Page last updated: 2024-12-10

maraviroc

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 CID3002977
CHEMBL ID256907
CHEMBL ID1201187
CHEMBL ID584744
CHEBI ID63608
CHEBI ID184662
SCHEMBL ID51991
SCHEMBL ID2177194
SCHEMBL ID4576508
MeSH IDM0493371

Synonyms (102)

Synonym
AC-558
HY-13004
4,4-difluoro-cyclohexanecarboxylic acid {(s)-3-[(1s,5s)-3-(3-isopropyl-5-methyl-[1,2,4]triazol-4-yl)-8-aza-bicyclo[3.2.1]oct-8-yl]-1-phenyl-propyl}-amide
4,4-difluoro-n-((s)-3-(3-(3-isopropyl-5-methyl-4h-1,2,4-triazol-4-yl)-8-aza-bicyclo[3.2.1]octan-8-yl)-1-phenylpropyl)cyclohexanecarboxamide
4,4-difluoro-n-((s)-3-((1s,3s,5r)-3-(3-isopropyl-5-methyl-4h-1,2,4-triazol-4-yl)-8-aza-bicyclo[3.2.1]octan-8-yl)-1-phenylpropyl)cyclohexanecarboxamide
bdbm50334986
4,4-difluoro-n-((1s)-3-(3-(3-isopropyl-5-methyl-4h-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]octan-8-yl)-1-phenylpropyl)cyclohexanecarboxamide
[3h]uk 427,857
gtpl803
[3h]uk-427,857
[3h]maraviroc
gtpl806
exo-4,4-difluoro-n-[3-[3-(3-isopropyl-5-methyl-4h-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]oct-8-yl]-1(s)-phenylpropyl]cyclohexanecarboxamide
selzentry(tm)
celsentri(tm)
uk-427,857 maraviroc (mvc)
376348-65-1
maraviroc ,
MVC ,
uk-427857
4,4-difluoro-n-[(1s)-3-[(1r,5s)-3-(3-isopropyl-5-methyl-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]octan-8-yl]-1-phenyl-propyl]cyclohexanecarboxamide
uk-427,857
selzentry
celsentri
pro 140 & maraviroc
4,4-difluoro-n-[(1s)-3-[(1s,5r)-3-(3-isopropyl-5-methyl-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]octan-8-yl]-1-phenyl-propyl]cyclohexanecarboxamide
pro 140 (anti-ccr5 monoclonal antibody) & exo-4,4-difluoro-n-[3-[3-(3-isopropyl-5-methyl-4h-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]oct-8-yl]-1(s)-phenylpropyl]cyclohexanecarboxamide
DB04835
cyclohexanecarboxamide, 4,4-difluoro-n-((1s)-3-((3-exo)-3-(3-methyl-5-(1-methylethyl)-4h-1,2,4-triazol-4-yl)-8-azabicyclo(3.2.1)oct-8-yl)-1-phenylpropyl)-
uk 427857
isopropyl, 4,4-difluoro-n-((1s)-3-{(1r,3s,5s)-3-(3-methyl-5-(propan-2-yl)-4h-1,2,4-triazol-4-yl)-8-azabicyclo(3.2.1)octan-8-yl}-1-phenylpropyl)cyclohexanecarboxamide
CHEMBL256907 ,
chebi:63608 ,
rel-maraviroc
CHEMBL1201187 ,
4,4-difluoro-n-[(1s)-3-[(1r,5s)-3-(3-methyl-5-propan-2-yl-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]octan-8-yl]-1-phenylpropyl]cyclohexane-1-carboxamide
CHEBI:184662
4,4-difluoro-n-[(1s)-3-[(1r,5s)-3-[3-methyl-5-(propan-2-yl)-4h-1,2,4-triazol-4-yl]-8-azabicyclo[3.2.1]octan-8-yl]-1-phenylpropyl]cyclohexane-1-carboximidic acid
4,4-diluoro-n-[(1s)-3-[(1s,5r)-3-(3-methyl-5-propan-2-yl-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]octan-8-yl]-1-phenylpropyl]cyclohexane-1-carboxamide
NCGC00183109-01
NCGC00183109-02
md6p741w8a ,
maraviroc [inn:ban:jan]
unii-md6p741w8a
hsdb 8021
tox21_113369
dtxsid8048949 ,
cas-376348-65-1
dtxcid5028875
4,4-difluoro-n-[(1s)-3-{(3-exo)-3-[3-methyl-5-(propan-2-yl)-4h-1,2,4-triazol-4-yl]-8-azabicyclo[3.2.1]oct-8-yl}-1-phenylpropyl]cyclohexanecarboxamide
MRV ,
4,4-difluoro-n-[(1s)-3-[(3-exo)-3-[3-methyl-5-(1-methylethyl)-4h-1,2,4-triazol-4-yl]-8-azabicyclo[3.2.1]oct-8-yl]-1-phenylpropyl]cyclohexanecarboxamide
maravirocum
4,4-difluoro-n-{(1s)-3-[(3-exo)-3-(3-isopropyl-5-methyl-4h-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]oct-8-yl]-1-phenylpropyl}cyclohexanecarboxamide
maraviroc [orange book]
maraviroc [who-dd]
maraviroc [mart.]
