Combining Raltegravir With Etravirine Maintains a High Level of Viral Suppression Over 96 Weeks in Long-term Experienced HIV-infected Individuals Over 45 Years on a PI-based Regimen: Results From the Phase II ANRS 163 ETRAL Study [NCT02212379]Phase 2170 participants (Actual)Interventional2015-01-31Completed
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00042289 (26) [back to overview]PK Parameter: Maximum Concentration (Cmax) in mg/L With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]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: Trough Concentration (C24) 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 (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]Pharmacokinetic (PK) Parameter: Infant Plasma Washout Half-life (T1/2) of ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Maximum Concentration (Cmax) in ng/mL With Median (95% CI) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Maximum Concentration (Cmax) in ng/mL With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: 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 (C12) With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Trough Concentration (C24) With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Trough Concentration (C24) With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]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]Area Under the Curve From 0 to 24 Hours (AUC24) of ARVs for Contraceptive Arms
NCT00042289 (26) [back to overview]Number of Women Who Met PK Target of Area Under the Curve (AUC) for ARVs
NCT00042289 (26) [back to overview]Number of Women Who Met PK Target of Area Under the Curve (AUC) for ARVs
NCT00042289 (26) [back to overview]Pharmacokinetic (PK) Parameter: Infant Plasma Washout Concentration of ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Area Under the Curve From 0 to 12 Hours (AUC12) With Geometric Mean (95% CI) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Area Under the Curve From 0 to 12 Hours (AUC12) With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Area Under the Curve From 0 to 12 Hours (AUC12) With Median (Range) for ARVs and TB Drugs
NCT00128830 (10) [back to overview]Median Change From TMC125-C229 Basline in Cluster of Differentiation 4 (CD4+) Cell Count at Week 48
NCT00128830 (10) [back to overview]Median Change in Cluster of Differentiation 4 (CD4+) Cell Count From Baseline in TMC125-C229 Feeder Study at Week 192
NCT00128830 (10) [back to overview]Median Change in Cluster of Differentiation 4 (CD4+) Cell Count From Baseline in TMC125-C229 Feeder Study at Week 96
NCT00128830 (10) [back to overview]Number of Participants Who Achieved Virologic Response (ie, Viral Load Less Than 50 Copies/mL) at Week 48
NCT00128830 (10) [back to overview]Number of Participants Who Achieved Virologic Response (ie, Viral Load Less Than 50 Copies/mL) at Week 96
NCT00128830 (10) [back to overview]Number of Participants With Adverse Events
NCT00128830 (10) [back to overview]Number of Participants Who Achieved Virologic Response (ie, Viral Load Less Than 50 Copies/mL; Viral Load Less Than 400 Copies/mL; and Greater Than or Equal to 1 log10 Decrease From Baseline) at Week 192
NCT00128830 (10) [back to overview]Number of Participants With Emerging Mutation (Reverse Transcriptase Mutation)
NCT00128830 (10) [back to overview]Median Change From TMC125-C229 Baseline in Cluster of Differentiation 4 (CD4+) Cell Count at Week 96
NCT00128830 (10) [back to overview]Number of Participants Who Achieved Virologic Response (ie, Viral Load Less Than 50 Copies/mL; Less Than 400 Copies/mL; and Greater Than or Equal to 1 Log 10 Decrease From Baseline) at Week 96
NCT00359021 (3) [back to overview]Change in Plasma Viral Load Versus Baseline (ie, Mean Change in log10 Plasma Viral Load From Baseline Over Time)
NCT00359021 (3) [back to overview]The Number of Participants Experiencing Adverse Events
NCT00359021 (3) [back to overview]The Percentage of Participants With Virologic Outcomes Over Time
NCT00460746 (10) [back to overview]Median Change From Baseline in Total Cholesterol at Week 48.
NCT00460746 (10) [back to overview]Median Change From Baseline in Triglycerides at Week 48.
NCT00460746 (10) [back to overview]Proportion of Patients Who Have Viral Load Measurements <50 Copies/ml at 2, 4, 8, 12, 16, 24, 36 and 48 Weeks After Switching to DRV/r and ETR, Missing Equals Failure.
NCT00460746 (10) [back to overview]Proportion of Patients Who Maintain Plasma HIV Viral Load Measurements < 400 Copies/ml at 2, 4, 8, 12, 16, 24, 36 and 48 Weeks After Switching to DRV/r and ETR, Missing Equals Failure.
NCT00460746 (10) [back to overview]CD4+ Cell Count (x 10^6 Cell/L): Baseline and Mean Changes From Baseline at 4, 8, 12, 16, 24,36 and 48 Weeks.
NCT00460746 (10) [back to overview]CD4+ Cell Count (x 10^6 Cell/L): Baseline and Median Changes From Baseline at 4, 8, 12, 16, 24, 36 and 48 Weeks.
NCT00460746 (10) [back to overview]Median Change From Baseline in Glucose at Week 48.
NCT00460746 (10) [back to overview]Median Change From Baseline in HDL Cholesterol.
NCT00460746 (10) [back to overview]Median Change From Baseline in LDL Cholesterol at Week 48.
NCT00460746 (10) [back to overview]Median Change From Baseline in Total Cholesterol (TC) / High Denisty Lipoprotein (HDL) Ratio at Week 48.
NCT00534352 (12) [back to overview]Virologic Response < 50 HIV-1 RNA Copies/mL (ITT-Observed Case)
NCT00534352 (12) [back to overview]Number of Participants With Treatment-Emergent Non-Graded Laboratory Abnormalities(Worst Abnormality): Lipids- High-density Lipoprotein (HDL)
NCT00534352 (12) [back to overview]Number of Participants With Treatment-Emergent Non-Graded Laboratory Abnormalities(Worst Abnormality): Glucose- Insulin
NCT00534352 (12) [back to overview]Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities(Worst Grade): Lipids- Triglycerides
NCT00534352 (12) [back to overview]Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities(Worst Grade): Lipids- Low-density Lipoprotein (LDL) Direct
NCT00534352 (12) [back to overview]Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities (Worst Grade): Lipids- Total Cholesteral
NCT00534352 (12) [back to overview]Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities (Worst Grade): Glucose-Hyperglycemia
NCT00534352 (12) [back to overview]Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities (Worst Grade): Glucose- Hypoglycemia
NCT00534352 (12) [back to overview]Log10 Viral Load (HIV-1 RNA Copies/mL): Mean Changes From Baseline(ITT-Observed Case)
NCT00534352 (12) [back to overview]CD4+ Cell Count (x 10^6 Cell/L): Baseline and Median Changes From Baseline (ITT-Observed Case)
NCT00534352 (12) [back to overview]CD4+ Cell Count (Percent): Baseline and Median Changes From Baseline (ITT-Observed Case)
NCT00534352 (12) [back to overview]Number of Participants Contributing to the Pharmacokinetic (PK) Evaluations: Cmin, Cmax, AUC24 & Css,av
NCT00537394 (15) [back to overview]Change in Cardiovascular Risk Score From Baseline
NCT00537394 (15) [back to overview]Change in CD4 Count From Baseline
NCT00537394 (15) [back to overview]Change in Fasting Non-HDL Cholesterol From Baseline
NCT00537394 (15) [back to overview]Change in Summarized Quality of Life Score
NCT00537394 (15) [back to overview]Number of Participants Self-reporting Non-adherence to Assigned Study ARVS (Excluding NRTIs, if Applicable)
NCT00537394 (15) [back to overview]Number of Participants With Plasma HIV-1 Viral Load < 50 Copies/ml
NCT00537394 (15) [back to overview]Time From Randomization to Confirmed Virological Failure
NCT00537394 (15) [back to overview]Time From Randomization to Discontinuation of Randomized NRTI Component of Study Treatment
NCT00537394 (15) [back to overview]Time From Treatment Dispensation to First Grade 3 or Higher (and at Least One Grade Higher Than Baseline) Signs/Symptom or Laboratory Abnormality
NCT00537394 (15) [back to overview]Time From Treatment Dispensation to First Study ARV Modification (Excluding NRTIs, if Applicable)
NCT00537394 (15) [back to overview]Time From Treatment Dispensation to Serious Non-AIDS-defining Events
NCT00537394 (15) [back to overview]Change in Plasma HIV-1 Viral Load From Baseline to Week 1
NCT00537394 (15) [back to overview]Number of Participants With Change in Virus Co-receptor Tropism Among Those With R5-only Tropic Virus at Study Entry
NCT00537394 (15) [back to overview]Participants With Newly Acquired HIV Drug Resistance Between Study Entry and Confirmed Virologic Failure
NCT00537394 (15) [back to overview]Percent of Participants With Regimen Failure, Defined as a Confirmed Virologic Failure or Discontinuation of Randomized NRTI Component of Study Treatment
NCT00624195 (1) [back to overview]Neuropsychological Performance Change
NCT00665847 (9) [back to overview]The Change From Baseline in CD4 Cell Counts Over Time
NCT00665847 (9) [back to overview]Population Pharmacokinetic (PK) Estimates of Etravirine/TMC125 (ETR): Trough Plasma Concentration (C0h)
NCT00665847 (9) [back to overview]The Percentage of Patients With Treatment-emergent Adverse Events (TEAEs)
NCT00665847 (9) [back to overview]The Emergence of Non-nucleoside Reverse Transcriptase Inhibitor Resistance-associated Mutations (NNRTI RAMs) in Patients Classified as Virologic Failures
NCT00665847 (9) [back to overview]The Number of Patients With Treatment-emergent Adverse Events (TEAEs)
NCT00665847 (9) [back to overview]Change From Baseline in Human Immunodeficiency Virus - Type 1 (HIV-1) Ribonucleic Acid (RNA) in Plasma Over Time
NCT00665847 (9) [back to overview]Percentage of Patients With Virologic Response at Week 24
NCT00665847 (9) [back to overview]Population Pharmacokinetic (PK) Estimates of Etravirine/TMC125 (ETR): Area Under the Plasma Concentration-time Curve Over 12 Hours at Steady-state (AUC12h)
NCT00665847 (9) [back to overview]Population Pharmacokinetic (PK) Estimates of Etravirine/TMC125 (ETR): Maximum Plasma Concentration (Cmax)
NCT00823979 (10) [back to overview]Percentage of Participants With HIV-1 RNA Levels <50 Copies/mL at Week 48 and 96
NCT00823979 (10) [back to overview]Percentage of Participants With Human Immunodeficiency Virus Type 1 Ribonucleic Acid (HIV-1 RNA) Levels Less Than (<) 50 Copies/Milliliter (mL) at Week 24
NCT00823979 (10) [back to overview]Percentage of Participants With Response as Determined by the Time to Loss of Virologic Response (TLOVR50) Algorithm at Week 24, 48 and 96
NCT00823979 (10) [back to overview]Time-Averaged Difference (TAD) in log10 Transformed HIV-1 RNA Levels at Week 24, 48 and 96
NCT00823979 (10) [back to overview]Percentage of Participants With HIV-1 RNA Levels <400 Copies/mL at Week 24, 48 and 96
NCT00823979 (10) [back to overview]Change From Baseline in Cluster of Differentiation 4 (CD4+) Absolute Lymphocyte Counts at Week 24, 48 and 96
NCT00823979 (10) [back to overview]Change From Baseline in Cluster of Differentiation 4 (CD4+) Percentage Lymphocyte Counts at Week 24, 48, 96
NCT00823979 (10) [back to overview]Change From Baseline in log10 Transformed HIV-1 RNA Levels at Week 24, 48 and 96
NCT00823979 (10) [back to overview]Number of Participants With Laboratory Test Abnormalities
NCT00823979 (10) [back to overview]Number of Participants With Non-nucleoside Reverse Transcriptase Inhibitors (NNRTI) Resistance-Associated Mutations (RAMs) and/or Phenotypic Susceptibility at Time of Treatment Failure Through Week 48
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]Number of Participants With Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA) Plasma Viral Load (<) 50 Copies/Milliliter (mL)
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]Number of Participants With Resistance at Virological Failure
NCT00855335 (15) [back to overview]Minimum Plasma Concentration (Cmin)
NCT00855335 (15) [back to overview]Plasma Concentration of Drug in the Cord Plasma and Maternal Plasma Samples Collected at the Time of Delivery
NCT00855335 (15) [back to overview]Mean Change From Baseline in Log10 Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA) Viral Load Value
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]Mean Change From Baseline in CD4+ Cell Count
NCT00855335 (15) [back to overview]Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT00855413 (38) [back to overview]Minimum Etravirine Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture
NCT00855413 (38) [back to overview]Minimum Darunavir Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture
NCT00855413 (38) [back to overview]Minimum Etravirine Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen
NCT00855413 (38) [back to overview]Minimum Etravirine Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure
NCT00855413 (38) [back to overview]Minimum Ritonavir Exposure Range in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture
NCT00855413 (38) [back to overview]Minimum Ritonavir Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure
NCT00855413 (38) [back to overview]Number of Participants Who Stopped Study Treatment Due to Adverse Event or Intolerance
NCT00855413 (38) [back to overview]Number of Participants With HIV RNA Measurement Above the Limits of Detection in Cerebrospinal Fluid
NCT00855413 (38) [back to overview]Number of Participants With Neurocognitive Impairment at Baseline
NCT00855413 (38) [back to overview]Number of Participants With Neurocognitive Impairment at Week 24
NCT00855413 (38) [back to overview]Number of Participants With Neurocognitive Impairment at Week 48
NCT00855413 (38) [back to overview]Number of Participants With Virologic Response
NCT00855413 (38) [back to overview]Number of Participants With Virologic Response
NCT00855413 (38) [back to overview]Overall Neurocognitive Impairment at Week 24
NCT00855413 (38) [back to overview]Overall Neurocognitive Impairment at Week 48
NCT00855413 (38) [back to overview]Overall Neurocognitive Impairment Score at Week 2 or 4
NCT00855413 (38) [back to overview]Minimum Ritonavir Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen
NCT00855413 (38) [back to overview]Adverse Events Possibly or Definitely Related to Study Treatment Through Week 48
NCT00855413 (38) [back to overview]Change in Overall Neurocognitive Impairment From Baseline to Week 24 or 48
NCT00855413 (38) [back to overview]Correlation of HIV RNA Levels in CSF and Drug Levels With Neurocognitive Functioning
NCT00855413 (38) [back to overview]Correlation of Time to HIV RNA Levels <200 Copies/mL With Improvement in Neurocognitive Functioning From Baseline to Week 24 and 48
NCT00855413 (38) [back to overview]HIV RNA Detection in Ileal Biopsy Specimens
NCT00855413 (38) [back to overview]HIV RNA Detection in Semen
NCT00855413 (38) [back to overview]HIV RNA Levels Immediately Prior to Initiating Study Treatment.
NCT00855413 (38) [back to overview]Maximum Darunavir Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture
NCT00855413 (38) [back to overview]Maximum Darunavir Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen
NCT00855413 (38) [back to overview]Maximum Darunavir Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure
NCT00855413 (38) [back to overview]Maximum Etravirine Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture
NCT00855413 (38) [back to overview]Maximum Etravirine Exposure in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure
NCT00855413 (38) [back to overview]Maximum Etravirine Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen
NCT00855413 (38) [back to overview]Maximum Ritonavir Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture
NCT00855413 (38) [back to overview]Maximum Ritonavir Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen
NCT00855413 (38) [back to overview]Maximum Ritonavir Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure
NCT00855413 (38) [back to overview]Median Change in CD4 Cell Count From Week 0 to Week 24.
NCT00855413 (38) [back to overview]Median Change in CD4 Cell Count From Week 0 to Week 48.
NCT00855413 (38) [back to overview]Median Time to HIV RNA Suppression to <200 Copies/mL
NCT00855413 (38) [back to overview]Minimum Darunavir Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure
NCT00855413 (38) [back to overview]Minimum Darunavir Exposure Range in Semen Among Participants Who Consented to an Optional Collection of Semen
NCT00871234 (1) [back to overview]Flow-mediated Dilation (FMD) of the Brachial Artery
NCT00884793 (4) [back to overview]"Average Change in Activated (CD38+HLADR+) CD8+ T Cells in the Ileum"
NCT00884793 (4) [back to overview]Number of Subjects Who Experienced an Increase in CD4% in the Ileum.
NCT00884793 (4) [back to overview]Number of Subjects Who Experienced an Increase in CD4+ T Cells (as a % of All Cells) in the Ileum.
NCT00884793 (4) [back to overview]Number of Subjects Who Had a Decrease in HIV RNA Per Million CD4+ T Cells in the Ileum
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Atazanavir (ATV): Treatment A: ATV/Low-Dose Ritonavir (Rtv) 300/100 mg (Results for C0h, Cmin, and Cmax)
NCT00896051 (18) [back to overview]Percentage of Participants With Undetectable Plasma Viral Load (VL) Values (<50 Copies/mL) at Week 48
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Atazanavir (ATV): Treatment A: ATV/Low-Dose Ritonavir (Rtv) 300/100 mg (Results for AUC24hr)
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Atazanavir (ATV): Treatment B: ATV/Low-Dose Ritonavir (Rtv) 400/100 mg (Results for AUC24hr)
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Etravirine (ETR) (Results for AUC12hr)
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Low-Dose Ritonavir (Rtv): Treatment A: Atazanavir (ATV)/Rtv 300/100 mg (Results for AUC24hr)
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Low-Dose Ritonavir (Rtv): Treatment B: Atazanavir (ATV)/Rtv 400/100 mg (Results for AUC24hr)
NCT00896051 (18) [back to overview]Change From Pre-Baseline in Log10 Viral Load Over Time
NCT00896051 (18) [back to overview]Change From Prebaseline in CD4+ Cell Count Over Time
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Etravirine (ETR) (Results for C0h, Cmin, and Cmax)
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Atazanavir (ATV): Treatment B: ATV/Low-Dose Ritonavir (Rtv) 400/100 mg (Results for C0h, Cmin, and Cmax)
NCT00896051 (18) [back to overview]Time to Virologic Failure
NCT00896051 (18) [back to overview]Time to Confirmed Virologic Response
NCT00896051 (18) [back to overview]The Percentage of Participants With a Virologic Response Using the Time to Loss of Virologic Response (TLOVR) Imputation Method
NCT00896051 (18) [back to overview]The Percentage of Participants With a Virologic Response Using the Non-Completing = Failure (NC=F) Imputation Method
NCT00896051 (18) [back to overview]The Percentage of Participants With a Virologic Response (Plasma Viral Load < 50 Copies/mL) at Week 48 Using the Snapshot Analysis Method
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Low-Dose Ritonavir (Rtv): Treatment B: Atazanavir (ATV)/Rtv 400/100 mg (Results for C0h, Cmin, and Cmax)
NCT00896051 (18) [back to overview]Pharmacokinetic Results of Low-Dose Ritonavir (Rtv): Treatment A: Atazanavir (ATV)/Rtv 300/100 mg (Results for C0h, Cmin, and Cmax)
NCT00903682 (7) [back to overview]Antiviral Activity of ETR vs. EFV
NCT00903682 (7) [back to overview]Neuropsychiatric Adverse Events by Week 48
NCT00903682 (7) [back to overview]Proportion of Patients With at Least 1 Treatment-emergent Grade 1-4 Central Nervous System or Psychiatric Adverse Event
NCT00903682 (7) [back to overview]Mean Change From Baseline in CD4+ Cell Count
NCT00903682 (7) [back to overview]Mean Change From Baseline in Neuropsychiatric and Total Tolerabililty Score
NCT00903682 (7) [back to overview]Resistance Determinations
NCT00903682 (7) [back to overview]Antiviral Activity of ETR vs. EFV
NCT00959894 (25) [back to overview]Probability of Remaining Free of a Safety/Tolerability Event at 96 Weeks
NCT00959894 (25) [back to overview]Change in Limb and Trunk Fat Distribution as Measured by DEXA Scan, in the Same Subgroup of up to 40 Participants (as in Aim 8), From Baseline to Week 96 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]Change in CD4+ Cell Count From Baseline to Week 24 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]Change in CD4+ Cell Count From Baseline to Week 48 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]Change in CD4+ Cell Count From Baseline to Week 96 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]Change in Fat Mass Ratio as Measured by DEXA Scan, in the Same Subgroup of up to 40 Participants (as in Aim 8), From Baseline to Week 24 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]Change in Fat Mass Ratio as Measured by DEXA Scan, in the Same Subgroup of up to 40 Participants (as in Aim 8), From Baseline to Week 96 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]Change in Glucose Metabolism (Insulin Resistance), in a Subgroup of up to 40 Participants, From Baseline to Week 24 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]Change in Glucose Metabolism (Insulin Resistance), in a Subgroup of up to 40 Participants, From Baseline to Week 48 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]Change in Glucose Metabolism (Insulin Resistance), in a Subgroup of up to 40 Participants, From Baseline to Week 96 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]Pharmacokinetics of Etravirine in Genital Secretions of up to 10 Men and up to 10 Women at Week 4 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]Population Pharmacokinetics of Etravirine 400 mg Once Daily, in Combination With Fixed-dose Emtricitabine-tenofovir Among Treatment-naïve HIV-1 Infected Adults: Etravirine AUC-24 Hours at Steady State
NCT00959894 (25) [back to overview]The Antiretroviral Activity of Etravirine 400 mg Given Once Daily, With Fixed-dose Truvada Once Daily, Among Treatment-naïve HIV-1 Infected Adults as Measured by the Percentage of Participants With HIV RNA < 50 Copies/mL at Week 24
NCT00959894 (25) [back to overview]The Proportion of Participants With HIV RNA <200 Copies/mL at Week 24 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]The Proportion of Participants With HIV RNA <200 Copies/mL at Week 48 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]The Proportion of Participants With HIV RNA <200 Copies/mL at Week 96 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]The Proportion of Participants With HIV RNA <50 Copies/mL at Week 48 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]The Proportion of Participants With HIV RNA <50 Copies/mL at Week 96 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]Change in Limb and Trunk Fat Distribution as Measured by DEXA Scan, in the Same Subgroup of up to 40 Participants (as in Aim 8), From Baseline to Week 24 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]Change in the Lipid Profile and Glucose Metabolism, in a Subgroup of up to 40 Participants, From Baseline to Week 24 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]Change in the Lipid Profile and Glucose Metabolism, in a Subgroup of up to 40 Participants, From Baseline to Week 48 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]Change in the Lipid Profile and Glucose Metabolism, in a Subgroup of up to 40 Participants, From Baseline to Week 96 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine
NCT00959894 (25) [back to overview]Population Pharmacokinetics of Etravirine 400 mg Once Daily, in Combination With Fixed-dose Emtricitabine-tenofovir Among Treatment-naïve HIV-1 Infected Adults
NCT00959894 (25) [back to overview]Resistance Mutations in the Subset of Patients With Confirmed Virologic Failure Who Have HIV RNA >500 Copies/mL and Genotype Resistance Results
NCT00959894 (25) [back to overview]Tolerability of Etravirine in HIV-1 Infected Adults Initiating Antiretroviral Therapy
NCT00980538 (1) [back to overview]Number of Participants With At-least One Adverse Event as a Measure of Safety Until Etravirine (ETR)-Based Treatment Regimen is Commercially Available
NCT01199731 (16) [back to overview]Percentage of Participants With Human Immunodeficiency Virus Type 1 (HIV-1) Ribonucleic Acid (RNA) <50 Copies Per Milliliter (/mL) at Week 16
NCT01199731 (16) [back to overview]Absolute Values of CD4+ Cell Counts After Switch of GSK2248761
NCT01199731 (16) [back to overview]Change From Baseline in CD4+ Cell Counts After Switch of GSK2248761
NCT01199731 (16) [back to overview]Change From Baseline in CD4+ Cell Counts After Switch of GSK2248761
NCT01199731 (16) [back to overview]Change From Baseline in CD4+ Cell Counts Prior to Switch of GSK2248761
NCT01199731 (16) [back to overview]Change From Baseline in CD4+ Cell Counts Prior to Switch of GSK2248761
NCT01199731 (16) [back to overview]Change From Baseline in Plasma HIV-1 RNA After Switch of GSK2248761
NCT01199731 (16) [back to overview]Change From Baseline in Plasma HIV-1 RNA Prior to Switch of GSK2248761
NCT01199731 (16) [back to overview]Change From Baseline in Plasma HIV-1 RNA Prior to Switch of GSK2248761
NCT01199731 (16) [back to overview]Number of Participants Who Experienced Disease Progression (HIV-associated Conditions, AIDS and Death)
NCT01199731 (16) [back to overview]Number of Participants With Electrocardiograph (ECG) With Values of Potential Critical Concern (PCI)
NCT01199731 (16) [back to overview]Number of Participants With Abnormal Clinical Chemistry Laboratory Data With Grade 3 or 4 Treatment-Emergent (TE) Toxicities
NCT01199731 (16) [back to overview]Number of Participants With Abnormal Hematology Laboratory Data With Grade 3 or 4 TE Toxicities
NCT01199731 (16) [back to overview]Number of Participants With Treatment-emergent Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT01199731 (16) [back to overview]Number of Participants Who Discontinued Treatment Due to AEs
NCT01199731 (16) [back to overview]Absolute Values of CD4+ Cell Counts After Switch of GSK2248761
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 48
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 8
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 24
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 20
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 16
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 12
NCT01199939 (14) [back to overview]Change From Baseline in Cluster of Differentiation 4 (CD4+) and Cluster of Differentiation 8 (CD8+) Cell Counts at Week 48
NCT01199939 (14) [back to overview]Time to Reach First Confirmed Virologic Response
NCT01199939 (14) [back to overview]Number of Participants With Virologic Failure
NCT01199939 (14) [back to overview]Number of Participants With Confirmed Virologic Response (CVR) at Week 48
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 36
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 4
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 42
NCT01199939 (14) [back to overview]Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 30
NCT01254656 (7) [back to overview]Number of Participants With Plasma HIV-1 RNA Level <50 Copies/mL up to Week 208
NCT01254656 (7) [back to overview]Number of Participants With Plasma Human Immunodeficiency Virus - 1 (HIV-1) Ribonucleic Acid (RNA) Level <50 Copies/mL at 144 Weeks From Day 1 of the Parent Protocol
NCT01254656 (7) [back to overview]Virology Analysis Participant Accountability From Week 96 Through Study Termination
NCT01254656 (7) [back to overview]Change From Baseline in CD4+ Lymphocyte Counts (Absolute) at 144 Weeks From Day 1 of the Parent Protocol
NCT01254656 (7) [back to overview]Change From Baseline in CD4+ Lymphocyte Counts (Absolute) at 192 Weeks From Day 1 of the Parent Protocol
NCT01254656 (7) [back to overview]Change From Baseline in CD4+ Lymphocyte Counts (Percentage) at 144 Weeks From Day 1 of the Parent Protocol
NCT01254656 (7) [back to overview]Change From Baseline in CD4+ Lymphocyte Counts (Percentage) at 192 Weeks From Day 1 of the Parent Protocol
NCT01427504 (12) [back to overview]Etravirine Cmin Pharmacokinetics Coadministered With Boceprevir
NCT01427504 (12) [back to overview]Boceprevir AUC Pharmacokinetics
NCT01427504 (12) [back to overview]Boceprevir AUC Pharmacokinetics Coadministered With Etravirine
NCT01427504 (12) [back to overview]Boceprevir C8 Pharmacokinetics
NCT01427504 (12) [back to overview]Boceprevir C8 Pharmacokinetics Coadministered With Etravirine
NCT01427504 (12) [back to overview]Boceprevir Cmax Pharmacokinetics
NCT01427504 (12) [back to overview]Boceprevir Cmax Pharmacokinetics Coadministered With Etravirine
NCT01427504 (12) [back to overview]Etravirine AUC Pharmacokinetics
NCT01427504 (12) [back to overview]Etravirine AUC Pharmacokinetics Coadministered With Boceprevir
NCT01427504 (12) [back to overview]Etravirine Cmax Pharmacokinetics
NCT01427504 (12) [back to overview]Etravirine Cmax Pharmacokinetics Coadministered With Boceprevir
NCT01427504 (12) [back to overview]Etravirine Cmin Pharmacokinetics
NCT01504841 (3) [back to overview]Area Under the Plasma Concentration-Time Curve Over 12 Hours of ETR
NCT01504841 (3) [back to overview]Decline in Absolute CD4 Percent of Greater Than 5 Percent Any Time After 12 Weeks of Therapy
NCT01504841 (3) [back to overview]HIV-1 RNA Virologic Failure Status at Weeks 24 and 48
NCT01625169 (10) [back to overview]Peak Concentration of Etravirine in Breast Milk
NCT01625169 (10) [back to overview]HIV Viral Load in Breast Milk and Plasma
NCT01625169 (10) [back to overview]Peak Plasma Concentration of Etravirine in Plasma
NCT01625169 (10) [back to overview]Peak Plasma Concentration of Etravirine in Plasma
NCT01625169 (10) [back to overview]HIV Viral Load in Breast Milk and Plasma
NCT01625169 (10) [back to overview]Area Under the Curve (AUC) 0-12 for Breast Milk
NCT01625169 (10) [back to overview]Area Under the Curve (AUC) 0-12 for Breast Milk
NCT01625169 (10) [back to overview]Area Under the Curve (AUC) 0-12 for Plasma
NCT01625169 (10) [back to overview]Area Under the Curve (AUC) 0-12 for Plasma
NCT01625169 (10) [back to overview]Peak Concentration of Etravirine in Breast Milk
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of Triglycerides [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of Triglycerides
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of Total Cholesterol [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of Total Cholesterol
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of High-density Lipoprotein Cholesterol [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of High-density Lipoprotein Cholesterol
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of Glucose [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of Glucose
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of Calculated Low-density Lipoprotein Cholesterol [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Change From Baseline in Fasting Values of Calculated Low-density Lipoprotein Cholesterol
NCT01641367 (60) [back to overview]Change From Baseline in CD4+ T-cell Count [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Change From Baseline in CD4+ T-cell Count
NCT01641367 (60) [back to overview]Proportion of Participants With Plasma HIV-1 RNA ≤200 Copies/mL at 72 Weeks [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Proportion of Participants With Plasma HIV-1 RNA ≤200 Copies/mL at 72 Weeks
NCT01641367 (60) [back to overview]Proportion of Participants With Plasma HIV-1 RNA ≤200 Copies/mL at 48 Weeks [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Proportion of Participants With Plasma HIV-1 RNA ≤200 Copies/mL at 48 Weeks
NCT01641367 (60) [back to overview]Proportion of Participants With Plasma HIV-1 RNA ≤200 Copies/mL at 24 Weeks [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to the First of Death or Hospitalization [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Percent of Participants With Treatment Modification or Discontinuation Due to Toxicity by Week 48
NCT01641367 (60) [back to overview]Percent of Participants With Treatment Modification or Discontinuation by Week 48 [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Percent of Participants With Treatment Modification or Discontinuation by Week 48
NCT01641367 (60) [back to overview]Percent of Participants With Death or Hospitalization by Week 48 [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Percent of Participants With Death or Hospitalization by Week 48
NCT01641367 (60) [back to overview]Percent of Participants With Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study, With a New Resistance-associated Mutation Detected in Population-based Sequencing, by Week 48 [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Percent of Participants With Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study, With a New Resistance-associated Mutation Detected in Population-based Sequencing, by Week 48
NCT01641367 (60) [back to overview]Percent of Participants With Confirmed Virologic Failure by Week 48 [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Percent of Participants With Confirmed Virologic Failure by Week 48
NCT01641367 (60) [back to overview]Percent of Participants With a Dose Modification Due to Grade 3 or 4 Toxicity by Week 48 [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Percent of Participants With a Dose Modification Due to Grade 3 or 4 Toxicity by Week 48
NCT01641367 (60) [back to overview]Percent of Participants That Developed Immune Reconstitution Inflammatory Syndrome (IRIS) by Week 48 [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Percent of Participants That Developed Immune Reconstitution Inflammatory Syndrome (IRIS) by Week 48
NCT01641367 (60) [back to overview]Percent of Participants Experiencing Death, AIDS-defining Event or a Non-AIDS-defining Event by Week 48 [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Percent of Participants Experiencing Death, AIDS-defining Event or a Non-AIDS-defining Event by Week 48
NCT01641367 (60) [back to overview]Percent of Participants Experiencing Death by Week 48 [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Percent of Participants Experiencing Death by Week 48
NCT01641367 (60) [back to overview]Number of Weeks of Follow-up [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Number of Participants With Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study, With a New Resistance-associated Mutation Detected in Population-based Sequencing [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Number of Weeks of Follow-up
NCT01641367 (60) [back to overview]Time to Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Time to Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to Treatment Modification or Discontinuation.
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to Treatment Modification or Discontinuation Due to Toxicity [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to Treatment Modification or Discontinuation Due to Toxicity
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to Treatment Modification or Discontinuation [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to the First of Death, an AIDS-defining Event or a Non-AIDS-defining Event [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to the First of Death, an AIDS-defining Event or a Non-AIDS-defining Event
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to the First of Death or Hospitalization.
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to the Development of Immune Reconstitution Inflammatory Syndrome (IRIS) [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to the Development of Immune Reconstitution Inflammatory Syndrome (IRIS)
NCT01641367 (60) [back to overview]Proportion of Participants With Plasma HIV-1 RNA ≤200 Copies/mL at 24 Weeks
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to Death [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Time From Study Entry/Randomization to Death
NCT01641367 (60) [back to overview]Percent of Participants With Treatment Modification or Discontinuation Due to Toxicity by Week 48 [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Number of Participants With Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study
NCT01641367 (60) [back to overview]Number of Participants With Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Number of Participants With Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study, With a New Resistance-associated Mutation Detected in Population-based Sequencing
NCT01641367 (60) [back to overview]Time to Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study, With a New Resistance-associated Mutation Detected in Population-based Sequencing
NCT01641367 (60) [back to overview]Time to Confirmed Virologic Failure, Defined as First HIV-1 RNA ≥1000 Copies/mL at or After 24 Weeks on Study, With a New Resistance-associated Mutation Detected in Population-based Sequencing [CPI+SOC v SOC]
NCT01641367 (60) [back to overview]Time to First Dose Modification Due to Grade 3 or 4 Toxicity
NCT01641367 (60) [back to overview]Time to First Dose Modification Due to Grade 3 or 4 Toxicity [CPI+SOC v SOC]
NCT02212379 (24) [back to overview]Percentage of Participants With Detectable Seminal HIV-RNA Viral Load at Week 48
NCT02212379 (24) [back to overview]Median Time of Virological Failure
NCT02212379 (24) [back to overview]Number of Patients With RAL and/or ETR Resistance Mutations Among Those With Virological Failure
NCT02212379 (24) [back to overview]Percent Change of Renal Function
NCT02212379 (24) [back to overview]Evolution of Body Fat Distribution From Day 0 to W96 (DXA Scan Sub-study, 80 Patients)
NCT02212379 (24) [back to overview]Evolution of CD4+, CD8+ T Cells Counts and CD4/CD8 Ratio
NCT02212379 (24) [back to overview]Evolution of Metabolic Parameters (Fasting Triglycerides, Total Cholesterol, HDL-cholesterol, LDL-cholesterol and Fasting Glycemia)
NCT02212379 (24) [back to overview]Evolution of the Calibrated 5-year Framingham Risk Score
NCT02212379 (24) [back to overview]Evolution of the Level of MCP1 From D0 to W48 on Frozen Samples
NCT02212379 (24) [back to overview]Evolution of the Ovarian Reserve From D0 to W48 Measured by AMH on Frozen Aliquots
NCT02212379 (24) [back to overview]Evolution of Total Cell-associated HIV-DNA
NCT02212379 (24) [back to overview]Factors Associated With the Occurrence of Plasma HIV-RNA Viral Load > 50 Copies/mL
NCT02212379 (24) [back to overview]Inflammatory Parameters
NCT02212379 (24) [back to overview]Number of Participants Experiencing Adverse Events and Effects
NCT02212379 (24) [back to overview]Percentage of Participants Compliant With Treatment Program.
NCT02212379 (24) [back to overview]Percentage of Participants Reporting a Very Good or an Excellent Quality of Life at Day 0, Weeks 48 and 96
NCT02212379 (24) [back to overview]Percentage of Participants With Successful Virological Suppression at Weeks 48 and 96
NCT02212379 (24) [back to overview]Percentage of Patients With High Grade of Virological Failure Defined as HIV RNA > 200 Copies/mL
NCT02212379 (24) [back to overview]Percentage of Patients With Therapeutic Success at Week 48 and Week 96
NCT02212379 (24) [back to overview]Percentage of Patients With Trial Treatment Interruption at Week 48 and Week 96
NCT02212379 (24) [back to overview]Percentage of Patients With With Grade Virological Failure (HIV-RNA Plasma VL Between 51 and 200 Copies/mL)
NCT02212379 (24) [back to overview]Sub-study in Women : Comparison of the Evolution Fat Distribution Measured by DXA Scan and Body Weight According to AMH Status
NCT02212379 (24) [back to overview]Sub-study in Women : Comparison of the Metabolic/Inflammatory Profile in Women According to Their Ovarian Reserve and Menopausal Status at D0 and Its Evolution up to Week 96
NCT02212379 (24) [back to overview]Sub-study: Bone Mineral Density
NCT04630002 (54) [back to overview]Cohort 1: Tmax of GSK3640254
NCT04630002 (54) [back to overview]Cohort 1: Time of Maximum Observed Concentration (Tmax) of DRV
NCT04630002 (54) [back to overview]Cohort 1: Plasma Concentration at the End of the Dosing Interval (Ctau) of DRV
NCT04630002 (54) [back to overview]Cohort 1: Maximum Observed Concentration (Cmax) of GSK3640254
NCT04630002 (54) [back to overview]Cohort 1: Ctau of RTV
NCT04630002 (54) [back to overview]Cohort 1: Ctau of GSK3640254
NCT04630002 (54) [back to overview]Cohort 1: Cmax of RTV
NCT04630002 (54) [back to overview]Cohort 1: Cmax of DRV
NCT04630002 (54) [back to overview]Cohort 1: AUC(0-tau) of RTV
NCT04630002 (54) [back to overview]Cohort 1: AUC(0-tau) of DRV
NCT04630002 (54) [back to overview]Cohort 1: Area Under the Plasma Concentration-time Curve From Time Zero to the End of the Dosing Interval at Steady State (AUC[0-tau]) of GSK3640254
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With Vital Sign Values of PCI Criteria
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With SAEs and Non-SAEs
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Urinalysis Parameters
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Hematology Parameters
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Glucose, Triglycerides, Lipase, Urate and Cholesterol
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Calcium, Creatine Kinase, Creatinine, Phosphate, Potassium and Sodium
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Alanine Aminotransferase, Albumin, Alkaline Phosphatase, Amylase, Aspartate Aminotransferase, Bilirubin and Direct Bilirubin
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With Clinically Significant Abnormal ECG Findings
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With Clinically Significant Abnormal ECG Findings
NCT04630002 (54) [back to overview]Cohort 3: Number of Participants With AEs Leading to Discontinuations and Deaths
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Vital Sign Values of PCI Criteria
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With SAEs and Non-SAEs
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Urinalysis Parameters
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Hematology Parameters
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Alanine Aminotransferase, Albumin, Alkaline Phosphatase, Amylase, Aspartate Aminotransferase, Bilirubin and Direct Bilirubin
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Clinically Significant Abnormal ECG Findings
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Clinically Significant Abnormal ECG Findings
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With AEs Leading to Discontinuations and Deaths
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Calcium, Creatine Kinase, Creatinine, Phosphate, Potassium and Sodium
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Vital Sign Values of Potential Clinical Importance (PCI) Criteria
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Serious Adverse Events (SAEs) and Non-serious Adverse Events (Non-SAEs)
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Urinalysis Parameters
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Glucose, Triglycerides, Lipase, Urate and Cholesterol
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Hematology Parameters
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Glucose, Triglycerides, Lipase, Urate and Cholesterol
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Calcium, Creatine Kinase, Creatinine, Phosphate, Potassium and Sodium
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Alanine Aminotransferase, Albumin, Alkaline Phosphatase, Amylase, Aspartate Aminotransferase, Bilirubin and Direct Bilirubin
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Clinically Significant Abnormal Electrocardiogram (ECG) Findings
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Clinically Significant Abnormal Electrocardiogram (ECG) Findings
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With Clinically Significant Abnormal Electrocardiogram (ECG) Findings
NCT04630002 (54) [back to overview]Cohort 1: Number of Participants With AEs Leading to Discontinuations and Deaths
NCT04630002 (54) [back to overview]Cohort 3: Cmax of GSK3640254
NCT04630002 (54) [back to overview]Cohort 3: AUC(0-tau) of GSK3640254
NCT04630002 (54) [back to overview]Cohort 2: Tmax of GSK3640254
NCT04630002 (54) [back to overview]Cohort 2: Tmax of ETR
NCT04630002 (54) [back to overview]Cohort 2: Number of Participants With Clinically Significant Abnormal ECG Findings
NCT04630002 (54) [back to overview]Cohort 2: Ctau of GSK3640254
NCT04630002 (54) [back to overview]Cohort 2: Ctau of ETR
NCT04630002 (54) [back to overview]Cohort 2: Cmax of GSK3640254
NCT04630002 (54) [back to overview]Cohort 2: Cmax of ETR
NCT04630002 (54) [back to overview]Cohort 2: AUC(0-tau) of GSK3640254
NCT04630002 (54) [back to overview]Cohort 2: AUC(0-tau) of ETR
NCT04630002 (54) [back to overview]Cohort 1: Tmax of RTV

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: 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: 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: 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]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

Median Change From TMC125-C229 Basline in Cluster of Differentiation 4 (CD4+) Cell Count at Week 48

The last visit of the TMC125 feeder study (TMC125-C203 [NCT00412646], TMC125-C223 [NCT00081978], TMC125 C211 [NCT00111280] or TMC125-C209 feeder studies) was considered to be the TMC125-C229 baseline. (NCT00128830)
Timeframe: Week 48

Interventionx 100000 cells/L (Median)
Etravirine21.50

[back to top]

Median Change in Cluster of Differentiation 4 (CD4+) Cell Count From Baseline in TMC125-C229 Feeder Study at Week 192

Baseline considered for this outcome is the baseline in the respective TMC125-C229 feeder study (TMC125-C203 [NCT00412646], TMC125-C223 [NCT00081978], TMC125 C211 [NCT00111280] or TMC125-C209 feeder studies). (NCT00128830)
Timeframe: Week 192

Interventionx 1000000 cells/mL (Median)
Etravirine149.50

[back to top]

Median Change in Cluster of Differentiation 4 (CD4+) Cell Count From Baseline in TMC125-C229 Feeder Study at Week 96

Baseline considered for this outcome is the baseline in the respective TMC125-C229 feeder study (TMC125-C203 [NCT00412646], TMC125-C223 [NCT00081978], TMC125 C211 [NCT00111280] or TMC125-C209 feeder studies). (NCT00128830)
Timeframe: Week 96

Interventionx 1000000 cells/mL (Median)
Etravirine120.00

[back to top]

Number of Participants Who Achieved Virologic Response (ie, Viral Load Less Than 50 Copies/mL) at Week 48

Number of participants who had viral load more than or equal to 50 copies/mL and less than 50 copies/mL at TMC125-C229 baseline and who achieved virologic response (ie, viral load less than 50 copies/mL) at Week 48. The last visit of the TMC125 feeder study (TMC125-C203 [NCT00412646], TMC125-C223 [NCT00081978], TMC125 C211 [NCT00111280] or TMC125-C209 feeder studies) was considered to be the TMC125-C229 baseline. (NCT00128830)
Timeframe: Week 48

InterventionParticipants (Number)
Viral Load More Than or Equal to 50 Copies/mL22
Viral Load Less Than 50 Copies/mL80

[back to top]

Number of Participants Who Achieved Virologic Response (ie, Viral Load Less Than 50 Copies/mL) at Week 96

Number of participants who had viral load more than or equal to 50 copies/mL and less than 50 copies/mL at TMC125-C229 baseline and who achieved virologic response (ie, viral load less than 50 copies/mL) at Week 96. The last visit of the TMC125 feeder study (TMC125-C203 [NCT00412646], TMC125-C223 [NCT00081978], TMC125 C211 [NCT00111280] or TMC125-C209 feeder studies) was considered to be the TMC125-C229 baseline. (NCT00128830)
Timeframe: Week 96

InterventionParticipants (Number)
Viral Load More Than or Equal to 50 Copies/mL28
Viral Load Less Than 50 Copies/mL72

[back to top]

Number of Participants With Adverse Events

Number of participants who reported at least 1 of the adverse events. (NCT00128830)
Timeframe: Up to 3 years

InterventionParticipants (Number)
Etravirine195

[back to top]

Number of Participants Who Achieved Virologic Response (ie, Viral Load Less Than 50 Copies/mL; Viral Load Less Than 400 Copies/mL; and Greater Than or Equal to 1 log10 Decrease From Baseline) at Week 192

Baseline considered for this outcome is the baseline in the respective TMC125-C229 feeder study (TMC125-C203 [NCT00412646], TMC125-C223 [NCT00081978], TMC125 C211 [NCT00111280] or TMC125-C209 feeder studies). (NCT00128830)
Timeframe: Week 192

InterventionParticipants (Number)
Viral load (VL) less than 50 copies/mLVL less than 400 copies/mLVL greater than or equal to 1log10
Etravirine698183

[back to top]

Number of Participants With Emerging Mutation (Reverse Transcriptase Mutation)

Emerging mutations are the mutation which are not present at baseline (last visit of the TMC125 feeder study [TMC125-C203 (NCT00412646), TMC125-C223 (NCT00081978), TMC125 C211 (NCT00111280) or TMC125-C209 feeder studies]) and are present at endpoint (last available timepoint during treatment period for each individual participant). (NCT00128830)
Timeframe: Baseline and Endpoint (ie, the last available time point during the treatment period)

InterventionParticipants (Number)
L100IY181CA98GV179IK103NH221Y
Etravirine876655

[back to top]

Median Change From TMC125-C229 Baseline in Cluster of Differentiation 4 (CD4+) Cell Count at Week 96

The last visit of the TMC125 feeder study (TMC125-C203 [NCT00412646], TMC125-C223 [NCT00081978], TMC125 C211 [NCT00111280] or TMC125-C209 feeder studies) was considered to be the TMC125-C229 baseline. (NCT00128830)
Timeframe: Week 96

Interventionx 1000000 cells/L (Median)
Etravirine18.50

[back to top]

Number of Participants Who Achieved Virologic Response (ie, Viral Load Less Than 50 Copies/mL; Less Than 400 Copies/mL; and Greater Than or Equal to 1 Log 10 Decrease From Baseline) at Week 96

Baseline considered for this outcome is the baseline in the respective TMC125-C229 feeder study (TMC125-C203 [NCT00412646], TMC125-C223 [NCT00081978], TMC125 C211 [NCT00111280] or TMC125-C209 feeder studies). (NCT00128830)
Timeframe: Week 96

InterventionParticipants (Number)
Viral load (VL) less than 50 copies/mLViral load less than 400 copies/mLViral load more than or equal to 1log10
Etravirine105124132

[back to top]

Change in Plasma Viral Load Versus Baseline (ie, Mean Change in log10 Plasma Viral Load From Baseline Over Time)

In the table below, the total number of participants analyzed in the Duet Placebo and Duet TMC125 groups, respectively at each time point were: Baseline (256;247 participants), Week 24 (251;240 participants), Week 48 (235;192 participants), and Week 96 (123;69 participants). (NCT00359021)
Timeframe: Baseline, Week 24, Week 48, and Week 96

,
Interventionlog10 copies/mL (Mean)
Week 24Week 48Week 96
DUET PLACEBO-0.8-0.7-0.5
DUET TMC1250-0.1-0.2

[back to top]

The Number of Participants Experiencing Adverse Events

The table below provides the number of participants who experienced Serious Adverse Events (SAEs) and Other Adverse Events (except SAEs) that started or worsened in severity during the overall TMC125-C217 treatment period. The duration of treatment ranged per patient from 1 week to 180 weeks, with a median of 62 weeks. (NCT00359021)
Timeframe: 1 week to 180 weeks, with a median of 62 weeks

,,
InterventionParticipants (Number)
Serious Adverse Events (SAEs)Other Adverse Events (AEs)
All Participants88297
DUET PLACEBO46160
DUET TMC12542137

[back to top]

The Percentage of Participants With Virologic Outcomes Over Time

The table below shows the percentage of participants with virologic suppression (< 50 copies/mL), the percentage of participants who were virologic failures (VF) (>50 copies/mL, discontinued prior to time X for reasons of VF or for other reasons, except for VF or adverse event, with a last viral load >50 copies/mL), and the percentage of participants with no viral load (VL) data available over time (ie, at Weeks 24, 48, and 96). (NCT00359021)
Timeframe: Weeks 24, 48, and 96

,
InterventionPercentage of Participants (Number)
Week 24 - Virologic Response (<50 cop/mL)Week 24 - Virologic FailureWeek 24 - No VL Data availableWeek 48 - Virologic Response (<50 cop/mL)Week 48 - Virologic FailureWeek 48 - No VL Data availableWeek 96 -Virologic Response (<50 cop/mL)Week 96 - Virologic FailureWeek 96 - No VL Data available
DUET PLACEBO43.046.910.235.250.414.57.448.044.5
DUET TMC12562.331.66.144.531.224.34.527.568.0

[back to top]

Median Change From Baseline in Total Cholesterol at Week 48.

(NCT00460746)
Timeframe: Week 48

Interventionmg/dL (Median)
Baseline (Day 1)Week 48 Change from Baseline
Darunavir(TMC114)/Eravirine(TMC125)189.0-27.5

[back to top]

Median Change From Baseline in Triglycerides at Week 48.

(NCT00460746)
Timeframe: Week 48

Interventionmg/dL (Median)
Baseline (Day 1)Week 48 Change from Baseline
Darunavir(TMC114)/Eravirine(TMC125)229.0-24.5

[back to top]

Proportion of Patients Who Have Viral Load Measurements <50 Copies/ml at 2, 4, 8, 12, 16, 24, 36 and 48 Weeks After Switching to DRV/r and ETR, Missing Equals Failure.

(NCT00460746)
Timeframe: 48 weeks

Interventionpercentage of participants (Number)
Baseline (Day 1)Week 2Week 4Week 8Week 12Week 16Week 24Week 36Week 48Post-Treatment Follow Up
Darunavir(TMC114)/Eravirine(TMC125)100909090809090908090

[back to top]

Proportion of Patients Who Maintain Plasma HIV Viral Load Measurements < 400 Copies/ml at 2, 4, 8, 12, 16, 24, 36 and 48 Weeks After Switching to DRV/r and ETR, Missing Equals Failure.

(NCT00460746)
Timeframe: 48 weeks

Interventionpercentage of participants (Number)
Baseline (Day 1)Week 2Week 4Week 8Week 12Week 16Week 24Week 36Week 48Post-Treatment Follow Up
Darunavir(TMC114)/Eravirine(TMC125)10010010090809090908090

[back to top]

CD4+ Cell Count (x 10^6 Cell/L): Baseline and Mean Changes From Baseline at 4, 8, 12, 16, 24,36 and 48 Weeks.