maraviroc [vandf]
maraviroc [ema epar]
maraviroc [jan]
maraviroc [mi]
cyclohexanecarboxamide, 4,4-difluoro-n-[(1s)-3-[(3-exo)-3-[3-methyl-5-(1-methylethyl)-4h-1,2,4-triazol-4-yl]-8-azabicyclo[3.2.1]oct-8-yl]-1-phenylpropyl]-
maraviroc [inn]
CS-0366
BRD-A23284911-001-02-4
SCHEMBL51991
CHEMBL584744 ,
4,4-difluoro-n-((1s)-3-((1r,5s)-3-(3-isopropyl-5-methyl-4h-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]octan-8-yl)-1-phenylpropyl)cyclohexanecarboxamide
SCHEMBL2177194
SCHEMBL4576508
MLS006011960
smr004703532
4,4-difluoro-n-((1s)-3-((1r,5s)-3-(3-isopropyl-5-methyl-4h-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]octan-8-yl)-1-phenylpropyl)cyclohexane-1-carboxamide
AKOS025402143
J-521678
EX-A200
sr-01000942244
SR-01000942244-1
maraviroc, >=98% (hplc)
4,4-difluoro-n-[(1s)-3-[(1r,3s,5s)-3-[3-methyl-5-(propan-2-yl)-4h-1,2,4-triazol-4-yl]-8-azabicyclo[3.2.1]octan-8-yl]-1-phenylpropyl]cyclohexane-1-carboxamide
AKOS032946723
AKOS032960315
Q421369
4,4-difluoro-n-((s)-3-((1r,3r,5s)-3-(3-isopropyl-5-methyl-4h-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]octan-8-yl)-1-phenylpropyl)cyclohexane-1-carboxamide
AS-75265
AMY12578
cyclohexanecarboxamide,4,4-difluoro-n-[(1s)-3-[(3-exo)-3-[3-methyl-5-(1-methylethyl)-4h-1,2,4-triazol-4-yl]-8-azabicyclo[3.2.1]oct-8-yl]-1-phenylpropyl]-
bdbm50464147
EN300-22828861
2414315-81-2
gsnhkudzzfzsjb-qumgssfmsa-n
gsnhkudzzfzsjb-hlmsnrgbsa-n
A3C7SRV8DV
cyclohexanecarboxamide, 4,4-difluoro-n-[(1s)-3-[(3-endo)-3-[3-methyl-5-(1-methylethyl)-4h-1,2,4-triazol-4-yl]-8-azabicyclo[3.2.1]oct-8-yl]-1-phenylpropyl]-
maraviroc, endo-
4,4-difluoro-n-[(1s)-3-[(3-endo)-3-[3-methyl-5-(1-methylethyl)-4h-1,2,4-triazol-4-yl]-8-azabicyclo[3.2.1]oct-8-yl]-1-phenylpropyl]cyclohexanecarboxamide
4,4-difluoro-n-[(1s)-3-[(1r,3r,5s)-3-[3-methyl-5-(propan-2-yl)-4h-1,2,4-triazol-4-yl]-8-azabicyclo[3.2.1]octan-8-yl]-1-phenylpropyl]cyclohexane-1-carboxamide
EN300-7367238
bdbm50583789
4,4-difluoro-n-[(1s)-3-[(1r,5s)-3-[3-methyl-5-(propan-2-yl)-4h-1,2,4-triazol-4-yl]-8-azabicyclo[3.2.1]octan-8-yl]-1-phenylpropyl]cyclohexane-1-carboxamide
EN300-128501
Z1614451557

Research Excerpts

Overview

Maraviroc is a new antiretroviral agent that has been associated with immunomodulatory proprieties. Its use may be a promising strategy to minimize the rate of rejections in HIV-infected kidney transplant (KT) recipients. MaraviroC (MVC) is a candidate drug for HIV preexposure prophylaxis (PrEP)