(NCT00460746)
Timeframe: Week 48

Interventioncells/mm^3 (Mean)
Baseline (Day 1)Week 4 Change from BaselineWeek 8 Change from BaselineWeek 12 Change from BaselineWeek 16 Change from BaselineWeek 24 Change from BaselineWeek 36 Change from BaselineWeek 48 Change from Baseline
Darunavir(TMC114)/Eravirine(TMC125)338.32.3-25.0-18.90.524.522.747.3

[back to top]

CD4+ Cell Count (x 10^6 Cell/L): Baseline and Median Changes From Baseline at 4, 8, 12, 16, 24, 36 and 48 Weeks.

(NCT00460746)
Timeframe: Week 48

Interventioncells/mm^3 (Median)
Baseline (Day 1)Week 4 Change from BaselineWeek 8 Change from BaselineWeek 12 Change from BaselineWeek 16 Change from BaselineWeek 24 Change from BaselineWeek 36 Change from BaselineWeek 48 Change from Baseline
Darunavir(TMC114)/Eravirine(TMC125)301.0-12.0-32.0-17.0-7.019.038.063.5

[back to top]

Median Change From Baseline in Glucose at Week 48.

(NCT00460746)
Timeframe: Week 48

Interventionmg/dL (Median)
Baseline (Day 1)Week 48 Change from Baseline
Darunavir(TMC114)/Eravirine(TMC125)92.51.0

[back to top]

Median Change From Baseline in HDL Cholesterol.

(NCT00460746)
Timeframe: Week 48

Interventionmg/dL (Median)
Baseline (Day 1)Week 48 Change from Baseline
Darunavir(TMC114)/Eravirine(TMC125)47.5-2.5

[back to top]

Median Change From Baseline in LDL Cholesterol at Week 48.

(NCT00460746)
Timeframe: Week 48

Interventionmg/dL (Median)
Baseline (Day 1)Week 48 Change from Baseline
Darunavir(TMC114)/Eravirine(TMC125)100.0-9.0

[back to top]

Median Change From Baseline in Total Cholesterol (TC) / High Denisty Lipoprotein (HDL) Ratio at Week 48.

(NCT00460746)
Timeframe: Week 48

Interventionratio of TC and HDL (Median)
Baseline (Day 1)Week 48 Change from Baseline
Darunavir(TMC114)/Eravirine(TMC125)4.7-0.3

[back to top]

Virologic Response < 50 HIV-1 RNA Copies/mL (ITT-Observed Case)

Virologic Response < 50 HIV-1 RNA Copies/mL (ITT-Observed Case). (NCT00534352)
Timeframe: Day 8, 14, 22, 28, 42 and Week 48

Interventionparticipants (Number)
Day 8 (n=19)Day 14 (n=21)Day 22 (n=19)Day 28 (n=20)Day 42 (n=20)Week 48 (n=13)
TDF/FTC +/- TMC125 +/- DRV/Rtv0346710

[back to top]

Number of Participants With Treatment-Emergent Non-Graded Laboratory Abnormalities(Worst Abnormality): Lipids- High-density Lipoprotein (HDL)

"Number of participants with Treatment-Emergent Non-Graded Laboratory Abnormalities(Worst Abnormality): Lipids- High-density lipoprotein (HDL).~Normal Range:~40 - 59 mG/dL 1.03 - 1.53 mmol/L" (NCT00534352)
Timeframe: Day 1 through 42 and Week 48

,,,
Interventionparticipants (Number)
Below 40 mG/dL (1.03 mmol/L)Above 59 mG/dL (1.53 mmol/L)
Optional Extension21
Treatment A40
Treatment B80
Treatment C60

[back to top]

Number of Participants With Treatment-Emergent Non-Graded Laboratory Abnormalities(Worst Abnormality): Glucose- Insulin

"Number of participants with Treatment-Emergent Non-Graded Laboratory Abnormalities(Worst Abnormality): Glucose- Insulin.~Normal Range: 3.0 - 27.0 ulU/mL" (NCT00534352)
Timeframe: Day 1 through 42 and Week 48

,,,
Interventionparticipant (Number)
Below 3.0 ulU/mLAbove 27.0 ulU/mL
Optional Extension13
Treatment A11
Treatment B23
Treatment C12

[back to top]

Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities(Worst Grade): Lipids- Triglycerides

"Number of participants with Treatment-Emergent Graded Laboratory Abnormalities(Worst Grade): Lipids- Triglycerides.~Worst Grade is based on the DAIDS toxicity grading scale, 0-5 : No Toxicity-Death." (NCT00534352)
Timeframe: Day 1 through 48 and Week 48

,,,
Interventionparticipants (Number)
Grade 1Grade 2
Optional Extension00
Treatment A00
Treatment B00
Treatment C00

[back to top]

Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities(Worst Grade): Lipids- Low-density Lipoprotein (LDL) Direct

"Number of participants with Treatment-Emergent Graded Laboratory Abnormalities(Worst Grade): Lipids- Low-density lipoprotein (LDL) Direct.~Worst Grade is based on the DAIDS toxicity grading scale, 0-5 : No Toxicity-Death." (NCT00534352)
Timeframe: Day 1 through 42 and Week 48

,,,
Interventionparticipants (Number)
Grade 1Grade 2
Optional Extension11
Treatment A00
Treatment B00
Treatment C11

[back to top]

Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities (Worst Grade): Lipids- Total Cholesteral

"Number of participants with Treatment-Emergent Graded Laboratory Abnormalities (Worst Grade): Lipids- Total Cholesteral.~Worst Grade is based on the DAIDS toxicity grading scale, 0-5 : No Toxicity-Death." (NCT00534352)
Timeframe: Day 1 through 42 and Week 48

,,,
Interventionparticipants (Number)
Grade 1Grade 2
Optional Extension10
Treatment A00
Treatment B00
Treatment C20

[back to top]

Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities (Worst Grade): Glucose-Hyperglycemia

"Number of Participants with Treatment-Emergent Graded Laboratory Abnormalities (Worst Grade): Glucose-Hyperglycemia.~Worst Grade is based on the National Institute of Allergy and Infectious Diseases Division of Acquired Immunodeficiency Syndrome (DAIDS) toxicity grading scale, 0,1,2,3,4 and 5 : None, Mild, Moderate, Severe, Life-threatening and Death." (NCT00534352)
Timeframe: Day 1 through 42 and Week 48

,,,
Interventionparticipants (Number)
Grade 1Grade 2
Optional Extension21
Treatment A11
Treatment B20
Treatment C01

[back to top]

Number of Participants With Treatment-Emergent Graded Laboratory Abnormalities (Worst Grade): Glucose- Hypoglycemia

"Number of participants with Treatment-Emergent Graded Laboratory Abnormalities (Worst Grade): Glucose- Hypoglycemia.~Worst Grade is based on the DAIDS toxicity grading scale 0-5: No Toxicity-Death." (NCT00534352)
Timeframe: Day 1 through 42 and Week 48

,,,
Interventionparticipants (Number)
Grade 1Grade 2
Optional Extension10
Treatment A00
Treatment B21
Treatment C00

[back to top]

Log10 Viral Load (HIV-1 RNA Copies/mL): Mean Changes From Baseline(ITT-Observed Case)

Log10 Viral Load (HIV-1 RNA copies/mL): Mean Changes From Baseline(ITT-Observed Case). (NCT00534352)
Timeframe: Baseline, Day 8, 14, 22, 28 & 42 and Week 48

Interventioncopies/mL (Mean)
Baseline (n=23)Day 8 (n=19)Day 14 (n=21)Day 22 (n=19)Day 28 (n=20)Day 42 (n=20)Week 48 (n=13)
TDF/FTC +/- TMC125 +/- DRV/Rtv4.19-1.41-1.71-1.77-1.86-2.04-2.30

[back to top]

CD4+ Cell Count (x 10^6 Cell/L): Baseline and Median Changes From Baseline (ITT-Observed Case)

CD4+ Cell Count (x 10^6 cell/L): Baseline and Median Changes From Baseline (ITT-Observed Case). (NCT00534352)
Timeframe: Baseline, Day 8, 14, 22, 28 & 42 ans Week 48

Interventionx 10^6 cell/L (Median)
Baseline (n=23)Day 8 (n=20)Day 14 (n=20)Day 22 (n=20)Day 28 (n=21)Day 42 (n=19)Week 48 (n=14)
TDF/FTC +/- TMC125 +/- DRV/Rtv403.045.594.059.062.056.0160.0

[back to top]

CD4+ Cell Count (Percent): Baseline and Median Changes From Baseline (ITT-Observed Case)

(NCT00534352)
Timeframe: Baseline, Day 8, 14, 22, 28 & 42 and Week 48

InterventionPercent Change from Baseline (Median)
Baseline (n=23)Day 8 (n=20)Day 14 (n=20)Day 22 (n=20)Day 28 (n=21)Day 42 (n=19)Week 48 (n=14)
TDF/FTC +/- TMC125 +/- DRV/Rtv26.20.14.21.83.04.23.8

[back to top]

Number of Participants Contributing to the Pharmacokinetic (PK) Evaluations: Cmin, Cmax, AUC24 & Css,av

At visit Days 14 & 28, samples were collected pre-dose and at 1, 2, 3, 4, 6, 9, and 12 hours post-dose. An additional sample was taken at 24 hours (Day 15 or 29 as applicable) post-dose. (NCT00534352)
Timeframe: 6 weeks

Interventionparticipants (Number)
Treatment A: TMC125 + TDF/FTC23
Treatment B: TMC125 + TDF/FTC + DRV/Rtv21

[back to top]

Change in Cardiovascular Risk Score From Baseline

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

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

[back to top]

Change in CD4 Count From Baseline

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

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

[back to top]

Change in Fasting Non-HDL Cholesterol From Baseline

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

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

[back to top]

Change in Summarized Quality of Life Score

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

Time From Randomization to Confirmed Virological Failure

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

Neuropsychological Performance Change

The outcome measure is change in performance from baseline to 16 weeks as measured by the global deficit score (GDS). The GDS is calculated by averaging the individual deficit scores from each neurocognitive test. Deficit scores for each test were calculated from age-, education-, gender-, and ethnicity-adjusted raw scores by methods that capture unexpectedly poor performance while ignoring better than expected performance. The GDS ranges in value from 0-5; higher scores indicate poorer cognitive functioning. Subjects with scores greater than or equal to 0.5 are considered cognitively impaired. (NCT00624195)
Timeframe: Baseline and 16 weeks

Interventionunits on a scale (Mean)
CNS-targeted-0.27
Non-CNS-targeted-0.17

[back to top]

The Change From Baseline in CD4 Cell Counts Over Time

(NCT00665847)
Timeframe: Baseline, Week 48

Intervention10E6 cells/L (Mean)
TMC125156

[back to top]

Population Pharmacokinetic (PK) Estimates of Etravirine/TMC125 (ETR): Trough Plasma Concentration (C0h)

(NCT00665847)
Timeframe: Week 48

Interventionng/mL (Mean)
TMC125346

[back to top]

The Percentage of Patients With Treatment-emergent Adverse Events (TEAEs)

The percentage of patients with a treatment-emergent adverse event (TEAE) (defined as an event that occurred in the 48-week treatment period during which it emerged [i.e. started or worsened in severity, relation, or other attribute], and not in the subsequent study periods, even if the event continued to be present] are provided below. Adverse events were graded from 1 to 4 in severity using the Division of Acquired Immunodeficiency Syndrome severity scale (grade 1 being less severe and grade 4 being more severe). ETR=etravirine/TMC125; OBR=optimized background regimen (NCT00665847)
Timeframe: 48 weeks

InterventionPercentage of patients (Number)
Any TEAETEAEs that were fatalTEAEs that were seriousTEAEs that were grade 3 or 4 in severityTEAEs leading to temporary ETR discontinuationTEAEs leading to permanent ETR discontinuationTEAEs possibly related to ETRTEAEs probably related to ETRTEAEs very likely related to ETRTEAEs at least possibly related to ETRTEAEs possibly related to OBRTEAEs probably related to OBRTEAEs very likely related to OBRTEAEs at least possibly related to OBRTEAEs of at least grade 2 in severityTEAEs of at least grade 3 in severityTEAEs of interest: Skin eventTEAEs of interest: RashTEAEs of interest: severe cutaneous reactionsTEAEs of interest: angioedemaTEAEs of interest: neuropsychiatric eventsTEAEs of interest: hepatic eventsTEAEs of interest: cardiac eventsTEAEs of interest: bleeding eventsTEAEs of interest: pancreatic eventsTEAEs of interest: lipid-related eventsTEAEs of interest: neoplasms
TMC12588.105.013.97.97.922.813.93.032.726.711.95.035.620.83.030.722.86.94.02.00001.05.91.0

[back to top]

The Emergence of Non-nucleoside Reverse Transcriptase Inhibitor Resistance-associated Mutations (NNRTI RAMs) in Patients Classified as Virologic Failures

Virologic failure (lack of response) was defined as: plasma viral load decline of < 0.5 log10 from Baseline by Week 8 and/or plasma viral load decline of <1.0 log10 from Baseline by Week 12. Virologic failure (loss of response) was defined as 2 consecutive measurements of plasma viral load > 0.5 log10 above the nadir after a minimum of 12 weeks of treatment. The table below provides data for 41 viologic failures of which 30 had mutation data available. In the table below, only the 4 most frequently emerging mutations are presented (emerging in at least 3 patients). (NCT00665847)
Timeframe: Baseline and Endpoint (up to Week 48)

InterventionPatients (Number)
V90IL100IE138AY181C
TMC1253338

[back to top]

The Number of Patients With Treatment-emergent Adverse Events (TEAEs)

A treatment-emergent adverse event (TEAE) was defined as an event that occurred in the 48-week treatment period during which it emerged (i.e. started or worsened in severity, relation, or other attribute), and not in the subsequent study periods, even if the event continued to be present. Adverse events were graded from 1 to 4 in severity using the Division of Acquired Immunodeficiency Syndrome severity scale (grade 1 being less severe and grade 4 being more severe). ETR=etravirine/TMC125; OBR=optimized background regimen (NCT00665847)
Timeframe: 48 weeks

InterventionPatients (Number)
Any TEAETEAEs that were fatalTEAEs that were seriousTEAEs that were grade 3 or 4 in severityTEAEs leading to temporary ETR discontinuationTEAEs leading to permanent ETR discontinuationTEAEs possibly related to ETRTEAEs probably related to ETRTEAEs very likely related to ETRTEAEs at least possibly related to ETRTEAEs possibly related to OBRTEAEs probably related to OBRTEAEs very likely related to OBRTEAEs at least possibly related to OBRTEAEs of at least grade 2 in severityTEAEs of at least grade 3 in severityTEAEs of interest: Skin eventTEAEs of interest: RashTEAEs of interest: severe cutaneous reactionsTEAEs of interest: angioedemaTEAEs of interest: neuropsychiatric eventsTEAEs of interest: hepatic eventsTEAEs of interest: cardiac eventsTEAEs of interest: bleeding eventsTEAEs of interest: pancreatic eventsTEAEs of interest: lipid-related eventsTEAEs of interest: neoplasms
TMC12589051488231433327125362133123742000161

[back to top]

Change From Baseline in Human Immunodeficiency Virus - Type 1 (HIV-1) Ribonucleic Acid (RNA) in Plasma Over Time

(NCT00665847)
Timeframe: Baseline, Week 48

Interventionlog10 copies/mL (Mean)
TMC125-1.53

[back to top]

Percentage of Patients With Virologic Response at Week 24

Virologic response was defined as the percentage of patients with plasma viral load < 50 copies/mL at Week 24 calculated according to the non-completer=failure (NC=F) imputation method. (NCT00665847)
Timeframe: Week 24

InterventionPercentage of Patients (Number)
TMC12552.5

[back to top]

Population Pharmacokinetic (PK) Estimates of Etravirine/TMC125 (ETR): Area Under the Plasma Concentration-time Curve Over 12 Hours at Steady-state (AUC12h)

The AUC12h is a Bayesian estimation based on a population pharmacokinetic model and sparse samples collected at each visit over the duration of trial. For each sparse sample taken, the time blood sample was recorded as well as the time of etravirine intake just prior to the time of blood sample. (NCT00665847)
Timeframe: Weeks 4-48

Interventionng.h/mL (Mean)
TMC1255216

[back to top]

Population Pharmacokinetic (PK) Estimates of Etravirine/TMC125 (ETR): Maximum Plasma Concentration (Cmax)

Etravirine/TMC125 (ETR) Cmax was approximated for each individual using the median value of plasma ETR concentrations taken 4 hours postdose (± 1 hour), when available, on the day of the Week 4 visit as shown in the table below. (NCT00665847)
Timeframe: Week 4

Interventionng/mL (Mean)
TMC125589

[back to top]

Percentage of Participants With HIV-1 RNA Levels <50 Copies/mL at Week 48 and 96

Plasma HIV-1 RNA level was determined by the Roche Amplicor HIV-1 Monitor standard assay (version 1.5). (NCT00823979)
Timeframe: Weeks 48, 96

,,
Interventionpercentage of participants (Number)
Week 48 (n=31, 32, 34)Week 96 (n=28, 28, 27)
Etravirine 200 mg61.837.0
Lersivirine 1000 mg31.314.3
Lersivirine 750 mg41.925.0

[back to top]

Percentage of Participants With Human Immunodeficiency Virus Type 1 Ribonucleic Acid (HIV-1 RNA) Levels Less Than (<) 50 Copies/Milliliter (mL) at Week 24

Plasma HIV-1 RNA level was determined by the Roche Amplicor HIV-1 Monitor standard assay (version 1.5). (NCT00823979)
Timeframe: Week 24

Interventionpercentage of participants (Number)
Lersivirine 750 mg48.4
Lersivirine 1000 mg43.8
Etravirine 200 mg67.6

[back to top]

Percentage of Participants With Response as Determined by the Time to Loss of Virologic Response (TLOVR50) Algorithm at Week 24, 48 and 96

TLOVR50 response (50 denotes lower limit of quantification [LLOQ] of assay=50 copies/mL): compliment to TLOVR50 failure. TLOVR50 failure based on observed HIV-1 RNA levels and failure events (death; permanent discontinuation of drug; lost to follow-up; new ARV drug; met treatment failure [TF] criteria). TF: an increase of at least (>=)3 times the baseline plasma HIV-1 RNA level at Week 2 or thereafter; failure to achieve HIV-1 RNA level <50 copies/mL at Week 24; starting at Week 2, an increase in HIV-1 RNA level to detectable levels (>50 copies/mL); HIV-1 RNA <1 log10 decrease from baseline at Week 4 or thereafter. TF were confirmed by second measurement >=14 days after first. Baseline value was calculated as the average of the measurements collected prior to and including Day 1 pre-dose. (NCT00823979)
Timeframe: Week 24, 48, 96

,,
Interventionpercentage of participants (Number)
Week 24Week 48
Etravirine 200 mg67.655.9
Lersivirine 1000 mg43.828.1
Lersivirine 750 mg48.441.9

[back to top]

Time-Averaged Difference (TAD) in log10 Transformed HIV-1 RNA Levels at Week 24, 48 and 96

TAD was calculated as (area under the curve of HIV-1 RNA levels [log10 copies/mL] from baseline to the time point of interest divided by time period in weeks) minus baseline HIV-1 RNA level (log10 copies/mL). Baseline value calculated as average of measurements collected at prior to and including Day 1 pre-dose. Due to early termination of the study decision was made not to derive TAD results for Week 96. (NCT00823979)
Timeframe: Week 24, 48, 96

,,
Interventionlog10 copies/mL (Mean)
Week 24Week 48
Etravirine 200 mg-2.10-1.89
Lersivirine 1000 mg-1.72-1.19
Lersivirine 750 mg-1.46-1.28

[back to top]

Percentage of Participants With HIV-1 RNA Levels <400 Copies/mL at Week 24, 48 and 96

Plasma HIV-1 RNA level was determined by the Roche Amplicor HIV-1 Monitor standard assay (version 1.5). (NCT00823979)
Timeframe: Week 24, 48, 96

,,
Interventionpercentage of participants (Number)
Week 24 (n=31, 32, 34)Week 48 (n=31, 32, 34)Week 96 (n=28, 28, 27)
Etravirine 200 mg79.470.637.0
Lersivirine 1000 mg50.034.414.3
Lersivirine 750 mg54.841.925.0

[back to top]

Change From Baseline in Cluster of Differentiation 4 (CD4+) Absolute Lymphocyte Counts at Week 24, 48 and 96

Blood samples for immunological status assessed by CD4+ lymphocyte count. Baseline value was calculated as the average of the measurements collected prior to and including Day 1 pre-dose. (NCT00823979)
Timeframe: Baseline, Week 24, 48, 96

,,
Interventioncells per microliter (cells/mcL) (Mean)
BaselineChange at Week 24Change at Week 48Change at Week 96
Etravirine 200 mg227110128184
Lersivirine 1000 mg259667489
Lersivirine 750 mg24798122148

[back to top]

Change From Baseline in Cluster of Differentiation 4 (CD4+) Percentage Lymphocyte Counts at Week 24, 48, 96

Blood samples for immunological status assessed by CD4+ lymphocyte count. Baseline value was calculated as the average of the measurements collected prior to and including Day 1 pre-dose. (NCT00823979)
Timeframe: Baseline, Week 24, 48, 96

,,
Interventionpercentage of total lymphocytes (Mean)
BaselineChange at Week 24Change at Week 48Change at Week 96
Etravirine 200 mg14.63.74.46.2
Lersivirine 1000 mg15.03.74.04.2
Lersivirine 750 mg16.13.04.65.4

[back to top]

Change From Baseline in log10 Transformed HIV-1 RNA Levels at Week 24, 48 and 96

Plasma HIV-1 RNA level was determined by the Roche Amplicor HIV-1 Monitor standard assay (version 1.5). For the log10 scale, all the HIV-1 RNA levels were log10 transformed prior to the average calculations. Baseline value calculated as average of measurements collected prior to and including Day 1 pre-dose. (NCT00823979)
Timeframe: Baseline, Week 24, 48, 96

,,
Interventionlog10 copies/mL (Mean)
BaselineChange at Week 24Change at Week 48Change at Week 96
Etravirine 200 mg4.59-2.32-2.06-0.96
Lersivirine 1000 mg4.76-1.89-1.08-0.36
Lersivirine 750 mg4.28-1.53-1.30-0.69

[back to top]

Number of Participants With Laboratory Test Abnormalities

Laboratory analysis included blood chemistry, hematology and urinalysis. (NCT00823979)
Timeframe: Baseline up to Week 48 or early termination

Interventionparticipants (Number)
Lersivirine 750 mg27
Lersivirine 1000 mg30
Etravirine 200 mg33

[back to top]

Number of Participants With Non-nucleoside Reverse Transcriptase Inhibitors (NNRTI) Resistance-Associated Mutations (RAMs) and/or Phenotypic Susceptibility at Time of Treatment Failure Through Week 48

Genotypic and phenotypic resistance to NNRTIs based on International Acquired Immunodeficiency Syndrome (AIDS) Society, United States of America (IAS-USA) RAM guidelines were evaluated using Monogram Biosciences PhenoSenseGT Assay at Baseline. This was then repeated for all participants with HIV-1 viral load >500 copies/mL at treatment failure, up to Week 48. (NCT00823979)
Timeframe: Baseline through Week 48

Interventionparticipants (Number)
Lersivirine 750 mg5
Lersivirine 1000 mg3
Etravirine 200 mg1

[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]

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]

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]

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]

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]

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]

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]

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]

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]

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]

Minimum Etravirine Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture

(NCT00855413)
Timeframe: Week 4 and week 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group4

[back to top]

Minimum Darunavir Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture

(NCT00855413)
Timeframe: Week 4 and week 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group10

[back to top]

Minimum Etravirine Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen

(NCT00855413)
Timeframe: Weeks 0-4 and weeks 12, 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group12

[back to top]

Minimum Etravirine Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure

(NCT00855413)
Timeframe: between Week 4-12 and between Weeks 36-48

Interventionng/g (Number)
Acute HIV Infection Treatment Group34

[back to top]

Minimum Ritonavir Exposure Range in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture

(NCT00855413)
Timeframe: Week 4 and week 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group2

[back to top]

Minimum Ritonavir Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure

(NCT00855413)
Timeframe: between week 4-12 and between weeks 36-48

Interventionng/g (Number)
Acute HIV Infection Treatment Group96

[back to top]

Number of Participants Who Stopped Study Treatment Due to Adverse Event or Intolerance

(NCT00855413)
Timeframe: Enrollment to Week 48

InterventionParticipants (Count of Participants)
Acute HIV Infection Treatment Group1

[back to top]

Number of Participants With HIV RNA Measurement Above the Limits of Detection in Cerebrospinal Fluid

(NCT00855413)
Timeframe: Week 4 and Week 48

Interventionparticipants (Number)
Acute HIV Infection Treatment Group0

[back to top]

Number of Participants With Neurocognitive Impairment at Baseline

(NCT00855413)
Timeframe: Week 2 or 4

InterventionParticipants (Count of Participants)
Acute HIV Infection Treatment Group8

[back to top]

Number of Participants With Neurocognitive Impairment at Week 24

(NCT00855413)
Timeframe: Week 24

InterventionParticipants (Count of Participants)
Acute HIV Infection Treatment Group3

[back to top]

Number of Participants With Neurocognitive Impairment at Week 48

(NCT00855413)
Timeframe: Week 48

InterventionParticipants (Count of Participants)
Acute HIV Infection Treatment Group3

[back to top]

Number of Participants With Virologic Response

Virologic response defined as plasma HIV RNA measurement <200 copies/mL at week 24 (NCT00855413)
Timeframe: 24 weeks

InterventionParticipants (Count of Participants)
Acute HIV Infection Treatment Group13

[back to top]

Number of Participants With Virologic Response

Virologic response to study treatment defined as plasma HIV RNA measurement <50 copies/mL at week 48 (NCT00855413)
Timeframe: 48 weeks from enrollment

InterventionParticipants (Count of Participants)
Acute HIV Infection Treatment Group9

[back to top]

Overall Neurocognitive Impairment at Week 24

Neuropsychological performance was assessed at week 2 or 4, week 24 and week 48 in the following measures: Premorbid/language (Wide Range Achievement Test 4 -Reading Subtest), Learning (Hopkins Verbal Learning Test - Revised), Memory (HVLT-R), Speed of Processing (Trailmaking A (Army Individual Test Battery, 1944), 1974, Stroop color, Attention (WAIS-III Symbol Search; Stroop word, Fine motor, Executive (Trailmaking B, Stroop interference, Letter, Category Fluency. An overall summary score of neurocognitive functioning was created by averaging all tests. Participants also completed Patient's Assessment of Own Functioning Inventory (PAOFI) and the Activities of Daily Living Scale (ADLS). Best available demographically corrected normative data were utilized to create z scores. A negative z score denotes below-average performance relative to a US normative comparison population. A higher z score is a better outcome. (NCT00855413)
Timeframe: Week 24

Interventionz score (Mean)
Acute HIV Infection Treatment Group-0.40

[back to top]

Overall Neurocognitive Impairment at Week 48

Neuropsychological performance was assessed at baseline (week 2 or 4), week 24 and week 48 in the following domain (measures): Premorbid/language (Wide Range Achievement Test (WRAT) 4 - Reading Subtest), Learning (HVLT-R, Hopkins Verbal Learning Test - Revised), Memory (HVLT-R), Speed of Processing (Trailmaking A (Army Individual Test Battery, 1944), 1974, Stroop color, Attention (WAIS-III Symbol Search; Stroop word, Fine motor, Executive (Trailmaking B, Stroop interference, Letter, Category Fluency. An overall summary score of neurocognitive functioning was created by averaging all tests. Participants also completed the self-reported functional status Patient's Assessment of Own Functioning Inventory (PAOFI) and the Activities of Daily Living Scale (ADLS). Best available demographically corrected normative data were utilized to create z scores. A negative z score denotes below-average performance relative to a US normative comparison population. A higher z score is a better outcome. (NCT00855413)
Timeframe: Week 48

Interventionz score (Mean)
Acute HIV Infection Treatment Group-.45

[back to top]

Overall Neurocognitive Impairment Score at Week 2 or 4

Neuropsychological performance was assessed at Week 2 or 4, Week 24 and Week 48 in the following measures: Premorbid/language (Wide Range Achievement Test 4 -Reading Subtest), Learning (HVLT-R, Hopkins Verbal Learning Test-Revised), Memory (HVLT-R), Speed of Processing (Trailmaking A (Army Individual Test Battery, 1944), 1974, Stroop color, Attention (WAIS-III Symbol Search; Stroop word, Fine motor, Executive (Trailmaking B, Stroop interference, Letter, Category Fluency. An overall summary score was created by averaging all tests. Participants also completed the self-reported functional status Patient's Assessment of Own Functioning Inventory (PAOFI) and the Activities of Daily Living Scale (ADLS). Best available demographically corrected normative data were utilized to create z scores. A negative z score denotes below-average performance relative to a US normative comparison population. A higher z score is a better outcome. (NCT00855413)
Timeframe: Week 2 or 4

Interventionz score (Mean)
Acute HIV Infection Treatment Group-0.69

[back to top]

Minimum Ritonavir Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen

(NCT00855413)
Timeframe: Weeks 0-4 and Weeks 12, 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group2

[back to top] [back to top]

Change in Overall Neurocognitive Impairment From Baseline to Week 24 or 48

Change in overall z score from baseline to week 24 or 48. A negative z score denotes below-average performance relative to a US normative comparison population. A higher z score is a better outcome. (NCT00855413)
Timeframe: Baseline to Week 24 or 48

Interventionz score (Mean)
Acute HIV Infection Treatment Group4.23

[back to top]

Correlation of HIV RNA Levels in CSF and Drug Levels With Neurocognitive Functioning

(NCT00855413)
Timeframe: From enrollment through Week 48

Interventionr value (Number)
Acute HIV Infection Treatment GroupNA

[back to top]

Correlation of Time to HIV RNA Levels <200 Copies/mL With Improvement in Neurocognitive Functioning From Baseline to Week 24 and 48

(NCT00855413)
Timeframe: Baseline to Week 24 and 48

Interventionr value (Number)
Acute HIV Infection Treatment Group-.82

[back to top]

HIV RNA Detection in Ileal Biopsy Specimens

Average HIV RNA detected in the ileal biopsy specimens per participant over weeks 4 and 48. (NCT00855413)
Timeframe: Weeks 4 and 48

Interventioncopies/mL (Mean)
Acute HIV Infection Treatment Group40

[back to top]

HIV RNA Detection in Semen

Total cumulative levels of HIV RNA detected in the semen of participants from enrollment through week 48. (NCT00855413)
Timeframe: From enrollment through 48 weeks

Interventioncopies/mL (Mean)
Acute HIV Infection Treatment Group2541

[back to top]

HIV RNA Levels Immediately Prior to Initiating Study Treatment.

(NCT00855413)
Timeframe: HIV RNA level at enrollment

Interventioncopies/mL (Median)
Acute HIV Infection Treatment Group1,000,000

[back to top]

Maximum Darunavir Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture

(NCT00855413)
Timeframe: Week 4 and week 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group240

[back to top]

Maximum Darunavir Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen

(NCT00855413)
Timeframe: Weeks 0-4 and weeks 12, 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group1296

[back to top]

Maximum Darunavir Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure

(NCT00855413)
Timeframe: between Week 4-12 and between Weeks 36-48

Interventionng/g (Number)
Acute HIV Infection Treatment Group83,749

[back to top]

Maximum Etravirine Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture

(NCT00855413)
Timeframe: Week 4 and week 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group25

[back to top]

Maximum Etravirine Exposure in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure

(NCT00855413)
Timeframe: between Week 4-12 and between Weeks 36-48

Interventionng/g (Number)
Acute HIV Infection Treatment Group83,749

[back to top]

Maximum Etravirine Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen

(NCT00855413)
Timeframe: Weeks 0-4 and weeks 12, 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group185

[back to top]

Maximum Ritonavir Exposure in Cerebrospinal Fluid Among Participants Who Consented to an Optional Lumbar Puncture

(NCT00855413)
Timeframe: Week 4 and Week 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group22

[back to top]

Maximum Ritonavir Exposure in Semen Among Participants Who Consented to an Optional Collection of Semen

(NCT00855413)
Timeframe: Weeks 0-4 and Weeks 12, 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group22

[back to top]

Maximum Ritonavir Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure

(NCT00855413)
Timeframe: between week 4-12 and between Weeks 36-48

Interventionng/g (Number)
Acute HIV Infection Treatment Group14,698

[back to top]

Median Change in CD4 Cell Count From Week 0 to Week 24.

(NCT00855413)
Timeframe: week 0, week 24

Interventioncells/mm^3 (Median)
Acute HIV Infection Treatment Group158

[back to top]

Median Change in CD4 Cell Count From Week 0 to Week 48.

(NCT00855413)
Timeframe: 48 weeks from enrollment

Interventioncells/mm^3 (Median)
Acute HIV Infection Treatment Group349

[back to top]

Median Time to HIV RNA Suppression to <200 Copies/mL

(NCT00855413)
Timeframe: From enrollment to the date of HIV RNA suppression, assessed up to Week 48

Interventiondays (Median)
Acute HIV Infection Treatment Group59

[back to top]

Minimum Darunavir Exposure Range in Ileal Tissue Among Participants Who Consented to an Optional Gut Biopsy Procedure

(NCT00855413)
Timeframe: between week 4-12 and between weeks 36-48

Interventionng/g (Number)
Acute HIV Infection Treatment Group987

[back to top]

Minimum Darunavir Exposure Range in Semen Among Participants Who Consented to an Optional Collection of Semen

(NCT00855413)
Timeframe: Weeks 0-4 and weeks 12, 48

Interventionng/mL (Number)
Acute HIV Infection Treatment Group9

[back to top]

Flow-mediated Dilation (FMD) of the Brachial Artery

FMD is measured as the percentage increase in brachial artery diameter after increase in blood flow. We measured the change in this percentage from entry (before etravirine was started) and again at four weeks after receiving etravirine. (NCT00871234)
Timeframe: Entry and four weeks

InterventionPercentage (Median)
Etravirine0.03

[back to top]

"Average Change in Activated (CD38+HLADR+) CD8+ T Cells in the Ileum"

Average of changes(week 0-week 12) in the % of CD8+ T cells that are CD38+HLA-DR+, by flow cytometry (NCT00884793)
Timeframe: 12 weeks

Interventionpercentage change (Mean)
Intensification Arm-5.4

[back to top]

Number of Subjects Who Experienced an Increase in CD4% in the Ileum.

Number of subjects who experienced an increase from week 0 to week 12 in CD4+ T cells (as a % of T cells, by flow cytometry) in the ileum (NCT00884793)
Timeframe: 12 weeks

Interventionparticipants (Number)
Intensification Arm5

[back to top]

Number of Subjects Who Experienced an Increase in CD4+ T Cells (as a % of All Cells) in the Ileum.

Number of subjects who experienced an increase in CD4+ T cells (as a % of all cells) in the ileum (by flow cytometry) from week 0 to week 12. (NCT00884793)
Timeframe: 12 weeks

Interventionparticipants (Number)
Intensification Arm6

[back to top]

Number of Subjects Who Had a Decrease in HIV RNA Per Million CD4+ T Cells in the Ileum

Number of subjects who had a decrease from week 0 to week 12 in unspliced cell-associated HIV RNA per million CD4+ T cells in the ileum (NCT00884793)
Timeframe: 12 weeks

Interventionparticipants (Number)
Intensification Arm5

[back to top]

Pharmacokinetic Results of Atazanavir (ATV): Treatment A: ATV/Low-Dose Ritonavir (Rtv) 300/100 mg (Results for C0h, Cmin, and Cmax)

The table below shows pharmacokinetic (PK) results of atazanavir (ATZ) when administered as ATV/rtv 300/100 mg pretreatment (Reference) and at Week 2 after treatment (Test). Results are expressed as the predose plasma concentration (C0h), minimum plasma concentration (Cmin), and maximum plasma concentration (Cmax). (NCT00896051)
Timeframe: Day -1 (Pretreatment); Week 2 (Test)

,
Interventionng/ml (Mean)
C0h, ng/ml (Reference, n=21; Test, n=19)Cmin, ng/ml (Reference, n=20; Test, n=18)Cmax, ng/ml (Reference, n=20; Test, n=19)
ATV/Rtv 300/100 mg (Reference)133911045652
ATV/Rtv 300/100 mg (Test)845.7758.65232

[back to top]

Percentage of Participants With Undetectable Plasma Viral Load (VL) Values (<50 Copies/mL) at Week 48

The table below shows the percentage of participants wih undetectable plasma viral load (VL) values (<50 copies/mL) at Week 48 using the Non-Completing = Failure (NC=F) imputation method (ie, participants who discontinued early were counted as nonresponders by having their VL values after discontinuation imputed with their baseline value, thus resulting in a 0 change). (NCT00896051)
Timeframe: Week 48

InterventionPercentage of Participants (Number)
ATV/Rtv 300/100 mg (Treatment A)50.0
ATV/Rtv 400/100 mg (Treatment B)45.5

[back to top]

Pharmacokinetic Results of Atazanavir (ATV): Treatment A: ATV/Low-Dose Ritonavir (Rtv) 300/100 mg (Results for AUC24hr)

The table below shows pharmacokinetic (PK) results of atazanavir (ATZ) when administered as ATV/rtv 300/100 mg pretreatment (Reference) and at Week 2 after treatment (Test). Results are expressed as the area under the plasma concentration-time curve from time of intake to 24 hours after dosing (AUC24hr). (NCT00896051)
Timeframe: Day -1 (Pretreatment); Week 2 (Test)

Interventionng.h/mL (Mean)
ATV/Rtv 300/100 mg (Reference)60030
ATV/Rtv 300/100 mg (Test)55070

[back to top]

Pharmacokinetic Results of Atazanavir (ATV): Treatment B: ATV/Low-Dose Ritonavir (Rtv) 400/100 mg (Results for AUC24hr)

The table below shows pharmacokinetic (PK) results of atazanavir (ATV) when administered as ATV/ritonavir (rtv) 300/100 mg pretreatment (Reference) and when administered as ATV/rtv 400/100 mg at Week 2 after treatment (Test). Results are expressed as the area under the plasma concentration-time curve from time of intake to 24 hours after dosing (AUC24hr). (NCT00896051)
Timeframe: Day -1 (Reference); Week 2 (Test)

Interventionng.h/mL (Mean)
ATV/Rtv 300/100 mg (Reference)74210
ATV/Rtv 300/100 mg (Test)72220

[back to top]

Pharmacokinetic Results of Etravirine (ETR) (Results for AUC12hr)

The table below shows pharmacokinetic (PK) results of ETR in the current study expressed as the area under the plasma concentration-time curve from time of intake to 12 hours after dosing (AUC12hr). (NCT00896051)
Timeframe: Week 2

Interventionng.h/mL (Mean)
ATV/Rtv 300/100 mg (Treatment A)7629
ATV/Rtv 400/100 mg (Treatment B)5171

[back to top]

Pharmacokinetic Results of Low-Dose Ritonavir (Rtv): Treatment A: Atazanavir (ATV)/Rtv 300/100 mg (Results for AUC24hr)

The table below shows the pharmacokinetic (PK) results of low-dose ritonavir (rtv) when administered as atazanavir (ATV)/rtv 300/100 mg pretreatment (Reference) and at Week 2 after treatment (Test). Results are expressed as the area under the plasma concentration-time curve from time of intake to 24 hours after dosing (AUC24hr). (NCT00896051)
Timeframe: Day -1 (Reference); Week 2 (Test)

Interventionng.h/ml (Mean)
ATV/Rtv 300/100 mg (Reference)12560
ATV/Rtv 300/100 mg (Test)11120

[back to top]

Pharmacokinetic Results of Low-Dose Ritonavir (Rtv): Treatment B: Atazanavir (ATV)/Rtv 400/100 mg (Results for AUC24hr)

The table below shows pharmacokinetic (PK) results of low-dose ritonavir (rtv) when administered as atazanavir (ATV)/ritonavir (rtv) 300/100 mg pretreatment (Reference) and when administered as ATV/rtv 400/100 mg at Week 2 after treatment (Test). Results are expressed as the area under the plasma concentration-time curve from time of intake to 24 hours after dosing (AUC24hr). (NCT00896051)
Timeframe: Day -1 (Reference); Week 2 (Test)

Interventionng.h/ml (Mean)
ATV/Rtv 300/100 mg (Reference)13880
ATV/Rtv 300/100 mg (Test)13660

[back to top]

Change From Pre-Baseline in Log10 Viral Load Over Time

The table below shows the mean change from prebaseline over time in log10 (Copies/mL) plasma viral load using the Non-Completing = Failure (NC=F) imputation method. (NCT00896051)
Timeframe: Pre-Baseline, Baseline, Weeks 4, 12, 24, 48

,
Interventionlog10 (Copies/mL) (Mean)
BaselineWeek 4Week 12Week 24Week 48
ATV/Rtv 300/100 mg (Treatment A)-1.4-1.9-1.7-1.8-1.4
ATV/Rtv 400/100 mg (Treatment B)-1.4-1.8-2.0-1.8-1.4

[back to top]

Change From Prebaseline in CD4+ Cell Count Over Time

The table below shows the mean change from prebaseline over time in CD4+ cell count using the Non-Completing = Failure (NC=F) imputation method. (NCT00896051)
Timeframe: Prebaseline, Baseline, Weeks 4, 12, 24, 48

,
InterventionCD4+ cell count (Mean)
BaselineWeek 4Week 12Week 24Week 48
ATV/Rtv 300/100 mg (Treatment A)16553154105
ATV/Rtv 400/100 mg (Treatment B)8467283132

[back to top]

Pharmacokinetic Results of Etravirine (ETR) (Results for C0h, Cmin, and Cmax)

The table below shows pharmacokinetic (PK) results of ETR in the current study expressed as the predose plasma concentration (C0h), minimum plasma concentration (Cmin) and maximum plasma concentration (Cmax). (NCT00896051)
Timeframe: Week 2

,
Interventionng/ml (Mean)
C0h (Treatment B, n=19)Cmin (Treatment A, n=16; Treatment B, n=18)Cmax (Treatment A, n=18; Treatment B, n=18)
ATV/Rtv 300/100 mg (Treatment A)422.2425.1773.0
ATV/Rtv 400/100 mg (Treatment B)316.6286.5628.7

[back to top]

Pharmacokinetic Results of Atazanavir (ATV): Treatment B: ATV/Low-Dose Ritonavir (Rtv) 400/100 mg (Results for C0h, Cmin, and Cmax)

The table below shows pharmacokinetic (PK) results of atazanavir (ATV) when administered as ATV/ritonavir (rtv) 300/100 mg pretreatment (Reference) and when administered as ATV/rtv 400/100 mg at Week 2 after treatment (Test). Results are expressed as the predose plasma concentration (C0h), minimum plasma concentration (Cmin), maximum plasma concentration (Cmax), and area under the plasma concentration-time curve from time of intake to 24 hours after dosing (AUC24hr). (NCT00896051)
Timeframe: Day -1 (Reference); Week 2 (Test)

,
Interventionng/ml (Mean)
C0h, ng/ml (Reference, n=22; Test, n=20)Cmin, ng/ml (Reference, n=21;Test, n=18)Cmax, ng/ml (Reference, n=22; Test, n=20)
ATV/Rtv 300/100 mg (Reference)189816716419
ATV/Rtv 400/100 mg (Test)154511076950

[back to top]

Time to Virologic Failure

The table below shows the number of days to virologic failure defined as a plasma viral load (VL) > 50 copies/mL for participants who had been virologic responders (ie, having a plasma VL <50, and <400 copies/mL according to the time to loss of virologic response [TLOVR] imputation method). Time to virologic failure was the time to subsequent loss of virologic response, and the time was calculated from Prebaseline (Week -2). Participants who never achieved a virologic response were defined as nonresponders and counted as virologic failures on Day 1. (NCT00896051)
Timeframe: Prebaseline to Week 48

,
InterventionDays (Median)
Virologic Responders (Plasma VL < 50 copies/mL)Virologic Responders (Plasma VL < 400 copies/mL)
ATV/Rtv 300/100 mg (Treatment A)318.0NA
ATV/Rtv 400/100 mg (Treatment B)NANA

[back to top]

Time to Confirmed Virologic Response

The table below provides the time in days it took participants to reach a confirmed virologic response defined as a plasma viral load (VL) <50 copies/mL, and plasma VL <400 copies/mL analyzed according to the Time to Loss of Virologic Response (TLOVR) imputation method. (NCT00896051)
Timeframe: Prebaseline to Week 48

,
InterventionDays (Median)
Plasma VL < 50 copies/mLPlasma VL < 400 copies/mL
ATV/Rtv 300/100 mg (Treatment A)71.028.0
ATV/Rtv 400/100 mg (Treatment B)76.028.0

[back to top]

The Percentage of Participants With a Virologic Response Using the Time to Loss of Virologic Response (TLOVR) Imputation Method

The table below shows the percentage of participants with a virologic response defined as a viral load <50 Copies/mL and <400 Copies/mL per time point calculated using the time to loss of virologic response (TLOVR) imputation method. (NCT00896051)
Timeframe: Baseline, Weeks 4, 12, 24, 48

,
InterventionPercentage of Particpants (Number)
<50 copies/mL, Baseline<50 copies/mL, Week 4<50 copies/mL, Week 12<50 copies/mL, Week 24<50 copies/mL, Week 48<400 copies/mL, Baseline<400 copies/mL, Week 4<400 copies/mL, Week 12<400 copies/mL, Week 24<400 copies/mL, Week 48
ATV/Rtv 300/100 mg (Treatment A)9.131.859.163.645.536.477.368.268.259.1
ATV/Rtv 400/100 mg (Treatment A)4.536.454.559.150.040.977.386.468.254.5

[back to top]

The Percentage of Participants With a Virologic Response Using the Non-Completing = Failure (NC=F) Imputation Method

The table below shows the percentage of participants per time point with a virologic response defined as having a plasma viral load (VL) <50 copies/mL, and with plasma VL <400 copies/mL using the Non-Completing = Failure (NC=F) imputation method (ie, participants who discontinued early were counted as nonresponders by having their VL values after discontinuation imputed with their Baseline value, thus resulting in a 0 change). (NCT00896051)
Timeframe: Baseline, Weeks 4, 12, 24, 48

,
InterventionPercentage of Participants (Number)
<50 copies/mL, Baseline<50 copies/mL, Week 4<50 copies/mL, Week 12<50 copies/mL, Week 24<50 copies/mL, Week 48<400 copies/mL, Baseline<400 copies/mL, Week 4<400 copies/mL, Week 12<400 copies/mL, Week 24<400 copies/mL, Week 48
ATV/Rtv 300/100 mg (Treatment A)9.131.859.163.650.040.977.368.272.750.0
ATV/Rtv 400/100 mg (Treatment B)9.136.459.163.645.540.977.381.872.759.1

[back to top]

The Percentage of Participants With a Virologic Response (Plasma Viral Load < 50 Copies/mL) at Week 48 Using the Snapshot Analysis Method

The table below provides the results from the snapshot analysis method that includes the percentage of participants with virologic response (<50 copies/mL), the percentage of participants who were virologic failures (VF) (>50 copies/mL, discontinued prior to time X for reasons of VF or for other reasons, except for VF or adverse event, with a last viral load >50 copies/mL), and the percentage of participants with no viral load (VL) data available at Week 48. (NCT00896051)
Timeframe: Week 48

,
InterventionPercentage of Participants (Number)
Virologic ResponseVirologic FailureNo VL Data Available
ATV/Rtv 300/100 mg (Treatment A)50.031.818.2
ATV/Rtv 400/100 mg (Treatment B)45.536.418.2

[back to top]

Pharmacokinetic Results of Low-Dose Ritonavir (Rtv): Treatment B: Atazanavir (ATV)/Rtv 400/100 mg (Results for C0h, Cmin, and Cmax)

The table below shows pharmacokinetic (PK) results of low-dose ritonavir (rtv) when administered as atazanavir (ATV)/ritonavir (rtv) 300/100 mg pretreatment (Reference) and when administered as ATV/rtv 400/100 mg at Week 2 after treatment (Test). Results are expressed as the predose plasma concentration (C0h), minimum plasma concentration (Cmin), maximum plasma concentration (Cmax), and area under the plasma concentration-time curve from time of intake to 24 hours after dosing (AUC24hr). (NCT00896051)
Timeframe: Day -1 (Reference); Week 2 (Test)

,
Interventionng/ml (Mean)
C0h, ng/ml (Reference, n=22; Test, n=20)Cmin, ng/mlCmax, ng/ml (Reference, n=22)
ATV/Rtv 300/100 mg (Reference)109.264.701882
ATV/Rtv 400/100 mg (Test)163.475.681847

[back to top]

Pharmacokinetic Results of Low-Dose Ritonavir (Rtv): Treatment A: Atazanavir (ATV)/Rtv 300/100 mg (Results for C0h, Cmin, and Cmax)

The table below shows the pharmacokinetic (PK) results of low-dose ritonavir (rtv) when administered as atazanavir (ATV)/rtv 300/100 mg pretreatment (Reference) and at Week 2 after treatment (Test). Results are expressed as the predose plasma concentration (C0h), minimum plasma concentration (Cmin), and maximum plasma concentration (Cmax). (NCT00896051)
Timeframe: Day -1 (Reference); Week 2 (Test)

,
Interventionng/ml (Mean)
C0h, ng/ml (Reference, n=21; Test, n=19)Cmin, ng/ml (Reference, n=20; Test, n=18)Cmax, ng/ml (Reference, n=20; Test, n=19)
ATV/Rtv 300/100 mg (Reference)143.460.421834
ATV/Rtv 300/100 mg (Test)102.543.971740

[back to top]

Antiviral Activity of ETR vs. EFV

The proportion of patients with confirmed plasma viral load <50 copies/mL at Week 48 as assessed by Time to Loss of Virologic Response (TLOVR) (NCT00903682)
Timeframe: between baseline and week 48

InterventionNumber of participants (Number)
Etravirine60
Efavirenz58

[back to top]

Neuropsychiatric Adverse Events by Week 48

The percentage of patients with at least 1 treatment emergent Grade 1 -4 neurologic or psychiatric adverse event, judged by the investigator to be at least possibly related to the study drug. (NCT00903682)
Timeframe: from baseline to week 48

Interventionpercentage of patients (Number)
Etravirine20.3
Efavirenz52.6

[back to top]

Proportion of Patients With at Least 1 Treatment-emergent Grade 1-4 Central Nervous System or Psychiatric Adverse Event

"Proportion of patients with at least 1 treatment-emergent Grade 1-4 Central Nervous System or psychiatric Adverse Event, observed between Baseline through Week 12 and judged by investigator to be at least possibly related to the study drug in ETR group versus EFV group. All Adverse Events were graded according to the Division of AIDS Table for Grading the Severity of Adult and Pediatric Adverse Events (DAIDS AE grading table). Grade 1-4 covers all severities." (NCT00903682)
Timeframe: between baseline and 12 weeks