ExcerptReferenceRelevance
"Maraviroc 1 is an approved antagonist of the human CCR5 receptor which prevents the entry of HIV."( An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
Batchelor, DV; Corless, M; Dorr, PK; Ellis, D; Fenwick, DR; Galan, SR; Jones, RM; Mason, HJ; Middleton, DS; Perros, M; Perruccio, F; Platts, MY; Pryde, DC; Rodrigues, D; Smith, NN; Stephenson, PT; Stupple, PA; Webster, R; Westby, M; Wood, A, 2011
)
1.09
"Maraviroc is an antiretroviral agent and C-C chemokine coreceptor 5 (CCR5) antagonist that is currently used to treat human immunodeficiency virus. "( The utility of maraviroc, an antiretroviral agent used to treat HIV, as treatment for opioid abuse? Data from MRI and behavioural testing in rats.
Borges, C; Cai, X; Ferris, CF; Iriah, SC; Kulkarni, PP; Madularu, D; Shalev, U, 2021
)
2.42
"Maraviroc is a new antiretroviral agent that has been associated with immunomodulatory proprieties; therefore, its use may be a promising strategy to minimize the rate of rejections in HIV-infected kidney transplant (KT) recipients."( Role of Maraviroc in minimizing the risk of graft rejection in HIV-infected kidney transplant recipients.
Alfano, G; Cappelli, G; Dolci, G; Fontana, F; Franceschini, E; Guaraldi, G; Mussini, C, 2020
)
1.71
"Maraviroc is an entry inhibitor of human immunodeficiency virus type 1 (HIV-1) that prevents the interaction between CCR5 and the surface subunit gp120 of the viral envelope glycoprotein (Env)."( In vitro inhibitory effect of maraviroc on the association of the simian immunodeficiency virus envelope glycoprotein with CCR5.
Affranchino, JL; Giraudy, I; González, SA; Ovejero, CA, 2021
)
1.63
"Maraviroc (MVC) is an allosteric inhibitor of human immunodeficiency virus type 1 (HIV-1) entry, and is the only CCR5 antagonist licensed for use as an anti-HIV-1 therapeutic. "( Analysis of Clinical HIV-1 Strains with Resistance to Maraviroc Reveals Strain-Specific Resistance Mutations, Variable Degrees of Resistance, and Minimal Cross-Resistance to Other CCR5 Antagonists.
Borm, K; Cashin, K; Churchill, M; Duncan, R; Ellenberg, P; Ellett, A; Flynn, JK; Gorry, PR; Gray, LR; Jubb, B; Lee, B; Lewin, SR; Lewis, M; Roche, M; Sterjovski, J; Westby, M; Zhou, J, 2017
)
2.15
"Maraviroc (MVC) is a candidate drug for HIV preexposure prophylaxis (PrEP)."( Safety and Tolerability of Maraviroc-Containing Regimens to Prevent HIV Infection in Women: A Phase 2 Randomized Trial.