Interventionpercentage of patients (Number)
Etravirine16.5
Efavirenz46.2

[back to top]

Mean Change From Baseline in CD4+ Cell Count

The mean change in CD4+ cell count from baseline was calculated with a last observation carried forward method; i.e. the last observed value was carried forward, irrespective of the reason for discontinuation. (NCT00903682)
Timeframe: at baseline and week 2, 6, 12, 24, 36 and 48

,
Interventionnumber of cells/L (x10^6) (Mean)
Week 2Week 6Week 12Week 24Week 36Week 48
Efavirenz72.45121.62151.46174.08180.18221.39
Etravirine69.96128.14143.24182.01213.45205.11

[back to top]

Mean Change From Baseline in Neuropsychiatric and Total Tolerabililty Score

"The HIV Patient Symptoms Profile measures the tolerability of HIV treatment from the patient's perspective, using 14 concept scales in maximum 84 questions. The response options include a no or yes answer to Did symptom occur?. If yes, there is a problem scale which ranges from 1 = I had this symptom and it was not a problem to 5 = I had this symptom and it was a severe problem. A neuropsychiatric tolerability score is composed as the sum of 21 items and ranges from 0 (best) to 105 (worse). A total Tolerability score (ie, the sum of all items) ranges from 0 (best) to 420 (worse)" (NCT00903682)
Timeframe: between baseline and week 48

,
Interventionpoints on a scale (Mean)
Total Tolerability ScoreNeuropsychiatric Tolerability Score
Efavirenz-0.01-0.07
Etravirine-0.04-0.04

[back to top]

Resistance Determinations

The evolution of viral genotype and phenotype was assessed by the number of patients with resistance-associated mutations emerging at the endpoint. A mutation was considered emerging if it was present at endpoint and not present at baseline or any pre-baseline assessment. (NNRTI = non-nucleoside reverse transcriptase inhibitor; NRTI = nucleoside reverse transcriptase inhibitor; RAM = resistance-associated mutation, IAS-USA = International AIDS Society - USA) (NCT00903682)
Timeframe: at baseline and all subsequent visits until week 48 in case if virologic failure

,
Interventionnumber of participants (Number)
>= 1 successful genotype after baseline>= 1 IAS-USA NRTI RAMs>= 1 NRTI Surveillance Drug Resistance Mutation>= 1 NNRTI RAMsno NRTI or NNRTI RAMs
Efavirenz92236
Etravirine110029

[back to top]

Antiviral Activity of ETR vs. EFV

The proportion of patients with confirmed plasma viral load <200 copies/mL at Week 48 as assessed by Time to Loss of Virologic Response (TLOVR) (NCT00903682)
Timeframe: between baseline and week 48

InterventionNumber of participants (Number)
Etravirine64
Efavirenz62

[back to top]

Probability of Remaining Free of a Safety/Tolerability Event at 96 Weeks

The Kaplan-Meier method was used to estimate the proportion of participants ever exposed to etravirine who remained event-free through Week 96, with a 95% CI using Greenwood's variance estimate and a log-log transformation. Time was handled as continuous (weeks from treatment start to event or censoring). (NCT00959894)
Timeframe: 96 weeks

Interventionproportion of participants (Number)
Etravirine 400 mg Once Daily0.69

[back to top]

Change in Limb and Trunk Fat Distribution as Measured by DEXA Scan, in the Same Subgroup of up to 40 Participants (as in Aim 8), From Baseline to Week 96 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

Changes from baseline to follow-up in limb fat, trunk fat, total body fat, and lean mass were calculated. Whole body Dual X-ray Absorptiometry (DEXA) scans (Hologic Discovery W, Hologic Inc., Bedford, MA) were conducted at baseline, Week 24, and Week 96 to assess body fat distribution. Calculations of change from baseline to follow-up used the value closest to schedule and within the analysis window, and were quantified with the estimated median and distribution-free 95% CI. (NCT00959894)
Timeframe: Baseline to 96 weeks

Interventionpercentage of body fat (Median)
Total body fatLimb fatTrunk fat
Etravirine 400 mg Once Daily1.440.821.93

[back to top]

Change in CD4+ Cell Count From Baseline to Week 24 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

The per-protocol analysis of change in CD4+ cell count from baseline to Week 24 was calculated using the measurement closest to schedule and within the analysis window, and quantified with an estimated median and distribution-free 95% confidence interval (CI). (NCT00959894)
Timeframe: Baseline to 24 weeks

Interventioncells/uL (Median)
Etravirine 400 mg Once Daily156

[back to top]

Change in CD4+ Cell Count From Baseline to Week 48 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

The per-protocol intention-to-treat analysis of change in CD4+ cell count from baseline to Week 48 was calculated using the measurement closest to schedule and within the analysis window, and quantified with an estimated median and distribution-free 95% CI. (NCT00959894)
Timeframe: Baseline to 48 weeks

Interventioncells/uL (Median)
Etravirine 400 mg Once Daily163

[back to top]

Change in CD4+ Cell Count From Baseline to Week 96 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

The per-protocol intention-to-treat analysis of change in CD4+ cell count from baseline to Week 96 was calculated using the measurement closest to schedule and within the analysis window, and quantified with an estimated median and distribution-free 95% CI. (NCT00959894)
Timeframe: Baseline to 96 weeks

Interventioncells/uL (Median)
Etravirine 400 mg Once Daily224

[back to top]

Change in Fat Mass Ratio as Measured by DEXA Scan, in the Same Subgroup of up to 40 Participants (as in Aim 8), From Baseline to Week 24 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

Change from baseline to follow-up in fat mass ratio was calculated. Whole body Dual X-ray Absorptiometry (DEXA) scans (Hologic Discovery W, Hologic Inc., Bedford, MA) were conducted at baseline, Week 24, and Week 96 to assess body fat distribution. Fat mass ratio was calculated as the ratio of trunk fat percentage and lower limb fat percentage (% trunk fat mass / % lower limb fat mass). Calculations of change from baseline to follow-up used the value closest to schedule and within the analysis window, and were quantified with the estimated median and distribution-free 95% CI. (NCT00959894)
Timeframe: Baseline to 24 weeks

Interventionratio of trunk fat % : lower limb fat % (Median)
Etravirine 400 mg Once Daily0.02

[back to top]

Change in Fat Mass Ratio as Measured by DEXA Scan, in the Same Subgroup of up to 40 Participants (as in Aim 8), From Baseline to Week 96 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

Change from baseline to follow-up in fat mass ratio was calculated. Whole body Dual X-ray Absorptiometry (DEXA) scans (Hologic Discovery W, Hologic Inc., Bedford, MA) were conducted at baseline, Week 24, and Week 96 to assess body fat distribution. Fat mass ratio was calculated as the ratio of trunk fat percentage and lower limb fat percentage (% trunk fat mass / % lower limb fat mass). Calculations of change from baseline to follow-up used the value closest to schedule and within the analysis window, and were quantified with the estimated median and distribution-free 95% CI. (NCT00959894)
Timeframe: Baseline to 96 weeks

Interventionratio of trunk fat % : lower limb fat % (Median)
Etravirine 400 mg Once Daily0.06

[back to top]

Change in Glucose Metabolism (Insulin Resistance), in a Subgroup of up to 40 Participants, From Baseline to Week 24 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

Metabolic data analyses were conducted as-treated. Insulin resistance was estimated by the homeostasis model assessment of insulin resistance (HOMA-IR), and was calculated as [fasting insulin (µU/mL) × fasting glucose (mmol/L)]/22.5. Changes from baseline to follow-up were calculated using the value closest to schedule and within the analysis window, and were quantified with the median and inter-quartile range. (NCT00959894)
Timeframe: Baseline to 24 weeks

InterventionµU/ml*mmol/L (Median)
Etravirine 400 mg Once Daily-0.08

[back to top]

Change in Glucose Metabolism (Insulin Resistance), in a Subgroup of up to 40 Participants, From Baseline to Week 48 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

Metabolic data analyses were conducted as-treated. Insulin resistance was estimated by the homeostasis model assessment of insulin resistance (HOMA-IR), and was calculated as [fasting insulin (µU/mL) × fasting glucose (mmol/L)]/22.5. Changes from baseline to follow-up were calculated using the value closest to schedule and within the analysis window, and were quantified with the median and inter-quartile range. (NCT00959894)
Timeframe: Baseline to 48 weeks

InterventionµU/ml*mmol/L (Median)
Etravirine 400 mg Once Daily0.71

[back to top]

Change in Glucose Metabolism (Insulin Resistance), in a Subgroup of up to 40 Participants, From Baseline to Week 96 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

Metabolic data analyses were conducted as-treated. Insulin resistance was estimated by the homeostasis model assessment of insulin resistance (HOMA-IR), and was calculated as [fasting insulin (µU/mL) × fasting glucose (mmol/L)]/22.5. Changes from baseline to follow-up were calculated using the value closest to schedule and within the analysis window, and were quantified with the median and inter-quartile range. (NCT00959894)
Timeframe: Baseline to 96 weeks

InterventionµU/ml*mmol/L (Median)
Etravirine 400 mg Once Daily0.23

[back to top]

Pharmacokinetics of Etravirine in Genital Secretions of up to 10 Men and up to 10 Women at Week 4 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

This secondary outcome measure assessed the ratio of semen:plasma concentration of etravirine in paired semen and plasma samples collected from 14 male participants at Week 4 of treatment with etravirine and fixed dose tenofovir/emtricitabine. (NCT00959894)
Timeframe: 4 weeks

Interventionratio of semen:plasma drug concentration (Median)
Etravirine 400 mg Once Daily0.192

[back to top]

Population Pharmacokinetics of Etravirine 400 mg Once Daily, in Combination With Fixed-dose Emtricitabine-tenofovir Among Treatment-naïve HIV-1 Infected Adults: Etravirine AUC-24 Hours at Steady State

Population pharmacokinetics were calculated using sparse sampling. Plasma concentrations of etravirine measured in samples from participants who provided blood samples at multiple study visits, with variation in sampling times relative to dosing of etravirine used to cover the spectrum of the dosing schedule. Model simulations and fitting were performed with NONMEM ® 7.3. (ICON, plc) and model exploration was performed with Berkeley Madonna (Berkeley, CA, USA) (NCT00959894)
Timeframe: At or after 4 weeks

Interventionng*hr/mL (Median)
Etravirine 400 mg Once Daily8024.40

[back to top]

The Antiretroviral Activity of Etravirine 400 mg Given Once Daily, With Fixed-dose Truvada Once Daily, Among Treatment-naïve HIV-1 Infected Adults as Measured by the Percentage of Participants With HIV RNA < 50 Copies/mL at Week 24

The primary study endpoint was the proportion of participants who achieved HIV-1 RNA <50 copies/ml at Week 24 of study participation. The per-protocol primary analysis was conducted intention-to-treat, with missing evaluations counted as failures. Achievement of HIV-1 viral load below 50 copies/ml was defined as having HIV-1 RNA <50 copies/ml during the Week 24 analysis window (>18 and <30 weeks post-entry). (NCT00959894)
Timeframe: 24 weeks

Interventionproportion of participants (Number)
Etravirine 400 mg Once Daily0.87

[back to top]

The Proportion of Participants With HIV RNA <200 Copies/mL at Week 24 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

This secondary outcome assessed the proportion of participants who achieved HIV-1 RNA <200 copies/ml at Week 24 of study treatment. The per-protocol analysis was conducted intention-to-treat, with missing evaluations counted as failures. (NCT00959894)
Timeframe: 24 weeks

Interventionproportion of participants (Number)
Etravirine 400 mg Once Daily0.89

[back to top]

The Proportion of Participants With HIV RNA <200 Copies/mL at Week 48 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

This secondary outcome assessed the proportion of participants who achieved HIV-1 RNA <200 copies/ml at Week 48 of study treatment. The per-protocol analysis was conducted intention-to-treat, with missing evaluations counted as failures. (NCT00959894)
Timeframe: 48 weeks

Interventionproportion of participants (Number)
Etravirine 400 mg Once Daily0.82

[back to top]

The Proportion of Participants With HIV RNA <200 Copies/mL at Week 96 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

This secondary outcome assessed the proportion of participants who achieved HIV-1 RNA 200 copies/ml at Week 96 of study treatment. The per-protocol analysis was conducted intention-to-treat, with missing evaluations counted as failures. (NCT00959894)
Timeframe: 96 weeks

Interventionproportion of participants (Number)
Etravirine 400 mg Once Daily0.77

[back to top]

The Proportion of Participants With HIV RNA <50 Copies/mL at Week 48 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

This secondary outcome assessed the proportion of participants who achieved HIV-1 RNA <50 copies/ml at Week 48 of study treatment. The per-protocol analysis was conducted intention-to-treat, with missing evaluations counted as failures. (NCT00959894)
Timeframe: 48 weeks

Interventionproportion of participants (Number)
Etravirine 400 mg Once Daily0.77

[back to top]

The Proportion of Participants With HIV RNA <50 Copies/mL at Week 96 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

This secondary outcome assessed the proportion of participants who achieved HIV-1 RNA <50 copies/ml at Week 96 of study treatment. The per-protocol analysis was conducted intention-to-treat, with missing evaluations counted as failures. (NCT00959894)
Timeframe: 96 weeks

Interventionproportion of participants (Number)
Etravirine 400 mg Once Daily0.71

[back to top]

Change in Limb and Trunk Fat Distribution as Measured by DEXA Scan, in the Same Subgroup of up to 40 Participants (as in Aim 8), From Baseline to Week 24 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

Changes from baseline to follow-up in limb fat, trunk fat, total body fat, and lean mass were calculated. Whole body Dual X-ray Absorptiometry (DEXA) scans (Hologic Discovery W, Hologic Inc., Bedford, MA) were conducted at baseline, Week 24, and Week 96 to assess body fat distribution. Calculations of change from baseline to follow-up used the value closest to schedule and within the analysis window, and were quantified with the estimated median and distribution-free 95% CI. (NCT00959894)
Timeframe: Baseline to 24 weeks

Interventionpercentage of body fat (Median)
Total body fatLimb fatTrunk fat
Etravirine 400 mg Once Daily0.430.480.32

[back to top]

Change in the Lipid Profile and Glucose Metabolism, in a Subgroup of up to 40 Participants, From Baseline to Week 24 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

Metabolic data analyses were conducted as-treated. Changes in total cholesterol, high-density lipoprotein (HDL) cholesterol, low-density lipoprotein (LDL) cholesterol, triglycerides, and fasting blood glucose from baseline to follow-up were calculated using the value closest to schedule and within the analysis window, and were quantified with the median and inter-quartile range. (NCT00959894)
Timeframe: Baseline to 24 weeks

Interventionmg/dL (Median)
Total cholesterolHDL-cholesterolLDL-cholesterolTriglyceridesFasting glucose
Etravirine 400 mg Once Daily-71-9-161

[back to top]

Change in the Lipid Profile and Glucose Metabolism, in a Subgroup of up to 40 Participants, From Baseline to Week 48 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

Metabolic data analyses were conducted as-treated. Changes in total cholesterol, high-density lipoprotein (HDL) cholesterol, low-density lipoprotein (LDL) cholesterol, triglycerides, and fasting blood glucose from baseline to follow-up were calculated using the value closest to schedule and within the analysis window, and were quantified with the median and inter-quartile range. (NCT00959894)
Timeframe: Baseline to 48 weeks

Interventionmg/dL (Median)
Total cholesterolHDL-cholesterolLDL-cholesterolTriglyceridesFasting glucose
Etravirine 400 mg Once Daily65-1-102

[back to top]

Change in the Lipid Profile and Glucose Metabolism, in a Subgroup of up to 40 Participants, From Baseline to Week 96 of Treatment With Etravirine and Fixed-dose Tenofovir/Emtricitabine

Metabolic data analyses were conducted as-treated. Changes in total cholesterol, high-density lipoprotein (HDL) cholesterol, low-density lipoprotein (LDL) cholesterol, triglycerides, and fasting blood glucose from baseline to follow-up were calculated using the value closest to schedule and within the analysis window, and were quantified with the median and inter-quartile range. Insulin resistance was estimated by the homeostasis model assessment of insulin resistance (HOMA-IR), and was calculated as [fasting insulin (µU/mL) × fasting glucose (mmol/L)]/22.5. (NCT00959894)
Timeframe: Baseline to 96 weeks

Interventionmg/dL (Median)
Total cholesterolHDL-cholesterolLDL-cholesterolTriglyceridesFasting glucose
Etravirine 400 mg Once Daily64-532

[back to top]

Population Pharmacokinetics of Etravirine 400 mg Once Daily, in Combination With Fixed-dose Emtricitabine-tenofovir Among Treatment-naïve HIV-1 Infected Adults

Population pharmacokinetics were calculated using sparse sampling. Plasma concentrations of etravirine measured in samples from participants who provided blood samples at multiple study visits, with variation in sampling times relative to dosing of etravirine used to cover the spectrum of the dosing schedule. Model simulations and fitting were performed with NONMEM ® 7.3. (ICON, plc) and model exploration was performed with Berkeley Madonna (Berkeley, CA, USA) (NCT00959894)
Timeframe: At or after 4 weeks

Interventionng/mL (Median)
Etravirine trough plasma concentrationEtravirine peak plasma concentration
Etravirine 400 mg Once Daily217.47480.99

[back to top]

Resistance Mutations in the Subset of Patients With Confirmed Virologic Failure Who Have HIV RNA >500 Copies/mL and Genotype Resistance Results

Per-protocol, genotype testing was conducted at confirmation of virologic failure if the confirmatory HIV-1 RNA was above the laboratory-specified threshold of 500 copies/mL. HIV-1 genotype was determined using the TRUGENE® HIV-1 assay (Siemens Healthcare Diagnostics, Tarrytown, NY) (NCT00959894)
Timeframe: 96 weeks

Interventionparticipants (Number)
Y181CE138KE138K, Y181C, M230L, M184I, K219E, V75INo resistance-associated mutations detected
Etravirine 400 mg Once Daily1113

[back to top]

Tolerability of Etravirine in HIV-1 Infected Adults Initiating Antiretroviral Therapy

"The safety/tolerability endpoint was defined as the first grade 3 or higher sign, symptom or laboratory abnormality that was at least one grade higher than baseline among participants ever exposed to etravirine (regardless of treatment status), or permanent discontinuation of etravirine due to any toxicity (regardless of grade). Modification of tenofovir/emtricitabine was not a safety/tolerability event.~The Kaplan-Meier method was used to estimate the proportion of participants ever exposed to etravirine who remained event-free through Week 96, with a 95% CI using Greenwood's variance estimate and a log-log transformation. Time was handled as continuous (weeks from treatment start to event or censoring)." (NCT00959894)
Timeframe: 96 weeks

Interventionparticipants (Number)
At least one safety/tolerability eventSigns or SymptomsLaboratory Abnormalities
Etravirine 400 mg Once Daily231310

[back to top]

Number of Participants With At-least One Adverse Event as a Measure of Safety Until Etravirine (ETR)-Based Treatment Regimen is Commercially Available

An adverse event is any untoward medical event that occurs in a participant administered an investigational product, and it does not necessarily indicate only events with clear causal relationship with the relevant investigational product. (NCT00980538)
Timeframe: Up to 10 years and 11 months

InterventionParticipants (Count of Participants)
Etravirine42

[back to top]

Percentage of Participants With Human Immunodeficiency Virus Type 1 (HIV-1) Ribonucleic Acid (RNA) <50 Copies Per Milliliter (/mL) at Week 16

Plasma for quantitative HIV-1 RNA was collected. The percentage of participants with HIV-1 RNA <50 copies/mL at Week 16 has been presented. A 2 ml plasma was assayed with the real-time NucliSens EasyQ HIV-1 assay (bioMerieux) capable of quantifying as low as 2.5 c/mL by using three modifications to the standard assay: 15 microliters (µl) of extracted eluate, 20 µl of primer, and 5 µl of 2X enzyme in place of standard kit volumes. The validated assay incorporated molecular beacons for detection. (NCT01199731)
Timeframe: At Week 16

InterventionPercentage of participants (Number)
ETV 200 mg33

[back to top]

Absolute Values of CD4+ Cell Counts After Switch of GSK2248761

CD4 is a receptor for the HIV virus. Most of the damage to an AIDS participant's immune system was done by the virus' destruction of CD4+ lymphocytes. Absolute values of CD4+ cell counts after Switch of GSK2248761 has been presented. (NCT01199731)
Timeframe: Baseline (Day 1) and post switch Week 1, 2, 4, withdrawal and Follow-up Week 4, 8, 12

InterventionCells per cubic millimeter (cells/mm^3) (Mean)
BaselineBaseline switchWeek 1 post switchWeek 2 post switchWeek 4 post switchWithdrawalWeek 4 follow-upWeek 8 follow-upWeek 12 follow-up
GSK2248761 200 mg162.5164.3189.9179.0199.6188.5237.9207.2329.3

[back to top]

Change From Baseline in CD4+ Cell Counts After Switch of GSK2248761

CD4 is a receptor for the HIV virus. Most of the damage to an AIDS patient's immune system is done by the virus' destruction of CD4+ lymphocytes. Change from Baseline was calculated as (observed value - Baseline value). Baseline was Day 1 value. (NCT01199731)
Timeframe: Baseline (Day 1) and post switch Week 1, 2, 4, withdrawal and Follow-up Week 4, 8, 12

InterventionCells/mm^3 (Mean)
Week 1 post switchWeek 2 post switchWeek 4 post switchWithdrawal4 Week follow-up8 Week follow-up
GSK2248761 100 mg48.258.874.044.159.866.3

[back to top]

Change From Baseline in CD4+ Cell Counts After Switch of GSK2248761

CD4 is a receptor for the HIV virus. Most of the damage to an AIDS patient's immune system is done by the virus' destruction of CD4+ lymphocytes. Change from Baseline was calculated as (observed value - Baseline value). Baseline was Day 1 value. (NCT01199731)
Timeframe: Baseline (Day 1) and post switch Week 1, 2, 4, withdrawal and Follow-up Week 4, 8, 12

InterventionCells/mm^3 (Mean)
Week 1 post switchWeek 2 post switchWeek 4 post switchWithdrawal4 Week follow-up8 Week follow-up12 Week follow-up
GSK2248761 200 mg7.44.316.626.072.032.3113.7

[back to top]

Change From Baseline in CD4+ Cell Counts Prior to Switch of GSK2248761

CD4 is a receptor for the HIV virus. Most of the damage to an AIDS patient's immune system is done by the virus' destruction of CD4+ lymphocytes. Change from Baseline was calculated as (observed value - Baseline value). Baseline was Day 1 value. (NCT01199731)
Timeframe: Baseline (Day 1) and Week 2, 4, 8, 12 and 16

,
InterventionCells/mm^3 (Mean)
Week 2Week 4Week 8Week 12
GSK2248761 100 mg43.0122.876.441.0
GSK2248761 200 mg21.260.757.330.0

[back to top]

Change From Baseline in CD4+ Cell Counts Prior to Switch of GSK2248761

CD4 is a receptor for the HIV virus. Most of the damage to an AIDS patient's immune system is done by the virus' destruction of CD4+ lymphocytes. Change from Baseline was calculated as (observed value - Baseline value). Baseline was Day 1 value. (NCT01199731)
Timeframe: Baseline (Day 1) and Week 2, 4, 8, 12 and 16

InterventionCells/mm^3 (Mean)
Week 2Week 4Week 8Week 12Week 16
ETV 200 mg62.281.471.061.5-7.0

[back to top]

Change From Baseline in Plasma HIV-1 RNA After Switch of GSK2248761

A switch was defined as any ART substitution or addition in the participant's ART regimen. Any ART switch permitted per protocol that was determined necessary and documented prior to the first on-treatment visit where HIV-1 RNA was assessed could occur without penalty. However, any participants with an ART switch not permitted per protocol or ART switch permitted per protocol with a viral load >=50 copies/mL at the time of the decision to switch was made was counted as a non-responder from that point onward for all assessment windows without a viral load measurement collected prior to the switch. Change from Baseline was calculated as (observed value - Baseline value ). Baseline was Day 1. (NCT01199731)
Timeframe: Baseline (Day 1) and post switch Week 1, 2, 4, withdrawal and follow-up Week 4, 8, 12

,
Interventionlog10 copies/mL (Mean)
Week 1 post switchWeek 2 post switchWeek 4 post switchWithdrawal4 Week follow-up8 Week follow-up12 Week follow-up
GSK2248761 100 mg-2.080-2.376-1.617-2.070-2.123-2.1010.278
GSK2248761 200 mg-2.197-2.229-2.545-1.978-1.999-1.251-2.272

[back to top]

Change From Baseline in Plasma HIV-1 RNA Prior to Switch of GSK2248761

A switch was defined as any ART substitution or addition in the participant's ART regimen. Any ART switch permitted per protocol that was determined necessary and documented prior to the first on-treatment visit where HIV-1 RNA was assessed could occur without penalty. However, any participants with an ART switch not permitted per protocol or ART switch permitted per protocol with a viral load >=50 copies/mL at the time of the decision to switch was made was counted as a non-responder from that point onward for all assessment windows without a viral load measurement collected prior to the switch. Change from Baseline was calculated as (observed value - Baseline value). Baseline was Day 1. The unit is log10 copies per milliliter (log10 copies/mL). (NCT01199731)
Timeframe: Baseline (Day 1) and Week 2, 4, 8, 12 and 16