Amico, KR; Andrade, A; Andrew, P; Chege, W; Chen, YQ; Cottle, LM; Eshleman, SH; Frank, I; Gulick, RM; Hendrix, CW; Ho, K; Hodder, S; Klingman, KL; Landovitz, RJ; Manabe, YC; Marcus, C; Marzinke, MA; Mayer, KH; McCauley, M; McGowan, I; Richardson, P; Rinehart, AR; Rooney, JF; Salata, RA; Santana, J; Siegel, M; Stekler, JD; Swaminathan, S; Wilkin, TJ; Young, AM, 2017
)
2.19
"Maraviroc is a CC-chemokine receptor 5 antagonist approved to treat adults infected with CC-chemokine receptor 5-tropic (R5) HIV-1. "( Pharmacokinetics, Safety and Efficacy of Maraviroc in Treatment-experienced Pediatric Patients Infected With CCR5-Tropic HIV-1.
Chokephaibulkit, K; Clark, A; Craig, C; Fang, A; Fourie, J; Giaquinto, C; Heera, J; Mawela, MP; McFadyen, L; Mitha, IH; Negra, MD; Valluri, SR; van der Ryst, E; Vourvahis, M; Zhang-Roper, RY, 2018
)
2.19
"Maraviroc is a CCR5 antagonist used in the treatment of HIV-1 infection. "( Maraviroc Is Associated with Latent HIV-1 Reactivation through NF-κB Activation in Resting CD4
Coronel Díaz, M; Díaz-de Santiago, A; Domínguez, E; Dronda, F; García-Bermejo, ML; Gutiérrez, C; Hernández-Novoa, B; López-Huertas, MR; Madrid-Elena, N; Moreno, S; Sastre, B; Serrano-Villar, S, 2018
)
3.37
"Maraviroc (MVC) is an orally dosed CCR5 antagonist approved for use in patients infected with CCR5-trophic HIV-1."( Towards a Maraviroc long-acting injectable nanoformulation.
Clark, A; Dwyer, A; Owen, A; Rannard, SP; Savage, AC; Scott, T; Siccardi, M; Tatham, LM; Vourvahis, M, 2019
)
1.64
"Maraviroc is a C-C chemokine receptor type-5 antagonist approved for the treatment of HIV-1. "( No Clinical Impact of CYP3A5 Gene Polymorphisms on the Pharmacokinetics and/or Efficacy of Maraviroc in Healthy Volunteers and HIV-1-Infected Subjects.
Chan, PLS; Clark, A; Fang, A; Fate, GD; Haynes, J; Heera, J; Marshall, JC; McFadyen, L; Nedderman, A; Nepal, S; Savage, ME; Smith, KY; Valluri, SR; Vourvahis, M; Wood, LS, 2019
)
2.18
"Maraviroc is a chemokine receptor 5 (CCR5) inhibitor used in the treatment of human immunodeficiency virus (HIV) that also shows therapeutic potential for several autoimmune, cancer, and inflammatory diseases that can afflict pregnant women. "( Interactions between Maraviroc and the ABCB1, ABCG2, and ABCC2 Transporters: An Important Role in Transplacental Pharmacokinetics.
Ambrus, C; Ceckova, M; Gaborik, Z; Ptackova, Z; Sorf, A; Staud, F; Tupova, L, 2019
)
2.28
"Maraviroc is a first-in-class chemokine coreceptor type-5 (CCR5) antagonist with demonstrated immunovirologic activity in treatment-experienced (TE) patients with CCR5 (R5)-tropic HIV-1; however, experience in regimens containing newer antiretroviral agents is limited. "( Safety and immunovirologic outcomes with maraviroc combination regimens in patients with a history of past treatment failures and virologic resistance in Brazil: an open-label, multicenter phase 3b study.
Bicudo, EL; Cassoli, LM; da Eira, M; de Andrade Neto, JL; Furtado, J; Leite, OH; Lewi, DS; Lima, MP; Lopes, MI; Machado, AA; Madruga, JV; Miranda, AF; Netto, EM; Pedro, Rde J; Portsmouth, S; Santini-Oliveira, M; Santos, BR; Tupinambas, U; Wajsbrot, DB, 2013
)
2.1
"Maraviroc (MVC) is a potent CCR5 coreceptor antagonist that is in clinical testing for daily oral pre-exposure prophylaxis (PrEP) for HIV prevention. "( Lack of prophylactic efficacy of oral maraviroc in macaques despite high drug concentrations in rectal tissues.
Aubert, R; Aung, W; Bachman, S; García-Lerma, JG; Heneine, W; Kersh, E; Martin, A; Massud, I; Mitchell, J; Pau, CP; Solomon Tsegaye, T, 2013
)
2.1
"Maraviroc is an antiretroviral drug in the CCR5 receptor antagonist class, which is used in the treatment of HIV. "( A validated stability-indicating UPLC method for the determination of impurities in Maraviroc.
Chilukuri, M; Hussainreddy, K; Narayanareddy, P; Venkataramana, M, 2014
)
2.07
"Maraviroc is an HIV entry inhibitor approved for treatment of both drug-sensitive and drug-resistant HIV strains."( Interactions between alcohol and the HIV entry inhibitor Maraviroc.
Aweeka, FT; Beatty, GW; Gruber, VA; Lum, PJ; McCance-Katz, EF; Rainey, PM,
)
1.1
"Maraviroc is a non-peptidic, low molecular weight CC chemokine receptor 5 (CCR5) ligand that has recently been marketed for the treatment of HIV infected individuals. "( Modeling the allosteric modulation of CCR5 function by Maraviroc.
Garcia-Perez, J; Kellenberger, E; Lagane, B, 2013
)
2.08
"Maraviroc is a CC-chemokine receptor 5 (CCR5) antagonist with potent antiviral and cancer preventive effects. "( Maraviroc attenuates trauma-hemorrhage-induced hepatic injury through PPAR gamma-dependent pathway in rats.
Liu, FC; Tsai, YF; Yu, HP, 2013
)
3.28
"Maraviroc is a CCR5 inhibitor approved in 2007 for treatment of therapy experienced adult patients infected with CCR5-tropic HIV-1 virus. "( A validated HPLC-MS method for quantification of the CCR5 inhibitor maraviroc in HIV+ human plasma.
Audino, A; Baietto, L; Bonora, S; D'Avolio, A; Di Perri, G; Sciandra, M; Simiele, M, 2014
)
2.08
"Maraviroc is a CCR5 antagonist that has been utilized as a viral entry inhibitor in the management of HIV-1. "( Development and bioanalytical validation of a liquid chromatographic-tandem mass spectrometric (LC-MS/MS) method for the quantification of the CCR5 antagonist maraviroc in human plasma.
Emory, JF; Marzinke, MA; Seserko, LA, 2014
)
2.04
"Maraviroc is a new CCR5 antagonist designed to block HIV entry."( CCR5 as a treatment target in pulmonary arterial hypertension.
Abid, S; Adnot, S; Amsellem, V; Bobe, R; Combadière, C; Delcroix, M; Dubois-Randé, JL; Gary-Bobo, G; Gladwin, MT; Houssaini, A; Lipskaia, L; Marcos, E; Mouraret, N; Norris, KA; Parpaleix, A; Poupel, L; Quarck, R; Saker, M, 2014
)
1.12
"Maraviroc is a first-in-class antiretroviral (ARV) drug acting on a host cell target (CCR5), which blocks the entry of the HIV virus into the cell. "( Preclinical discovery and development of maraviroc for the treatment of HIV.
Glisic, S; Radi, M; Tassini, S; Veljkovic, N; Veljkovic, V; Vucicevic, J, 2015
)
2.13
"Maraviroc is an HIV entry inhibitor that alters the conformation of CCR5 and is poorly efficient in patients infected by viruses that use CXCR4 as an entry coreceptor. "( HIV-1 Coreceptor Usage Assessment by Ultra-Deep Pyrosequencing and Response to Maraviroc.
Bellecave, P; Calvez, V; Charpentier, C; Descamps, D; Flandre, P; Marcelin, AG; Masquelier, B; Pawlotsky, JM; Recordon-Pinson, P; Rodriguez, C; Soulié, C, 2015
)
2.09
"Maraviroc (MVC) is an allosteric CCR5 inhibitor used against HIV-1 infection. "( A single-residue change in the HIV-1 V3 loop associated with maraviroc resistance impairs CCR5 binding affinity while increasing replicative capacity.