,
InterventionLog10 copies/mL (Mean)
Week 2Week 4Week 8Week 12
GSK2248761 100 mg-1.845-1.985-2.332-1.997
GSK2248761 200 mg-2.075-2.621-3.030-3.085

[back to top]

Change From Baseline in Plasma HIV-1 RNA Prior to Switch of GSK2248761

A switch was defined as any ART substitution or addition in the participant's ART regimen. Any ART switch permitted per protocol that was determined necessary and documented prior to the first on-treatment visit where HIV-1 RNA was assessed could occur without penalty. However, any participants with an ART switch not permitted per protocol or ART switch permitted per protocol with a viral load >=50 copies/mL at the time of the decision to switch was made was counted as a non-responder from that point onward for all assessment windows without a viral load measurement collected prior to the switch. Change from Baseline was calculated as (observed value - Baseline value). Baseline was Day 1. The unit is log10 copies per milliliter (log10 copies/mL). (NCT01199731)
Timeframe: Baseline (Day 1) and Week 2, 4, 8, 12 and 16

InterventionLog10 copies/mL (Mean)
Week 2Week 4Week 8Week 12Week 16
ETV 200 mg-1.730-2.064-1.850-1.678-0.820

[back to top]

Number of Participants Who Experienced Disease Progression (HIV-associated Conditions, AIDS and Death)

Clinical DP was defined as progression from Baseline HIV disease status:Category A at Baseline to Centers for Disease Control and Prevention(CDC) category B event, category A at Baseline to CDC category C event, category B at Baseline to CDC category C event, category C at Baseline to new CDC category C event or category A, B or C at Baseline to death. Category A consisted of one or more of the conditions like asymptomatic HIV infection, persistent generalized lymphadenopathy and acute (primary) HIV infection with accompanying illness in an adolescent or adult(>13 years) with documented HIV infection. Category B consisted like Bacillary angiomatosis, Candidiasis, oropharyngeal (thrush), Candidiasis, vulvovaginal; persistent, frequent, oral, Herpes zoster etc. Category C included clinical conditions listed like Candidiasis of bronchi, trachea, or lungs, Candidiasis, esophageal, Cervical cancer, Coccidioidomycosis, disseminated or extrapulmonary etc in AIDS surveillance case definition. (NCT01199731)
Timeframe: Up to follow-up i.e. 2-4 weeks after the last study visit (due to early termination the last visit was on 19 July 2011)

,,
InterventionParticipants (Count of Participants)
Disease progressionDeath
ETV 200 mg00
GSK2248761 100 mg00
GSK2248761 200 mg00

[back to top]

Number of Participants With Electrocardiograph (ECG) With Values of Potential Critical Concern (PCI)

Number of participants with ECG of PCI above 480 has been presented. ECG was be performed twice on Day 1 at least 5 minutes apart and following 5 minutes of rest in a semi supine position at ∼1 hour prior to first dose. ECG evaluations at other visits was obtained after dosing, preferably at 2 hours post dosing. An ECG machine that automatically calculated the heart rate and measured PR, QRS, QT, and corrected QT (QTc) intervals was used. (NCT01199731)
Timeframe: Up to follow-up i.e. 2-4 weeks after the last study visit (due to early termination the last visit was on 19 July 2011)

InterventionParticipants (Count of Participants)
GSK2248761 100 mg0
GSK2248761 200 mg0
ETV 200 mg0

[back to top]

Number of Participants With Abnormal Clinical Chemistry Laboratory Data With Grade 3 or 4 Treatment-Emergent (TE) Toxicities

A toxicity was considered to be TE if it was greater than the Baseline grade, and if it had developed or increased post-Baseline in intensity (and prior to the last dose of IP). Division of acquired immunodeficiency syndrome (AIDS) toxicity scale for Grading the Severity of Adult and Pediatric Adverse Events Version 1.0 was used for grading i.e. Grade 3=severe and Grade 4=potentially life threatening. Categories with values have been presented. No toxicity-related dose reductions of IP was allowed. IP and background antiretroviral therapy (ART) was restarted as soon as medically appropriate; in general, this was no longer than 14] days after discontinuation (unless Grade 3 or 4 toxicities persisted). (NCT01199731)
Timeframe: Up to follow-up i.e. 2-4 weeks after the last study visit (due to early termination the last visit was on 19 July 2011)

,,
InterventionParticipants (Count of Participants)
Grade 3: GlucoseGrade 3: Hyperglycaemia
ETV 200 mg00
GSK2248761 100 mg11
GSK2248761 200 mg00

[back to top]

Number of Participants With Abnormal Hematology Laboratory Data With Grade 3 or 4 TE Toxicities

A toxicity was considered to be TE if it was greater than the Baseline grade, and if it had developed or increased post-Baseline in intensity (and prior to the last dose of IP). The hematology parameters included hemoglobin, total neutrophils, and white blood cells (WBC) count. Categories with values has been presented. Grade 3=severe and Grade 4=potentially life threatening. No toxicity-related dose reductions of IP was allowed. IP and background ART was restarted as soon as medically appropriate; in general, this was no longer than 14] days after discontinuation (unless Grade 3 or 4 toxicities persisted). (NCT01199731)
Timeframe: Up to follow-up i.e. 2-4 weeks after the last study visit (due to early termination the last visit was on 19 July 2011)

,,
InterventionParticipants (Count of Participants)
Grade 3: Total NeutrophilsGrade 3: WBC
ETV 200 mg00
GSK2248761 100 mg10
GSK2248761 200 mg11

[back to top]

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

An AE was defined as any untoward medical occurrence that occurred during the course of the trial after study treatment had started. An adverse event was therefore any unfavorable and unintended sign, symptom, or disease temporally associated with the use of study drug, whether or not considered related to the study drug. An SAE is any untoward medical occurrence that at any dose results in death, are life threatening, requires hospitalization or prolongation of hospitalization or results in disability/incapacity, and congenital anomaly/birth defect. (NCT01199731)
Timeframe: Up to follow-up i.e. 2-4 weeks after the last study visit (due to early termination the last visit was on 19 July 2011)

,,
InterventionParticipants (Count of Participants)
Any AEAny SAE
ETV 200 mg60
GSK2248761 100 mg71
GSK2248761 200 mg94

[back to top]

Number of Participants Who Discontinued Treatment Due to AEs

An AE is defined as any untoward medical occurrence in a participant temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product. An AE can therefore be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease (new or exacerbated) temporally associated with the use of a medicinal product. (NCT01199731)
Timeframe: Up to follow-up i.e. 2-4 weeks after the last study visit (due to early termination the last visit was on 19 July 2011)

InterventionParticipants (Count of Participants)
GSK2248761 100 mg0
GSK2248761 200 mg2
ETV 200 mg0

[back to top]

Absolute Values of CD4+ Cell Counts After Switch of GSK2248761

CD4 is a receptor for the HIV virus. Most of the damage to an AIDS participant's immune system was done by the virus' destruction of CD4+ lymphocytes. Absolute values of CD4+ cell counts after Switch of GSK2248761 has been presented. (NCT01199731)
Timeframe: Baseline (Day 1) and post switch Week 1, 2, 4, withdrawal and Follow-up Week 4, 8, 12

InterventionCells per cubic millimeter (cells/mm^3) (Mean)
BaselineBaseline switchWeek 1 post switchWeek 2 post switchWeek 4 post switchWithdrawalWeek 4 follow-upWeek 8 follow-up
GSK2248761 100 mg229.2311.0253.0263.7471.5254.6289.0277.5

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 48

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 48

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

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 8

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 8

Interventionlog10 Copies/mL (Median)
ETR + DRV/Rtv-2.04

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 24

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 24

Interventionlog10 Copies/mL (Median)
ETR + DRV/Rtv-2.17

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 20

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 20

Interventionlog10 Copies/mL (Median)
ETR + DRV/Rtv-2.27

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 16

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

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

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 12

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 12

Interventionlog10 Copies/mL (Median)
ETR + DRV/Rtv-2.18

[back to top]

Change From Baseline in Cluster of Differentiation 4 (CD4+) and Cluster of Differentiation 8 (CD8+) Cell Counts at Week 48

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 48

Interventioncells/uL (Mean)
CD4CD8
ETR + DRV/Rtv168-69

[back to top]

Time to Reach First Confirmed Virologic Response

CVR is defined as confirmed plasma Viral Load of less than 50 human immunodeficiency virus - type 1 (HIV-1) ribonucleic acid (RNA) copies/mL. (NCT01199939)
Timeframe: Baseline (Day 1) to Week 48

InterventionDays (Mean)
ETR + DRV/Rtv82.98

[back to top]

Number of Participants With Virologic Failure

Virologic Failure is defined as participant who is a rebounder or a non-responder. Rebounder participant is defined as a participant who is still in the study at Week 12 and first achieves 2 consecutive virologic responses (<50 copies/mL) followed by 2 consecutive non-responses or a discontinued participant (any reason) for which the last observed time point shows a non-response. Non responder participant is defined as a participant who is still in the study at Week 12 and never achieves 2 consecutive responses. (NCT01199939)
Timeframe: Baseline (Day 1) to Week 48

InterventionParticipants (Number)
ETR + DRV/Rtv7

[back to top]

Number of Participants With Confirmed Virologic Response (CVR) at Week 48

CVR is defined as confirmed plasma Viral Load of less than 50 human immunodeficiency virus - type 1 (HIV-1) ribonucleic acid (RNA) copies/mL. (NCT01199939)
Timeframe: Week 48

InterventionParticipants (Number)
ETR + DRV/Rtv40

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 36

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 36

Interventionlog10 Copies/mL (Median)
ETR + DRV/Rtv-2.24

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 4

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 4

Interventionlog10 Copies/mL (Median)
ETR + DRV/Rtv-1.77

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 42

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 42

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

[back to top]

Change From Baseline in Log10 Plasma Human Immunodeficiency Virus - Type 1 (HIV-1) Viral Load at Week 30

(NCT01199939)
Timeframe: Baseline (Day 1) and Week 30

Interventionlog10 Copies/mL (Median)
ETR + DRV/Rtv-2.28

[back to top]

Number of Participants With Plasma HIV-1 RNA Level <50 Copies/mL up to Week 208

Number of participants with HIV-1 RNA level <50 copies/mL plasma was summarized at last visit. Abbott RealTime HIV-1 assay was used to measure the HIV-1 RNA level. (NCT01254656)
Timeframe: Up to Week 208

InterventionParticipants (Number)
Lersivirine (LRV) 500 mg39
LRV 750 mg38
Efavirenz (EFV) 600 mg13

[back to top]

Number of Participants With Plasma Human Immunodeficiency Virus - 1 (HIV-1) Ribonucleic Acid (RNA) Level <50 Copies/mL at 144 Weeks From Day 1 of the Parent Protocol

Number of participants with HIV-1 RNA level <50 copies/mL plasma was summarized at 48 weeks i.e. 144 weeks from Day 1 of the parent protocol. Roche Amplicor HIV-1 Monitor assay was used to measure the HIV-1 RNA level. (NCT01254656)
Timeframe: 144 Weeks from Day 1 of the parent protocol

InterventionParticipants (Number)
Lersivirine (LRV) 500 mg40
LRV 750 mg40
Efavirenz (EFV) 600 mg16

[back to top]

Virology Analysis Participant Accountability From Week 96 Through Study Termination

"Virology analysis included virus susceptibility (phenotype and genotype)to a standard panel of approved antiretrovirals as determined by the Monogram Biosciences PhenoSense GT assay. Below analysis table included the following parameters: 1. protocol-defined treatment failure was defined as an increase in HIV-1 RNA to detectable levels (≥50 copies/mL) on 2 consecutive measurements, the second measurement taken no more than 14 days after the first measurement); 2. Treatment failure: treatment failure (both virologic and non-virologic) was defined as a subject who met the protocol-defined treatment failure criterion or discontinued from the study; 3. NRTI or NNRTI resistance mutations: nucleoside reverse transcriptase inhibitor or lersivirine-associated resistance-associated mutations (RAM) based on the International AIDS Society-USA (IAS-USA) RAM guidelines; 4. 'with result' meant an analyzed sample returned genotypic result or phenotypic result or both." (NCT01254656)
Timeframe: Week 96 through study termination

,,
InterventionParticipants (Number)
Treatment failure (TF)Protocol defined treatment failureTF analysed - HIV-1 RNA >500 c/mLTF analysed - HIV-1 RNA >500 c/mL with resultTF analysed - HIV-1 RNA <= 500 c/mLTF analysed - HIV-1 RNA <= 500 c/mL with resultNRTI or NNRTI resistance mutations
Efavirenz (EFV) 600 mg5000200
Lersivirine (LRV) 500 mg8411520
LRV 750 mg5300210

[back to top]

Change From Baseline in CD4+ Lymphocyte Counts (Absolute) at 144 Weeks From Day 1 of the Parent Protocol

Participant's immunological status assessed by CD4+ lymphocyte count (absolute and percentage) at 48 weeks (ie, 144 weeks from Day 1 of the parent protocol) (NCT01254656)
Timeframe: 144 Weeks from Day 1 of the parent protocol

Interventioncells/uL (Mean)
Lersivirine (LRV) 500 mg293
LRV 750 mg302
Efavirenz (EFV) 600 mg303

[back to top]

Change From Baseline in CD4+ Lymphocyte Counts (Absolute) at 192 Weeks From Day 1 of the Parent Protocol

Participant's immunological status assessed by CD4+ lymphocyte count (absolute and percentage) at 192 weeks from Day 1 of the parent protocol. (NCT01254656)
Timeframe: 192 Weeks from Day 1 of the parent protocol

Interventioncells/uL (Mean)
Lersivirine (LRV) 500 mg308
LRV 750 mg304
Efavirenz (EFV) 600 mg300

[back to top]

Change From Baseline in CD4+ Lymphocyte Counts (Percentage) at 144 Weeks From Day 1 of the Parent Protocol

Participant's immunological status assessed by CD4+ lymphocyte count (absolute and percentage) at 48 weeks (ie, 144 weeks from Day 1 of the parent protocol) (NCT01254656)
Timeframe: 144 Weeks from Day 1 of the parent protocol

InterventionPercentage (Mean)
Lersivirine (LRV) 500 mg13
LRV 750 mg13
Efavirenz (EFV) 600 mg13

[back to top]

Change From Baseline in CD4+ Lymphocyte Counts (Percentage) at 192 Weeks From Day 1 of the Parent Protocol

Participant's immunological status assessed by CD4+ lymphocyte count (absolute and percentage) at 192 weeks from Day 1 of the parent protocol. (NCT01254656)
Timeframe: 192 Weeks from Day 1 of the parent protocol

InterventionPercentage (Mean)
Lersivirine (LRV) 500 mg13
LRV 750 mg15
Efavirenz (EFV) 600 mg12

[back to top]

Etravirine Cmin Pharmacokinetics Coadministered With Boceprevir

Determine etravirine Cmin when coadministered with boceprevir. [Ratio = etravirine administered with boceprevir / etravirine administered alone] (NCT01427504)
Timeframe: Pre-dose, 1, 2, 3, 4, 5, 6, 8, 10, and 12 hours post-dose on day 11-14

InterventionRatio (Geometric Mean)
Etravirine Cmin Coadministered With Boceprevir0.71

[back to top]

Boceprevir AUC Pharmacokinetics

Determine boceprevir area-under-the concentration time curve (AUC) when administered alone. (NCT01427504)
Timeframe: Pre-dose and, 1, 2, 3, 4, 5, 6, and 8 hours post dose on day 11-14

Interventionng*hr/mL (Geometric Mean)
Boceprevir AUC4601

[back to top]

Boceprevir AUC Pharmacokinetics Coadministered With Etravirine

Determine boceprevir AUC when coadministered with etravirine. [Ratio = boceprevir administered with etravirine/ boceprevir alone] (NCT01427504)
Timeframe: Pre-dose and, 1, 2, 3, 4, 5, 6, and 8 hours post dose on day 11-14

InterventionRatio (Geometric Mean)
Boceprevir AUC Coadministered With Etravirine1.10

[back to top]

Boceprevir C8 Pharmacokinetics

Determine boceprevir 8 hour concentration when administered alone. (NCT01427504)
Timeframe: Pre-dose and, 1, 2, 3, 4, 5, 6, and 8 hours post dose on day 11-14

Interventionng/mL (Geometric Mean)
Boceprevir C8106

[back to top]

Boceprevir C8 Pharmacokinetics Coadministered With Etravirine

Determine boceprevir 8 hour concentration when coadministered with etravirine. [Ratio = boceprevir administered with etravirine / boceprevir administered alone] (NCT01427504)
Timeframe: Pre-dose, 1, 2, 3, 4, 5, 6, and 8 hours post dose on day 11-14

InterventionRatio (Geometric Mean)
Boceprevir C8 Coadministered With Etravirine0.88

[back to top]

Boceprevir Cmax Pharmacokinetics

Determine the Cmax of boceprevir when administered alone. (NCT01427504)
Timeframe: Pre-dose and, 1, 2, 3, 4, 5, 6, and 8 hours post dose on day 11-14

Interventionng/mL (Geometric Mean)
Boceprevir Cmax1423

[back to top]

Boceprevir Cmax Pharmacokinetics Coadministered With Etravirine

Determine boceprevir Cmax when coadministered with etravirine. [Ratio = boceprevir administered with etravirine / boceprevir alone] (NCT01427504)
Timeframe: Pre-dose and, 1, 2, 3, 4, 5, 6, and 8 hours post dose on day 11-14

InterventionRatio (Geometric Mean)
Boceprevir Cmax Coadministered With Etravirine1.10

[back to top]

Etravirine AUC Pharmacokinetics

Determine etravirine area under the concentration vs. time curve (AUC)when administered alone. (NCT01427504)
Timeframe: Pre-dose and, 1, 2, 3, 4, 5, 6, 8, 10 and 12 hours post dose on day 11-14

Interventionng*hr/mL (Geometric Mean)
Etravirine AUC7698

[back to top]

Etravirine AUC Pharmacokinetics Coadministered With Boceprevir

Determine etravirine AUC when coadministered with boceprevir. [Ratio = Etravirine administered with bocepreivr / etravirine administered alone] (NCT01427504)
Timeframe: Pre-dose, 1, 2, 3, 4, 5, 6, 8, 10, and 12 hours Post-dose on day 11-14

InterventionRatio (Geometric Mean)
Etravirine AUC Coadministered With Boceprevir0.77

[back to top]

Etravirine Cmax Pharmacokinetics

Determine etravirine Cmax when administered alone (NCT01427504)
Timeframe: Pre-dose and, 1, 2, 3, 4, 5, 6, 8, 10 and 12 hours post dose on day 11-14

Interventionng/mL (Geometric Mean)
Etravirine Cmax900

[back to top]

Etravirine Cmax Pharmacokinetics Coadministered With Boceprevir

Determine etravirine Cmax when coadministered with boceprevir. [Ratio = etravirine administered with boceprevir / etravirine administered alone] (NCT01427504)
Timeframe: Pre-dose, 1, 2, 3, 4, 5, 6, 8, 10, and 12 hours post-dose on day 11-14

InterventionRatio (Geometric Mean)
Etravirine Cmax0.76

[back to top]

Etravirine Cmin Pharmacokinetics

Determine etravirine Cmin when administered alone (NCT01427504)
Timeframe: Pre-dose and, 1, 2, 3, 4, 5, 6, 8, 10 and 12 hours post dose on day 11-14

Interventionng/mL (Geometric Mean)
Etravirine Cmin439

[back to top]

Area Under the Plasma Concentration-Time Curve Over 12 Hours of ETR

Geometric Mean (Standard Deviation) of the area under the plasma concentration-time curve over 12 hours (AUC12h) of ETR. (NCT01504841)
Timeframe: Pre-dose, 1, 2, 4, 6, 9, and 12 hours post-dose measured at intensive PK visit (within 7-10 days after last dose of study drug administration)

Interventionng*h/mL (Geometric Mean)
Cohort I: Treatment Experienced, 2 to 6 Years of Age5512.85
Cohort II: Treatment Experienced, 1 to 2 Years of Age4821.76

[back to top]

Decline in Absolute CD4 Percent of Greater Than 5 Percent Any Time After 12 Weeks of Therapy

Number (%) of participants with a >5% decline in absolute CD4 percent from baseline at weeks 12, 24, and 48, by Cohort, including Clopper-Pearson confidence intervals. (NCT01504841)
Timeframe: Measured at baseline and at Weeks 12, 24, and 48

InterventionParticipants (Count of Participants)
Week 1272045667Week 1272045668Week 2472045667Week 2472045668Week 4872045667Week 4872045668
>5% decline in CD4 % from baselineIncrease or <5% decline in CD4 % from baseline
Cohort I: Treatment Experienced, 2 to 6 Years of Age1
Cohort II: Treatment Experienced, 1 to 2 Years of Age2
Cohort I: Treatment Experienced, 2 to 6 Years of Age10
Cohort II: Treatment Experienced, 1 to 2 Years of Age1
Cohort II: Treatment Experienced, 1 to 2 Years of Age3
Cohort I: Treatment Experienced, 2 to 6 Years of Age2
Cohort I: Treatment Experienced, 2 to 6 Years of Age9

[back to top]

HIV-1 RNA Virologic Failure Status at Weeks 24 and 48

Number (%) of participants with confirmed Virologic Failure, defined as: failure to suppress plasma HIV-1 RNA to fewer than 400 copies/ml and failure to achieve at least a 2-log10 reduction (from baseline) in HIV-1 RNA at Weeks 24 or 48, by Cohort, with Clopper-Pearson confidence intervals. The initial HIV-1 RNA results that met the Virologic Failure definition were each confirmed by a second result obtained within 1 to 4 weeks of the initial result obtained at Week 24 and/or 48. (NCT01504841)
Timeframe: Baseline, Week 24, and Week 48

InterventionParticipants (Count of Participants)
Week 2472045667Week 2472045668Week 4872045667Week 4872045668
Virologic FailureNo Virologic Failure
Cohort I: Treatment Experienced, 2 to 6 Years of Age2
Cohort I: Treatment Experienced, 2 to 6 Years of Age9
Cohort I: Treatment Experienced, 2 to 6 Years of Age3
Cohort II: Treatment Experienced, 1 to 2 Years of Age3
Cohort I: Treatment Experienced, 2 to 6 Years of Age8
Cohort II: Treatment Experienced, 1 to 2 Years of Age1

[back to top]

Peak Concentration of Etravirine in Breast Milk

Cmax ng/mL (NCT01625169)
Timeframe: day 14

Interventionng/ml (Mean)
HIV + Pregnant Women1805.4

[back to top]

HIV Viral Load in Breast Milk and Plasma

Positive HIV RNA in breast milk and plasma- LDL 40 copies/ml (NCT01625169)
Timeframe: Day 5

Interventionparticipants (Number)
Breast milkPlasma
HIV + Pregnant Women01

[back to top]

Peak Plasma Concentration of Etravirine in Plasma

"Cmax ng/ml~Note: One participant did not complete the Day 5 evaluation." (NCT01625169)
Timeframe: Day 5

Interventionng/ml (Mean)
HIV + Pregnant Women497.6

[back to top]

Peak Plasma Concentration of Etravirine in Plasma

Cmax ng/mL (NCT01625169)
Timeframe: day 14

Interventionng/ml (Mean)
HIV + Pregnant Women564.6

[back to top]

HIV Viral Load in Breast Milk and Plasma

Positive HIV RNA in breast milk and plasma- LDL 40 copies/ml (NCT01625169)
Timeframe: Day 14

Interventionparticipants (Number)
Breast milkPlasma
HIV + Pregnant Women20

[back to top]

Area Under the Curve (AUC) 0-12 for Breast Milk

AUC 0-12 ng*hr/ml (NCT01625169)
Timeframe: Day 14

Interventionng*hr/ml (Mean)
HIV + Pregnant Women12953.7

[back to top]

Area Under the Curve (AUC) 0-12 for Breast Milk

AUC 0-12 ng*hr/ml (NCT01625169)
Timeframe: Day 5

Interventionng*hr/ml (Mean)
HIV + Pregnant Women4371.9

[back to top]

Area Under the Curve (AUC) 0-12 for Plasma

AUC 0-12 ng*hr/ml (NCT01625169)
Timeframe: Day 14: 0, 2,4, 8 and 24 hours post dose

Interventionng*hr/ml (Mean)
HIV + Pregnant Women4096

[back to top]

Area Under the Curve (AUC) 0-12 for Plasma

AUC 0-12 ng*hr/ml (NCT01625169)
Timeframe: Day 5: 0, 2,4, 8 and 24 hours post dose

Interventionng*hr/ml (Mean)
HIV + Pregnant Women3622.1

[back to top]

Peak Concentration of Etravirine in Breast Milk

"Cmax ng/ml~Note: One participant did not complete the Day 5 evaluation." (NCT01625169)
Timeframe: day 5

Interventionng/ml (Mean)
HIV + Pregnant Women685.4

[back to top]

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

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

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

[back to top]

Change From Baseline in Fasting Values of Triglycerides

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

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

[back to top]

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

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

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

[back to top]

Change From Baseline in Fasting Values of Total Cholesterol

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

Change From Baseline in Fasting Values of Glucose

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

Change From Baseline in CD4+ T-cell Count

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

Percent of Participants With Treatment Modification or Discontinuation by Week 48

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

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

[back to top]

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

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

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

[back to top]

Percent of Participants With Death or Hospitalization by Week 48

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

Percent of Participants With Confirmed Virologic Failure by Week 48

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

Percent of Participants Experiencing Death by Week 48

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

Number of Weeks of Follow-up

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

Time From Study Entry/Randomization to Death

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

Percentage of Participants With Detectable Seminal HIV-RNA Viral Load at Week 48

• Assessment of HIV-RNA viral load in human male genital compartment (20 patients) at week 48 (NCT02212379)
Timeframe: week 48