Alcami, J; Arenzana-Seisdedos, F; Azoulay, S; Cascajero, A; Colin, P; Garcia-Perez, J; Heinrich, JT; Kellenberger, E; Lagane, B; Lortat-Jacob, H; Staropoli, I, 2015
)
2.1
"Maraviroc is a first-in-class selective CCR5 antagonist only approved in combination with other antiretrovirals for the treatment of HIV-infection. "( Off-label use of maraviroc in clinical practice.
Blanco, JR; Ochoa-Callejero, L, 2016
)
2.22
"Maraviroc (MVC) is a potential candidate for 'on demand' preexposure prophylaxis. "( Single oral dose of maraviroc does not prevent ex-vivo HIV infection of rectal mucosa in HIV-1 negative human volunteers.
Back, D; Boix, J; Cabrera, C; Carrillo, A; Clotet, B; Coll, J; Else, L; Escrig, R; García, E; Gómez-Mora, E; Moltó, J; Ouchi, D; Paredes, R, 2015
)
2.18
"The Maraviroc Switch Study is a randomized, multicenter, 96-week, open-label switch study in HIV type 1-infected adults with R5-tropic virus, virologically suppressed on a ritonavir-boosted protease inhibitor (PI/r) plus double nucleoside/nucleotide reverse transcriptase inhibitor (2 N(t)RTI) backbone. "( Maraviroc, as a Switch Option, in HIV-1-infected Individuals With Stable, Well-controlled HIV Replication and R5-tropic Virus on Their First Nucleoside/Nucleotide Reverse Transcriptase Inhibitor Plus Ritonavir-boosted Protease Inhibitor Regimen: Week 48 R
Amin, J; Andrade-Villanueva, J; Arnaiz, JA; Belloso, W; Clark, A; Clarke, A; Cooper, D; Emery, S; Fisher, M; Gatell, J; Gill, J; Harrigan, R; Horban, A; Kaiser, R; Kelleher, A; Losso, M; Mallon, P; Pett, SL; Porteiro, N; Prazuck, T; Rockstroh, JK; Ruxrungtham, K; Sierra Madero, J; Silk, D; Sugiura, W; Tu, E; Wolff, M; Woolley, I, 2016
)
2.44
"Maraviroc is an allosteric small molecule antagonist of chemokine receptor type 5 (CCR5) and has been used in adult allogeneic hematopoietic stem cell transplant (HSCT) recipients to prevent acute graft-versus-host disease (GVHD) of the gastrointestinal (GI) tract and liver. "( A Pharmacokinetic and Pharmacodynamic Study of Maraviroc as Acute Graft-versus-Host Disease Prophylaxis in Pediatric Allogeneic Stem Cell Transplant Recipients with Nonmalignant Diagnoses.
Davies, SM; Fukuda, T; Kashuba, ADM; Khandelwal, P; Marsh, RA; Mehta, PA; Mizuno, K; Teusink-Cross, A; Vinks, AA, 2016
)
2.13
"Maraviroc (MVC) is a candidate for human immunodeficiency virus (HIV) pre-exposure prophylaxis."( Phase 2 Study of the Safety and Tolerability of Maraviroc-Containing Regimens to Prevent HIV Infection in Men Who Have Sex With Men (HPTN 069/ACTG A5305).
Amico, KR; Andrade, A; Andrew, P; Chege, W; Chen, YQ; Cottle, LM; Eshleman, SH; Farley, JE; Frank, I; Gulick, RM; Hendrix, CW; Ho, K; Klingman, KL; Landovitz, RJ; Liu, A; Magnus, M; Marcus, C; Marzinke, MA; Mayer, KH; McCauley, M; McGowan, I; Richardson, P; Rinehart, AR; Rooney, JF; Salata, RA; Santana, J; Stekler, JD; Wilkin, TJ; Young, AM, 2017
)
2.15
"Maraviroc (MVC) is a suitable drug for aviraemic subjects on antiretroviral treatment (ART) developing toxicity. "( Genotypic tropism testing of proviral DNA to guide maraviroc initiation in aviraemic subjects: 48-week analysis of results from the PROTEST study.