InterventionParticipants (Count of Participants)
Raltegravir and Etravirine1

[back to top]

Median Time of Virological Failure

Time between the date of the study treatment initiation and the date of virological failure (NCT02212379)
Timeframe: week 96

Interventionweeks (Median)
Raltegravir and Etravirine96

[back to top]

Number of Patients With RAL and/or ETR Resistance Mutations Among Those With Virological Failure

(NCT02212379)
Timeframe: week 96

Interventionparticipants (Number)
Raltegravir and Etravirine1

[back to top]

Percent Change of Renal Function

Percent change of the estimated Glomerular Filtration Rate (eGFR) calculated using the CKD-EPI (Chronic Kidney Disease Epidemiology Collaboration Calculator) formula (NCT02212379)
Timeframe: from day 0 to week 96

InterventionMedian percent change, as median (IQR) (Median)
Raltegravir and Etravirine-0.6

[back to top]

Evolution of Body Fat Distribution From Day 0 to W96 (DXA Scan Sub-study, 80 Patients)

Evolution of total fat mass, limb fat and trunk fat from day 0 to week 96 (NCT02212379)
Timeframe: from day 0 to week 96

Interventionpercentage of change (Median)
Total fat mass, KgLimb fat, KgTrunk fat, Kg
Raltegravir and Etravirine12.211.612.2

[back to top]

Evolution of CD4+, CD8+ T Cells Counts and CD4/CD8 Ratio

(NCT02212379)
Timeframe: from day 0 to week 48 and week 96

Interventionpercentage of change (Median)
CD4: 0-48 weekCD8: 0-48 weekCD4/CD8: 0-48 weekCD4: 0-96 weekCD8: 0-96 weekCD4/CD8: 0-96 week
Raltegravir and Etravirine1.1-1.85.75.0-5.27.4

[back to top]

Evolution of Metabolic Parameters (Fasting Triglycerides, Total Cholesterol, HDL-cholesterol, LDL-cholesterol and Fasting Glycemia)

(NCT02212379)
Timeframe: from day 0 to week 96

Interventionpercentage of change (Median)
fasting triglyceridestotal cholesterolHDL-cholesterolLDL-cholesterolfasting glycemia
Raltegravir and Etravirine-18.8-0.55.4-4.30

[back to top]

Evolution of the Calibrated 5-year Framingham Risk Score

"The Framingham risk score is expressed as a percentage. Higher scores mean a worse outcome and lower scores mean better outcome.~Median percent change expressed as median (interquartile range (IQR))" (NCT02212379)
Timeframe: from day 0 to week 48 and at week 96

InterventionMedian percent change as median (IQR) (Median)
from D0 to week 48from D0 to week 96
Raltegravir and Etravirine1.09.7

[back to top]

Evolution of the Level of MCP1 From D0 to W48 on Frozen Samples

(NCT02212379)
Timeframe: from day 0, to week 48

InterventionPercentage change (Median)
Premenopausal with mesurable AMHPremenopausal with reduced ovarian reservePost-menopausal
Raltegravir and Etravirine3.13.5-2.4

[back to top]

Evolution of the Ovarian Reserve From D0 to W48 Measured by AMH on Frozen Aliquots

We measured the Anti-mullerian Hormone (AMH) level to evaluate the ovarian reserve (from D0 to W48) (NCT02212379)
Timeframe: from day 0, to week 48

Interventionng/mL (Median)
Reproductive activity: D0Pre-menopausal: D0Post-menopausal: D0Reproductive activity: W48Pre-menopausal: W48Post-menopausal: W48
Raltegravir and Etravirine0.1720.0090.0090.1520.0080.005

[back to top]

Evolution of Total Cell-associated HIV-DNA

(NCT02212379)
Timeframe: from day 0 to week 48 and week 96

Interventionpercentage of change (Median)
Change from baseline to week 48Change from baseline to week 96
Raltegravir and Etravirine00

[back to top]

Factors Associated With the Occurrence of Plasma HIV-RNA Viral Load > 50 Copies/mL

(NCT02212379)
Timeframe: week 96

Interventionhazard ratio (Number)
Age >60 years>=2 glasses/day alcohol consumption
Raltegravir and Etravirine3.711.3

[back to top]

Inflammatory Parameters

• Evolution of the inflammation markers (IL-6hs, sCD14, sCD163, D-Dimers, IP-10, IgG, CRPus and insulin) on frozen plasma aliquots (NCT02212379)
Timeframe: from day 0 to week 96

Interventionpercentage of change (Median)
IL-6IP-10sCD163sCD14IgGhsCRPD-DimerInsulin
Raltegravir and Etravirine0.8-8.10.7-270016.54.6

[back to top]

Number of Participants Experiencing Adverse Events and Effects

Number of all clinical and biological adverse events effects. Number of grade 3 or 4 clinical and biological adverse events and effects. (NCT02212379)
Timeframe: From day 0 to week 48 and week 96

InterventionParticipants (Count of Participants)
Any AE: 0-48 weekGrade 3 or 4 AE: 0-48 weekAny AE : 48-96Grade 3 or 4 AE: 48-96
Raltegravir and Etravirine1541510811

[back to top]

Percentage of Participants Compliant With Treatment Program.

The compliance rate was estimated as the number of pills consumed (recorded using the self-reported 90 questionnaire) divided by the number of pills theoretically consumed, classified as low (80%), medium (80%-95%) or high (95%). (NCT02212379)
Timeframe: at week 0, week 48, and week 96

InterventionParticipants (Count of Participants)
low (<80) at W0medium (80-95) at W0high (>95) at W0low (<80) at W48medium (80-95) at W48high (>95) at W48low (<80) at W96medium (80-95) at W96high (>95) at W96
Raltegravir and Etravirine133139716123129115

[back to top]

Percentage of Participants Reporting a Very Good or an Excellent Quality of Life at Day 0, Weeks 48 and 96

(NCT02212379)
Timeframe: day 0 and weeks 48 and 96

InterventionParticipants (Count of Participants)
day 0week 48week 96
Raltegravir and Etravirine637877

[back to top]

Percentage of Participants With Successful Virological Suppression at Weeks 48 and 96

"Virological success is defined as the absence of 2 consecutive plasma viral loads (VL) > 50 copies/mL within 2 to 4 weeks of a dual raltegravir/etravirine regimen.~The proportion of patients who maintained viral suppression under raltegravir plus etravirine was 99.4% (95% confidence interval (95% CI:95.6 -99.9) at week 48 and 98.7% (95% CI: 95.0 -99.7) at week 96" (NCT02212379)
Timeframe: at week48 and at week 96

Interventionpercentage of participant (Number)
at week 48at week 96
Raltegravir and Etravirine99.498.7

[back to top]

Percentage of Patients With High Grade of Virological Failure Defined as HIV RNA > 200 Copies/mL

(NCT02212379)
Timeframe: weeks 48 and 96

Interventionpercentage of participants (Mean)
week 48week 96
Raltegravir and Etravirine0.60

[back to top]

Percentage of Patients With Therapeutic Success at Week 48 and Week 96

Therapeutic success was defined as the absence of virological failure (i.e. 2 consecutive plasma viral loads (VL) > 50 copies/mL within 2 to 4 weeks) and the absence of treatment interruption due to adverse event judged by DSMB as related to the study treatment or procedure (NCT02212379)
Timeframe: weeks 48 and 96

Interventionpercentage of participants (Number)
at week 48at week 96
Raltegravir and Etravirine95.192.7

[back to top]

Percentage of Patients With Trial Treatment Interruption at Week 48 and Week 96

(NCT02212379)
Timeframe: weeks 48 and 96

Interventionpercentage of participant (Number)
at week 48at week 96
Raltegravir and Etravirine4.36.1

[back to top]

Percentage of Patients With With Grade Virological Failure (HIV-RNA Plasma VL Between 51 and 200 Copies/mL)

(NCT02212379)
Timeframe: weeks 48 and 96

Interventionpercentage of participants (Number)
at week 48at week 96
Raltegravir and Etravirine00.6

[back to top]

Sub-study in Women : Comparison of the Evolution Fat Distribution Measured by DXA Scan and Body Weight According to AMH Status

BMI, Hip circumference, Waist circumference, waist/hip ratio, Limb fat, Trunk fat, Total fat, Limb lean, Trunk lean, and Total lean (NCT02212379)
Timeframe: from day 0, to week 96

,,
InterventionPercentage of change (Median)
Body mass index (BMI)Hip circumferenceWaist circumferenceWaist/hip ratioLimb fatTrunk fatTotal fatLimb leanTrunk leanTotal lean
Postmenopausal2.073.096.521.8510.8921.2724.39-1.44-3.24-3.01
Premenopausal With Mesurable AMH-0.967.062.74-3.43-2.94-6.90-2.59-0.70-0.79-1.35
Premenopausal With Reduced Ovarian Reserve5.692.804.200.026.7418.8016.772.135.535.83

[back to top]

Sub-study in Women : Comparison of the Metabolic/Inflammatory Profile in Women According to Their Ovarian Reserve and Menopausal Status at D0 and Its Evolution up to Week 96

"Metabolic markers measures are total cholesterol, LDL-cholesterol, HDL-cholesterol, triglycerides.~Inflammatory and innate immune activation markers measures are: IL-6hs, sCD14, sCD163, D-Dimers, IP-10, IgG, hsCRP and Insulin.~Ovarian reserve measure is AMH" (NCT02212379)
Timeframe: from day 0, to week 96

,,
InterventionPercentage of change (Median)
Total cholesterolLDL-cholesterolHDL-cholesterolTriglyceridessCD14sCD163hsCRPD-dimersIgGIL-6hsIP-10Insulin
Postmenopausal-11.35-6.58-4.84-0.99-18.84.8-31.423.1-1.2-0.9-5.411.4
Premenopausal With Mesurable AMH6.625.5218.475.59-31.610.85.76.80.75.9-18.3-29.2
Premenopausal With Reduced Ovarian Reserve6.915.6227.74-38.37-32.815.931.834.81.8-49.98.230.3

[back to top]

Sub-study: Bone Mineral Density

"• Evolution of bone mineral density (BMD) measured by DXA scans (DXA scan sub-study, 81 patients)~Lumbar spine BMD, mg/cm2~Total hip BMD, mg/cm2" (NCT02212379)
Timeframe: from day 0, to week 48 and week 96

Interventionpercentage of change (Median)
Lumbar spine BMD from D0 to week 48Lumbar spine BMD at week 96Total hip BMD from D0 to week 48Total hip BMD at week 96
Raltegravir and Etravirine0.7-1.00.60

[back to top]

Cohort 1: Tmax of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 3

InterventionHours (Median)
Cohort 1: GSK3640254 200 mg4.000
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg4.000

[back to top]

Cohort 1: Time of Maximum Observed Concentration (Tmax) of DRV

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionHours (Median)
Cohort 1: DRV/RTV 600/100 mg3.000
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg3.000

[back to top]

Cohort 1: Plasma Concentration at the End of the Dosing Interval (Ctau) of DRV

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 1: DRV/RTV 600/100 mg2.957
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg2.637

[back to top]

Cohort 1: Maximum Observed Concentration (Cmax) of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 1: GSK3640254 200 mg1.752
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg1.863

[back to top]

Cohort 1: Ctau of RTV

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 1: DRV/RTV 600/100 mg0.3194
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg0.3314

[back to top]

Cohort 1: Ctau of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 1: GSK3640254 200 mg0.8152
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg0.9530

[back to top]

Cohort 1: Cmax of RTV

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 1: DRV/RTV 600/100 mg1.178
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg1.306

[back to top]

Cohort 1: Cmax of DRV

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 1: DRV/RTV 600/100 mg7.037
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg7.268

[back to top]

Cohort 1: AUC(0-tau) of RTV

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionHours* micrograms per milliliter (Geometric Mean)
Cohort 1: DRV/RTV 600/100 mg7.303
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg7.790

[back to top]

Cohort 1: AUC(0-tau) of DRV

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionHours*micrograms per milliliter (Geometric Mean)
Cohort 1: DRV/RTV 600/100 mg57.47
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg53.37

[back to top]

Cohort 1: Area Under the Plasma Concentration-time Curve From Time Zero to the End of the Dosing Interval at Steady State (AUC[0-tau]) of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 3

InterventionHours*micrograms per milliliter (Geometric Mean)
Cohort 1: GSK3640254 200 mg27.03
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg30.70

[back to top]

Cohort 3: Number of Participants With Vital Sign Values of PCI Criteria

Vital signs including SBP, DBP and pulse rate were measured in a supine position after atleast 5 minutes of rest. The PCI ranges for vitals were as follows; for SBP <85 or >140 mmHg, for DBP <45 or >90 mmHg, for pulse rate <40 or >100 beats per minute. The number of participants with vital signs of PCI were presented. (NCT04630002)
Timeframe: Up to Day 26

,
InterventionParticipants (Count of Participants)
SBPDBPPulse rate
Cohort 3: GSK3640254 200 mg100
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg000

[back to top]

Cohort 3: Number of Participants With SAEs and Non-SAEs

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study intervention, whether or not considered related to the study intervention. An SAE is defined as any untoward medical occurrence that, at any dose that results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect or other situations as per medical or scientific judgment. Adverse events which were not Serious were considered as Non-Serious adverse events. (NCT04630002)
Timeframe: Up to Day 26

,
InterventionParticipants (Count of Participants)
SAEnon-SAE
Cohort 3: GSK3640254 200 mg01
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg08

[back to top]

Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Urinalysis Parameters

Urine samples were collected for urinalysis parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Erythrocytes High, Grade 3: Gross, with or without clots OR with RBC casts OR intervention indicated, Grade 4: Life-threatening consequences; Glucose High, Grade 3: >2+ (proportionate concentration by dipstick test) or >500 mg, Grade 4: >500 mg; Protein High, Grade 3: 3+ (proportionate concentration by dipstick test) or higher, Grade 4: Not Applicable. Baseline was defined as the latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 26

,
InterventionParticipants (Count of Participants)
Erythrocytes, High, Increase to Grade 3Erythrocytes, High, Increase to Grade 4Glucose, High, Increase to Grade 3Glucose, High, Increase to Grade 4Protein, High, Increase to Grade 3Protein, High, Increase to Grade 4
Cohort 3: GSK3640254 200 mg000000
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg000000

[back to top]

Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Hematology Parameters

Blood samples were collected for analysis of hematology parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Hemoglobin Low, Grade 3: 7.0 to <9.0 g/dL (males) and 6.5 to <8.5 g/dL (females),Grade 4: <7.0 g/dL (males) and <6.5 g/dL (females); Leukocytes Low, Grade 3: 1000 to 1499 cells/mm^3,Grade 4: <1000 cells/mm^3; Lymphocytes Low, Grade 3: 350 to <500 cells/L,Grade 4: <350 cells/L; Neutrophils Low, Grade 3: 400 to 599 cells/mm^3, Grade 4: <400 cells/mm^3; Platelets Low, Grade 3: 25,000 to <50,000 cells/mm^3, Grade 4: <25,000 cells/mm^3. Baseline was defined as latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 26

,
InterventionParticipants (Count of Participants)
Hemoglobin, Low, Increase to Grade 3Hemoglobin, Low, Increase to Grade 4Leukocytes, Low, Increase to Grade 3Leukocytes, Low, Increase to Grade 4Lymphocytes, Low, Increase to Grade 3Lymphocytes, Low, Increase to Grade 4Neutrophils, Low, Increase to Grade 3Neutrophils, Low, Increase to Grade 4Platelets, Low, Increase to Grade 3Platelets, Low, Increase to Grade 4
Cohort 3: GSK3640254 200 mg0000000000
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg0000000000

[back to top]

Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Glucose, Triglycerides, Lipase, Urate and Cholesterol

Blood samples were collected for analysis of clinical chemistry parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Glucose High, Grade 3: >250 to 500 mg/dL, Grade 4: >=500 mg/dL, Glucose Low, Grade 3: 30 to<40 mg/dL, Grade 4:<30 mg/dL; Triglycerides High, Grade 3: >500 to <1.000 mg/dL, Grade 4:>1000 mg/dL; Lipase High, Grade 3: 3.0 to <5.0×ULN, Grade 4:>=5.0×ULN; Urate High, Grade 3: 12.0 to <15.0 mEq/L, Grade 4:>=15.0 mEq/L; Cholesterol High, Grade 3: >=300 mg/dL, Grade 4: Not Applicable. Baseline was defined as the latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 26

,
InterventionParticipants (Count of Participants)
Glucose, Low, Increase to Grade 3Glucose, Low, Increase to Grade 4Glucose, High, Increase to Grade 3Glucose, High, Increase to Grade 4Triglycerides, High, Increase to Grade 3Triglycerides, High, Increase to Grade 4Lipase, High, Increase to Grade 3Lipase, High, Increase to Grade 4Urate, High, Increase to Grade 3Urate, High, Increase to Grade 4Cholesterol, High, Increase to Grade 3Cholesterol, High, Increase to Grade 4
Cohort 3: GSK3640254 200 mg000000000000
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg000000000000

[back to top]

Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Calcium, Creatine Kinase, Creatinine, Phosphate, Potassium and Sodium

Blood samples were collected for analysis of clinical chemistry parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Calcium High, Grade 3: 12.5 to <13.5 mg/dL, Grade 4: >=13.5 mg/dL; Calcium Low, Grade 3: 6.1 to <7.0 mg/dL, Grade 4: <6.1 mg/dL; Creatine Kinase High, Grade 3: 10 to <20 × ULN, Grade 4: >=20 × ULN; Creatinine High, Grade 3: >1.8 to <3.5 ULN, Grade 4: >=3.5 × ULN; Phosphate Low, Grade 3: 1.0 to <1.4 mg/dL, Grade 4: <1.0 mg/dL; Potassium High, Grade 3: 6.5 to <7.0 mEq/L,Grade 4: >=7.0 mEq/L; Potassium Low, Grade 3: 2.0 to <2.5 mEq/L, Grade 4: <2.00 mEq/L; Sodium High, Grade 3: 154 to <160 mEq/L, Grade 4:>=160 mEq/L; Sodium Low, Grade 3: 121 to <125 mEq/L, Grade 4:<=120 mEq/L. Baseline was defined as the latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 26

,
InterventionParticipants (Count of Participants)
Calcium, Low, Increase to Grade 3Calcium, Low, Increase to Grade 4Calcium, High, Increase to Grade 3Calcium, High, Increase to Grade 4Creatine Kinase, High, Increase to Grade 3Creatine Kinase, High, Increase to Grade 4Creatinine, High, Increase to Grade 3Creatinine, High, Increase to Grade 4Phosphate, Low, Increase to Grade 3Phosphate, Low, Increase to Grade 4Potassium, Low, Increase to Grade 3Potassium, Low, Increase to Grade 4Potassium, High, Increase to Grade 3Potassium, High, Increase to Grade 4Sodium, Low, Increase to Grade 3Sodium, Low, Increase to Grade 4Sodium, High, Increase to Grade 3Sodium, High, Increase to Grade 4
Cohort 3: GSK3640254 200 mg000000000000000000
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg000000000000000000

[back to top]

Cohort 3: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Alanine Aminotransferase, Albumin, Alkaline Phosphatase, Amylase, Aspartate Aminotransferase, Bilirubin and Direct Bilirubin

Blood samples were collected for analysis of clinical chemistry parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Alanine Aminotransferase High; Grade 3: 5.0 to <10.0 × ULN, Grade 4: >=10.0 × ULN; Albumin Low, Grade 3: <2.0 g/dL, Grade 4: Not Applicable; Alkaline Phosphatase High, Grade 3: 5.0 to <10.0 × ULN, Grade 4: >=10.0 × ULN; Amylase High, Grade 3: 3.0 to <5.0 × ULN, Grade 4: >=5.0 × ULN; Aspartate Aminotransferase High, Grade 3: 5.0 to <10.0 × ULN, Grade 4: >=10.0 × ULN; Bilirubin High, Grade 3: 2.6 to<5.0 × ULN, Grade 4: >=5.0 × ULN and Direct Bilirubin High, Grade 3: >ULN with other signs and symptoms of hepatotoxicity, Grade 4: >ULN with life-threatening consequences. Baseline was defined as latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 26

,
InterventionParticipants (Count of Participants)
Alanine Aminotransferase, High, Increase to Grade 3Alanine Aminotransferase, High, Increase to Grade 4Albumin, Low, Increase to Grade 3Albumin, Low, Increase to Grade 4Alkaline Phosphatase, High, Increase to Grade 3Alkaline Phosphatase, High, Increase to Grade 4Amylase, High, Increase to Grade 3Amylase, High, Increase to Grade 4Aspartate Aminotransferase, High, Increase to Grade 3Aspartate Aminotransferase, High, Increase to Grade 4Bilirubin, High, Increase to Grade 3Bilirubin, High, Increase to Grade 4Direct Bilirubin, High, Increase to Grade 3Direct Bilirubin, High, Increase to Grade 4
Cohort 3: GSK3640254 200 mg00000000000000
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg00000000000000

[back to top]

Cohort 3: Number of Participants With Clinically Significant Abnormal ECG Findings

A 12-lead ECG was recorded with the participant in a supine position using an automated ECG machine. Clinically significant abnormal findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. Data for number of participants with clinically significant abnormal ECG findings were reported. Data has been presented for the participants with respect to the actual treatment received in respective treatment periods. (NCT04630002)
Timeframe: Day 1 (2,4,6 Hours) in Treatment Period 1; Day 8 (2,4,6 Hours), Day 9 (2,4,6 Hours), Day 26 in Treatment Period 2

InterventionParticipants (Count of Participants)
2 Hours, Day 84 Hours, Day 86 Hours, Day 82 Hours; Day 94 Hours; Day 96 Hours; Day 9Day 26
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg0000000

[back to top]

Cohort 3: Number of Participants With Clinically Significant Abnormal ECG Findings

A 12-lead ECG was recorded with the participant in a supine position using an automated ECG machine. Clinically significant abnormal findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. Data for number of participants with clinically significant abnormal ECG findings were reported. Data has been presented for the participants with respect to the actual treatment received in respective treatment periods. (NCT04630002)
Timeframe: Day 1 (2,4,6 Hours) in Treatment Period 1; Day 8 (2,4,6 Hours), Day 9 (2,4,6 Hours), Day 26 in Treatment Period 2

InterventionParticipants (Count of Participants)
2 Hours, Day 14 Hours, Day 16 Hours, Day 1
Cohort 3: GSK3640254 200 mg000

[back to top]

Cohort 3: Number of Participants With AEs Leading to Discontinuations and Deaths

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study intervention, whether or not considered related to the study intervention. Number of participants with AEs leading to discontinuations and deaths were reported. (NCT04630002)
Timeframe: Up to Day 26

,
InterventionParticipants (Count of Participants)
AEs leading to discontinuationsAES leading to deaths
Cohort 3: GSK3640254 200 mg00
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg10

[back to top]

Cohort 2: Number of Participants With Vital Sign Values of PCI Criteria

Vital signs including SBP, DBP and pulse rate were measured in a supine position after atleast 5 minutes of rest. The PCI ranges for vitals were as follows; for SBP <85 or >140 mmHg, for DBP <45 or >90 mmHg, for pulse rate <40 or >100 beats per minute. The number of participants with vital signs of PCI were presented. (NCT04630002)
Timeframe: Up to Day 36

,,
InterventionParticipants (Count of Participants)
SBPDBPPulse rate
Cohort 2: ETR 200 mg000
Cohort 2: GSK3640254 200 mg000
Cohort 2: GSK3640254 200 mg + ETR 200 mg010

[back to top]

Cohort 2: Number of Participants With SAEs and Non-SAEs

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study intervention, whether or not considered related to the study intervention. An SAE is defined as any untoward medical occurrence that, at any dose that results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect or other situations as per medical or scientific judgment. Adverse events which were not Serious were considered as Non-Serious adverse events. (NCT04630002)
Timeframe: Up to Day 36

,,
InterventionParticipants (Count of Participants)
SAEnon-SAE
Cohort 2: ETR 200 mg04
Cohort 2: GSK3640254 200 mg03
Cohort 2: GSK3640254 200 mg + ETR 200 mg07

[back to top]

Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Urinalysis Parameters

Urine samples were collected for urinalysis parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Erythrocytes High, Grade 3: Gross, with or without clots OR with RBC casts OR intervention indicated, Grade 4: Life-threatening consequences; Glucose High, Grade 3: >2+ (proportionate concentration by dipstick test) or >500 mg, Grade 4: >500 mg; Protein High, Grade 3: 3+ (proportionate concentration by dipstick test) or higher, Grade 4: Not Applicable. Baseline was defined as the latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 36

,,
InterventionParticipants (Count of Participants)
Erythrocytes, High, Increase to Grade 3Erythrocytes, High, Increase to Grade 4Glucose, High, Increase to Grade 3Glucose, High, Increase to Grade 4Protein, High, Increase to Grade 3Protein, High, Increase to Grade 4
Cohort 2: ETR 200 mg000000
Cohort 2: GSK3640254 200 mg000000
Cohort 2: GSK3640254 200 mg + ETR 200 mg000000

[back to top]

Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Hematology Parameters

Blood samples were collected for analysis of hematology parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Hemoglobin Low, Grade 3: 7.0 to <9.0 g/dL (males) and 6.5 to <8.5 g/dL (females),Grade 4: <7.0 g/dL (males) and <6.5 g/dL (females); Leukocytes Low, Grade 3: 1000 to 1499 cells/mm^3,Grade 4: <1000 cells/mm^3; Lymphocytes Low, Grade 3: 350 to <500 cells/L,Grade 4: <350 cells/L; Neutrophils Low, Grade 3: 400 to 599 cells/mm^3, Grade 4: <400 cells/mm^3; Platelets Low, Grade 3: 25,000 to <50,000 cells/mm^3, Grade 4: <25,000 cells/mm^3. Baseline was defined as latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 36

,,
InterventionParticipants (Count of Participants)
Hemoglobin, Low, Increase to Grade 3Hemoglobin, Low, Increase to Grade 4Leukocytes, Low, Increase to Grade 3Leukocytes, Low, Increase to Grade 4Lymphocytes, Low, Increase to Grade 3Lymphocytes, Low, Increase to Grade 4Neutrophils, Low, Increase to Grade 3Neutrophils, Low, Increase to Grade 4Platelets, Low, Increase to Grade 3Platelets, Low, Increase to Grade 4
Cohort 2: ETR 200 mg0000000000
Cohort 2: GSK3640254 200 mg0000000000
Cohort 2: GSK3640254 200 mg + ETR 200 mg0000000000

[back to top]

Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Alanine Aminotransferase, Albumin, Alkaline Phosphatase, Amylase, Aspartate Aminotransferase, Bilirubin and Direct Bilirubin

Blood samples were collected for analysis of clinical chemistry parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Alanine Aminotransferase High; Grade 3: 5.0 to <10.0 × ULN, Grade 4: >=10.0 × ULN; Albumin Low, Grade 3: <2.0 g/dL, Grade 4: Not Applicable; Alkaline Phosphatase High, Grade 3: 5.0 to <10.0 × ULN, Grade 4: >=10.0 × ULN; Amylase High, Grade 3: 3.0 to <5.0 × ULN, Grade 4: >=5.0 × ULN; Aspartate Aminotransferase High, Grade 3: 5.0 to <10.0 × ULN, Grade 4: >=10.0 × ULN; Bilirubin High, Grade 3: 2.6 to<5.0 × ULN, Grade 4: >=5.0 × ULN and Direct Bilirubin High, Grade 3: >ULN with other signs and symptoms of hepatotoxicity, Grade 4: >ULN with life-threatening consequences. Baseline was defined as latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 36

,,
InterventionParticipants (Count of Participants)
Alanine Aminotransferase, High, Increase to Grade 3Alanine Aminotransferase, High, Increase to Grade 4Albumin, Low, Increase to Grade 3Albumin, Low, Increase to Grade 4Alkaline Phosphatase, High, Increase to Grade 3Alkaline Phosphatase, High, Increase to Grade 4Amylase, High, Increase to Grade 3Amylase, High, Increase to Grade 4Aspartate Aminotransferase, High, Increase to Grade 3Aspartate Aminotransferase, High, Increase to Grade 4Bilirubin, High, Increase to Grade 3Bilirubin, High, Increase to Grade 4Direct Bilirubin, High, Increase to Grade 3Direct Bilirubin, High, Increase to Grade 4
Cohort 2: ETR 200 mg00000000000000
Cohort 2: GSK3640254 200 mg00000000000000
Cohort 2: GSK3640254 200 mg + ETR 200 mg00000000000000

[back to top]

Cohort 2: Number of Participants With Clinically Significant Abnormal ECG Findings

A 12-lead ECG was recorded with the participant in a supine position using an automated ECG machine. Clinically significant abnormal findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. Data for number of participants with clinically significant abnormal ECG findings were reported. Data has been presented for the participants with respect to the actual treatment received in respective treatment periods. (NCT04630002)
Timeframe: Day 1 (2,4,6 Hours), Day 7 and Day 11 in Treatment Period 1; Day 12 (2,4,6 Hours), Day 21 in Treatment Period 2; Day 22 (2,4,6 Hours), Day 26 and Day 36 in Treatment Period 3

InterventionParticipants (Count of Participants)
2 Hours, Day 14 Hours, Day 16 Hours; Day 1Day 7Day 11
Cohort 2: GSK3640254 200 mg00000

[back to top]

Cohort 2: Number of Participants With Clinically Significant Abnormal ECG Findings

A 12-lead ECG was recorded with the participant in a supine position using an automated ECG machine. Clinically significant abnormal findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. Data for number of participants with clinically significant abnormal ECG findings were reported. Data has been presented for the participants with respect to the actual treatment received in respective treatment periods. (NCT04630002)
Timeframe: Day 1 (2,4,6 Hours), Day 7 and Day 11 in Treatment Period 1; Day 12 (2,4,6 Hours), Day 21 in Treatment Period 2; Day 22 (2,4,6 Hours), Day 26 and Day 36 in Treatment Period 3

InterventionParticipants (Count of Participants)
2 Hours, Day 124 Hours, Day 126 Hours; Day 12Day 21
Cohort 2: ETR 200 mg0000

[back to top]

Cohort 2: Number of Participants With AEs Leading to Discontinuations and Deaths

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study intervention, whether or not considered related to the study intervention. Number of participants with AEs leading to discontinuations and deaths were reported. (NCT04630002)
Timeframe: Up to Day 36

,,
InterventionParticipants (Count of Participants)
AEs leading to discontinuationsAES leading to deaths
Cohort 2: ETR 200 mg00
Cohort 2: GSK3640254 200 mg00
Cohort 2: GSK3640254 200 mg + ETR 200 mg10

[back to top]

Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Calcium, Creatine Kinase, Creatinine, Phosphate, Potassium and Sodium

Blood samples were collected for analysis of clinical chemistry parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Calcium High, Grade 3: 12.5 to <13.5 mg/dL, Grade 4: >=13.5 mg/dL; Calcium Low, Grade 3: 6.1 to <7.0 mg/dL, Grade 4: <6.1 mg/dL; Creatine Kinase High, Grade 3: 10 to <20 × ULN, Grade 4: >=20 × ULN; Creatinine High, Grade 3: >1.8 to <3.5 ULN, Grade 4: >=3.5 × ULN; Phosphate Low, Grade 3: 1.0 to <1.4 mg/dL, Grade 4: <1.0 mg/dL; Potassium High, Grade 3: 6.5 to <7.0 mEq/L,Grade 4: >=7.0 mEq/L; Potassium Low, Grade 3: 2.0 to <2.5 mEq/L, Grade 4: <2.00 mEq/L; Sodium High, Grade 3: 154 to <160 mEq/L, Grade 4:>=160 mEq/L; Sodium Low, Grade 3: 121 to <125 mEq/L, Grade 4:<=120 mEq/L. Baseline was defined as the latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 36

,,
InterventionParticipants (Count of Participants)
Calcium, Low, Increase to Grade 3Calcium, Low, Increase to Grade 4Calcium, High, Increase to Grade 3Calcium, High, Increase to Grade 4Creatine Kinase, High, Increase to Grade 3Creatine Kinase, High, Increase to Grade 4Creatinine, High, Increase to Grade 3Creatinine, High, Increase to Grade 4Phosphate, Low, Increase to Grade 3Phosphate, Low, Increase to Grade 4Potassium, Low, Increase to Grade 3Potassium, Low, Increase to Grade 4Potassium, High, Increase to Grade 3Potassium, High, Increase to Grade 4Sodium, Low, Increase to Grade 3Sodium, Low, Increase to Grade 4Sodium, High, Increase to Grade 3Sodium, High, Increase to Grade 4
Cohort 2: ETR 200 mg000000000000000000
Cohort 2: GSK3640254 200 mg000000000000000000
Cohort 2: GSK3640254 200 mg + ETR 200 mg000000000000000000

[back to top]

Cohort 1: Number of Participants With Vital Sign Values of Potential Clinical Importance (PCI) Criteria

Vital signs including systolic blood pressure (SBP), diastolic blood pressure (DBP) and pulse rate were measured in a supine position after atleast 5 minutes of rest. The PCI ranges for vitals were as follows; for SBP <85 or >140 millimeters of mercury (mmHg), for DBP <45 or >90 mmHg, for pulse rate <40 or >100 beats per minute. The number of participants with vital signs of PCI were presented. (NCT04630002)
Timeframe: Up to Day 35

,,
InterventionParticipants (Count of Participants)
SBPDBPPulse rate
Cohort 1: DRV/RTV 600/100 mg200
Cohort 1: GSK3640254 200 mg000
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg000

[back to top]

Cohort 1: Number of Participants With Serious Adverse Events (SAEs) and Non-serious Adverse Events (Non-SAEs)

An adverse event (AE) is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study intervention, whether or not considered related to the study intervention. An SAE is defined as any untoward medical occurrence that, at any dose that results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect or other situations as per medical or scientific judgment. Adverse events which were not Serious were considered as Non-Serious adverse events. (NCT04630002)
Timeframe: Up to Day 35

,,
InterventionParticipants (Count of Participants)
SAEnon-SAE
Cohort 1: DRV/RTV 600/100 mg04
Cohort 1: GSK3640254 200 mg06
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg05

[back to top]

Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Urinalysis Parameters

Urine samples were collected for urinalysis parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Erythrocytes High, Grade 3: Gross, with or without clots OR with Red Blood Cells (RBC) casts OR intervention indicated, Grade 4: Life-threatening consequences; Glucose High, Grade 3: >2+ (proportionate concentration by dipstick test) or >500 mg, Grade 4: >500 mg; Protein High, Grade 3: 3+ (proportionate concentration by dipstick test) or higher, Grade 4: Not Applicable. Baseline was defined as the latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 35

,,
InterventionParticipants (Count of Participants)
Erythrocytes, High, Increase to Grade 3Erythrocytes, High, Increase to Grade 4Glucose, High, Increase to Grade 3Glucose, High, Increase to Grade 4Protein, High, Increase to Grade 3Protein, High, Increase to Grade 4
Cohort 1: DRV/RTV 600/100 mg000000
Cohort 1: GSK3640254 200 mg000000
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg000000

[back to top]

Cohort 2: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Glucose, Triglycerides, Lipase, Urate and Cholesterol

Blood samples were collected for analysis of clinical chemistry parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Glucose High, Grade 3: >250 to 500 mg/dL, Grade 4: >=500 mg/dL, Glucose Low, Grade 3: 30 to<40 mg/dL, Grade 4:<30 mg/dL; Triglycerides High, Grade 3: >500 to <1.000 mg/dL, Grade 4:>1000 mg/dL; Lipase High, Grade 3: 3.0 to <5.0×ULN, Grade 4:>=5.0×ULN; Urate High, Grade 3: 12.0 to <15.0 mEq/L, Grade 4:>=15.0 mEq/L; Cholesterol High, Grade 3: >=300 mg/dL, Grade 4: Not Applicable. Baseline was defined as the latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 36

,,
InterventionParticipants (Count of Participants)
Glucose, Low, Increase to Grade 3Glucose, Low, Increase to Grade 4Glucose, High, Increase to Grade 3Glucose, High, Increase to Grade 4Triglycerides, High, Increase to Grade 3Triglycerides, High, Increase to Grade 4Lipase, High, Increase to Grade 3Lipase, High, Increase to Grade 4Urate, High, Increase to Grade 3Urate, High, Increase to Grade 4Cholesterol, High, Increase to Grade 3Cholesterol, High, Increase to Grade 4
Cohort 2: ETR 200 mg000000000000
Cohort 2: GSK3640254 200 mg000000000000
Cohort 2: GSK3640254 200 mg + ETR 200 mg000000000000

[back to top]

Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Hematology Parameters

Blood samples were collected for analysis of hematology parameters. Laboratory abnormalities were graded according to Division of Acquired Immunodeficiency Syndrome (DAIDS) grading table Version 2.1. For Hemoglobin Low, Grade 3: 7.0 to <9.0 Grams per deciliter (g/dL) (males) and 6.5 to <8.5 g/dL (females),Grade 4: <7.0 g/dL (males) and <6.5 g/dL (females); Leukocytes Low, Grade 3: 1000 to 1499 cells per cubic millimeter (cells/mm^3),Grade 4: <1000 cells/mm^3; Lymphocytes Low, Grade 3: 350 to <500 cells per liter (cells/L),Grade 4: <350 cells/L; Neutrophils Low, Grade 3: 400 to 599 cells/mm^3, Grade 4: <400 cells/mm^3; Platelets Low, Grade 3: 25,000 to <50,000 cells/mm^3, Grade 4: <25,000 cells/mm^3. Baseline was defined as latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 35

,,
InterventionParticipants (Count of Participants)
Hemoglobin, Low, Increase to Grade 3Hemoglobin, Low, Increase to Grade 4Leukocytes, Low, Increase to Grade 3Leukocytes, Low, Increase to Grade 4Lymphocytes, Low, Increase to Grade 3Lymphocytes, Low, Increase to Grade 4Neutrophils, Low, Increase to Grade 3Neutrophils, Low, Increase to Grade 4Platelets, Low, Increase to Grade 3Platelets, Low, Increase to Grade 4
Cohort 1: DRV/RTV 600/100 mg0000000000
Cohort 1: GSK3640254 200 mg0000000000
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg0000000000

[back to top]

Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Glucose, Triglycerides, Lipase, Urate and Cholesterol

Blood samples were collected for analysis of clinical chemistry parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Glucose High, Grade 3: >250 to 500 mg/dL, Grade 4: >=500 mg/dL, Glucose Low, Grade 3: 30 to<40 mg/dL, Grade 4:<30 mg/dL; Triglycerides High, Grade 3: >500 to <1.000 mg/dL, Grade 4:>1000 mg/dL; Lipase High, Grade 3: 3.0 to <5.0×ULN, Grade 4:>=5.0×ULN; Urate High, Grade 3: 12.0 to <15.0 mEq/L, Grade 4:>=15.0 mEq/L; Cholesterol High, Grade 3: >=300 mg/dL, Grade 4: Not Applicable. Baseline was defined as the latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 35

,,
InterventionParticipants (Count of Participants)
Glucose, Low, Increase to Grade 3Glucose, Low, Increase to Grade 4Glucose, High, Increase to Grade 3Glucose, High, Increase to Grade 4Triglycerides, High, Increase to Grade 3Triglycerides, High, Increase to Grade 4Lipase, High, Increase to Grade 3Lipase, High, Increase to Grade 4Urate, High, Increase to Grade 3Urate, High, Increase to Grade 4Cholesterol, High, Increase to Grade 3Cholesterol, High, Increase to Grade 4
Cohort 1: DRV/RTV 600/100 mg000000000000
Cohort 1: GSK3640254 200 mg000000000000
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg000000000000

[back to top]

Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Calcium, Creatine Kinase, Creatinine, Phosphate, Potassium and Sodium

Blood samples were collected for analysis of clinical chemistry parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Calcium High, Grade 3: 12.5 to <13.5 milligrams/deciliter (mg/dL), Grade 4: >=13.5 mg/dL; Calcium Low, Grade 3: 6.1 to <7.0 mg/dL, Grade 4: <6.1 mg/dL; Creatine Kinase High, Grade 3: 10 to <20 × ULN, Grade 4: >=20 × ULN; Creatinine High, Grade 3: >1.8 to <3.5 ULN, Grade 4: >=3.5 × ULN; Phosphate Low, Grade 3: 1.0 to <1.4 mg/dL, Grade 4: <1.0 mg/dL; Potassium High, Grade 3: 6.5 to <7.0 Milliequivalents per liter (mEq/L),Grade 4: >=7.0 mEq/L; Potassium Low, Grade 3: 2.0 to <2.5 mEq/L, Grade 4: <2.00 mEq/L; Sodium High, Grade 3: 154 to <160 mEq/L, Grade 4:>=160 mEq/L; Sodium Low, Grade 3: 121 to <125 mEq/L, Grade 4:<=120 mEq/L. Baseline was defined as the latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 35

,,
InterventionParticipants (Count of Participants)
Calcium, Low, Increase to Grade 3Calcium, Low, Increase to Grade 4Calcium, High, Increase to Grade 3Calcium, High, Increase to Grade 4Creatine Kinase, High, Increase to Grade 3Creatine Kinase, High, Increase to Grade 4Creatinine, High, Increase to Grade 3Creatinine, High, Increase to Grade 4Phosphate, Low, Increase to Grade 3Phosphate, Low, Increase to Grade 4Potassium, Low, Increase to Grade 3Potassium, Low, Increase to Grade 4Potassium, High, Increase to Grade 3Potassium, High, Increase to Grade 4Sodium, Low, Increase to Grade 3Sodium, Low, Increase to Grade 4Sodium, High, Increase to Grade 3Sodium, High, Increase to Grade 4
Cohort 1: DRV/RTV 600/100 mg000000000000000000
Cohort 1: GSK3640254 200 mg000000000000000000
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg000000000000000000

[back to top]

Cohort 1: Number of Participants With Grade Increase Post-Baseline Relative to Baseline in Clinical Chemistry Parameters: Alanine Aminotransferase, Albumin, Alkaline Phosphatase, Amylase, Aspartate Aminotransferase, Bilirubin and Direct Bilirubin

Blood samples were collected for analysis of clinical chemistry parameters. Laboratory abnormalities were graded according to DAIDS grading table Version 2.1. For Alanine Aminotransferase High; Grade 3: 5.0 to <10.0 times (×) Upper Limit Normal (ULN), Grade 4: >=10.0 × ULN; Albumin Low, Grade 3: <2.0 grams per deciliter (g/dL), Grade 4: Not Applicable; Alkaline Phosphatase High, Grade 3: 5.0 to <10.0 × ULN, Grade 4: >=10.0 × ULN; Amylase High, Grade 3: 3.0 to <5.0 × ULN, Grade 4: >=5.0 × ULN; Aspartate Aminotransferase High, Grade 3: 5.0 to <10.0 × ULN, Grade 4: >=10.0 × ULN; Bilirubin High, Grade 3: 2.6 to<5.0 × ULN, Grade 4: >=5.0 × ULN and Direct Bilirubin High, Grade 3: >ULN with other signs and symptoms of hepatotoxicity, Grade 4: >ULN with life-threatening consequences. Baseline was defined as latest pre-dose assessment with a non-missing value, including those from unscheduled visits. An increase is defined as an increase in DAIDS grade relative to Baseline grade. (NCT04630002)
Timeframe: Baseline (Pre-dose, Day-1) and up to Day 35

,,
InterventionParticipants (Count of Participants)
Alanine Aminotransferase, High, Increase to Grade 3Alanine Aminotransferase, High, Increase to Grade 4Albumin, Low, Increase to Grade 3Albumin, Low, Increase to Grade 4Alkaline Phosphatase, High, Increase to Grade 3Alkaline Phosphatase, High, Increase to Grade 4Amylase, High, Increase to Grade 3Amylase, High, Increase to Grade 4Aspartate Aminotransferase, High, Increase to Grade 3Aspartate Aminotransferase, High, Increase to Grade 4Bilirubin, High, Increase to Grade 3Bilirubin, High, Increase to Grade 4Direct Bilirubin, High, Increase to Grade 3Direct Bilirubin, High, Increase to Grade 4
Cohort 1: DRV/RTV 600/100 mg00000000000000
Cohort 1: GSK3640254 200 mg00000000000000
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg00000000000000

[back to top]

Cohort 1: Number of Participants With Clinically Significant Abnormal Electrocardiogram (ECG) Findings

A 12-lead ECG was recorded with the participant in a supine position using an automated ECG machine. Clinically significant abnormal findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. Data for number of participants with clinically significant abnormal ECG findings were reported. Data has been presented for the participants with respect to the actual treatment received in respective treatment periods. (NCT04630002)
Timeframe: Day 1 (2,4,6 Hours), Day 7 and Day 11 in Treatment Period 1; Day 12 (2,4,6 Hours), Day 21 in Treatment Period 2; Day 22 (2,4,6 Hours), Day 26 and Day 35 in Treatment Period 3

InterventionParticipants (Count of Participants)
2 Hours; Day 224 Hours; Day 226 Hours; Day 22Day 26Day 35
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg00000

[back to top]

Cohort 1: Number of Participants With Clinically Significant Abnormal Electrocardiogram (ECG) Findings

A 12-lead ECG was recorded with the participant in a supine position using an automated ECG machine. Clinically significant abnormal findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. Data for number of participants with clinically significant abnormal ECG findings were reported. Data has been presented for the participants with respect to the actual treatment received in respective treatment periods. (NCT04630002)
Timeframe: Day 1 (2,4,6 Hours), Day 7 and Day 11 in Treatment Period 1; Day 12 (2,4,6 Hours), Day 21 in Treatment Period 2; Day 22 (2,4,6 Hours), Day 26 and Day 35 in Treatment Period 3

InterventionParticipants (Count of Participants)
2 Hours, Day 14 Hours, Day 16 Hours; Day 1Day 7Day 11
Cohort 1: GSK3640254 200 mg00000

[back to top]

Cohort 1: Number of Participants With Clinically Significant Abnormal Electrocardiogram (ECG) Findings

A 12-lead ECG was recorded with the participant in a supine position using an automated ECG machine. Clinically significant abnormal findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. Data for number of participants with clinically significant abnormal ECG findings were reported. Data has been presented for the participants with respect to the actual treatment received in respective treatment periods. (NCT04630002)
Timeframe: Day 1 (2,4,6 Hours), Day 7 and Day 11 in Treatment Period 1; Day 12 (2,4,6 Hours), Day 21 in Treatment Period 2; Day 22 (2,4,6 Hours), Day 26 and Day 35 in Treatment Period 3

InterventionParticipants (Count of Participants)
2 Hours, Day 124 Hours, Day 126 Hours; Day 12Day 21
Cohort 1: DRV/RTV 600/100 mg0000

[back to top]

Cohort 1: Number of Participants With AEs Leading to Discontinuations and Deaths

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study intervention, whether or not considered related to the study intervention. Number of participants with AEs leading to discontinuations and deaths were reported. (NCT04630002)
Timeframe: Up to Day 35

,,
InterventionParticipants (Count of Participants)
AEs leading to discontinuationsAES leading to deaths
Cohort 1: DRV/RTV 600/100 mg10
Cohort 1: GSK3640254 200 mg00
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg00

[back to top]

Cohort 3: Cmax of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 2

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 3: GSK3640254 200 mg1.578
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg1.383

[back to top]

Cohort 3: AUC(0-tau) of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 2

InterventionHours*micrograms per milliliter (Geometric Mean)
Cohort 3: GSK3640254 200 mg24.99
Cohort 3: GSK3640254 200 mg+ DRV/RTV 600/100 mg+ ETR 200 mg22.78

[back to top]

Cohort 2: Tmax of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 3

InterventionHours (Median)
Cohort 2: GSK3640254 200 mg4.000
Cohort 2: GSK3640254 200 mg + ETR 200 mg3.000

[back to top]

Cohort 2: Tmax of ETR

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionHours (Median)
Cohort 2: ETR 200 mg3.500
Cohort 2: GSK3640254 200 mg + ETR 200 mg4.000

[back to top]

Cohort 2: Number of Participants With Clinically Significant Abnormal ECG Findings

A 12-lead ECG was recorded with the participant in a supine position using an automated ECG machine. Clinically significant abnormal findings are those which are not associated with the underlying disease, unless judged by the investigator to be more severe than expected for the participant's condition. Data for number of participants with clinically significant abnormal ECG findings were reported. Data has been presented for the participants with respect to the actual treatment received in respective treatment periods. (NCT04630002)
Timeframe: Day 1 (2,4,6 Hours), Day 7 and Day 11 in Treatment Period 1; Day 12 (2,4,6 Hours), Day 21 in Treatment Period 2; Day 22 (2,4,6 Hours), Day 26 and Day 36 in Treatment Period 3

InterventionParticipants (Count of Participants)
2 Hours; Day 224 Hours; Day 226 Hours; Day 22Day 26Day 36
Cohort 2: GSK3640254 200 mg + ETR 200 mg00000

[back to top]

Cohort 2: Ctau of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 2: GSK3640254 200 mg0.7706
Cohort 2: GSK3640254 200 mg + ETR 200 mg0.3901

[back to top]

Cohort 2: Ctau of ETR

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 2: ETR 200 mg0.4705
Cohort 2: GSK3640254 200 mg + ETR 200 mg0.5837

[back to top]

Cohort 2: Cmax of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 2: GSK3640254 200 mg1.889
Cohort 2: GSK3640254 200 mg + ETR 200 mg1.136

[back to top]

Cohort 2: Cmax of ETR

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionMicrograms per milliliter (Geometric Mean)
Cohort 2: ETR 200 mg0.9749
Cohort 2: GSK3640254 200 mg + ETR 200 mg1.102

[back to top]

Cohort 2: AUC(0-tau) of GSK3640254

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours, 16 Hours, 24 Hours post-dose in Treatment Periods 1 and 3

InterventionHours*micrograms per milliliter (Geometric Mean)
Cohort 2: GSK3640254 200 mg27.86
Cohort 2: GSK3640254 200 mg + ETR 200 mg14.73

[back to top]

Cohort 2: AUC(0-tau) of ETR

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionHours*micrograms per milliliter (Geometric Mean)
Cohort 2: ETR 200 mg8.340
Cohort 2: GSK3640254 200 mg + ETR 200 mg9.791

[back to top]

Cohort 1: Tmax of RTV

Blood samples were collected at indicated time points. Pharmacokinetic analysis was conducted using standard non-compartmental methods. (NCT04630002)
Timeframe: Pre-dose and 0.5 Hours, 1 Hour, 1.5 Hours, 2 Hours, 3 Hours, 4 Hours, 6 Hours, 8 Hours, 12 Hours post-dose in Treatment Periods 2 and 3

InterventionHours (Median)
Cohort 1: DRV/RTV 600/100 mg4.000
Cohort 1: GSK3640254 200 mg + DRV/RTV 600/100 mg4.000

[back to top]