Aiestaran, A; Artero, A; Bernal, E; Blanco, F; Flores, J; García, F; García-Deltoro, M; Gutiérrez, F; Hernández-Quero, J; Imaz, A; Martínez-Madrid, OJ; Mínguez-Gallego, C; Navarro, J; Ocampo, A; Paredes, R; Pasquau, J; Pérez, N; Pérez-Elías, MJ; Poveda, E; Ribas, MA, 2017
)
2.15
"Maraviroc is a substrate for CYP3A4, and exposure will therefore be modulated by CYP3A4 inhibitors. "( Maraviroc: in vitro assessment of drug-drug interaction potential.
Collins, C; Dickins, M; Hyland, R; Jones, B; Jones, H, 2008
)
3.23
"Maraviroc is a selective, reversible, small-molecule CCR5-receptor antagonist. "( Maraviroc: a CCR5-receptor antagonist for the treatment of HIV-1 infection.
Bandres, JC; Fung, HB; Lieberman-Blum, SS, 2008
)
3.23
"2. Maraviroc is a novel CCR5 antagonist for the treatment of HIV-1 infection."( Preclinical assessment of the distribution of maraviroc to potential human immunodeficiency virus (HIV) sanctuary sites in the central nervous system (CNS) and gut-associated lymphoid tissue (GALT).
Bowers, SJ; Mitchell, RJ; Potchoiba, MJ; Schroeder, CM; Small, HF; Walker, DK, 2008
)
1.12
"Maraviroc is a potent inhibitor of HIV-1 replication and contributes to effective viral suppression in combination with traditional antiretroviral medications. "( Developing clinical role of a CCR5 co-receptor antagonist in HIV-1 infection.
Caldwell, DJ; Evans, JD, 2008
)
1.79
"Maraviroc is a novel drug and different from the rest of the antiretrovirals due to the special characteristics of its mechanism of action and is also the first antiretroviral directed towards a cell target."( [Introduction. A brief history of AIDS].
Alcamí, J, 2008
)
1.07
"Maraviroc (MVC) is a new antagonist of the CCR5 coreceptor and is the first antiviral compound available that has a cell factor essential for HIV entry as a target. "( [Mechanisms of resistance and failure of treatment with maraviroc].
Delgado, R, 2008
)
2.03
"Maraviroc is a selective and slowly reversible antagonist of the CCR5 co-receptor which has shown to have powerful antiviral activity, in vitro, against a wide range of HIV clinical isolates, including strains multi-resistant to 4 classes of pre-existing antiretroviral drugs. "( [At what time and with which combinations should maraviroc be indicated in the new antiretroviral treatment scenario?].
Camacho, A; Genebat, M; Leal, M; Rivero, A, 2008
)
2.04
"Maraviroc is a small molecule and a member of a new class of antiretroviral compounds known as CCR5 antagonists, which block R5-tropic HIV entry into CD4 cells. "( Maraviroc: a new CCR5 antagonist.
Khanlou, H; Sayana, S, 2009
)
3.24
"Maraviroc is a potent selective CCR5 antagonist and is the first of this new class of oral agents to be approved for the treatment of CCR5-tropic HIV type-1. "( Maraviroc: pharmacokinetics and drug interactions.
Abel, S; Back, DJ; Vourvahis, M, 2009
)
3.24
"* Maraviroc is a CCR5 receptor antagonist, while raltegravir is a HIV-1 integrase inhibitor. "( Assessment of the pharmacokinetics of co-administered maraviroc and raltegravir.
Andrews, E; Crownover, P; Damle, B; Fang, J; Glue, P; Tressler, R, 2010
)
1.33
"Maraviroc is an orally available antagonist of the CCR5 chemokine receptor, which acts as a human immunodeficiency virus type 1 (HIV-1) coreceptor. "( Development and application of a simple LC-MS method for the determination of plasma maraviroc concentrations.
Hirano, A; Kaneda, T; Kinoshita, E; Nomura, T; Okubo, N; Takahashi, M, 2010
)
2.03
"Maraviroc is a first-in-class CCR5 antagonist that shows potent anti-HIV-1 activity in vitro and in vivo and is well tolerated in both healthy volunteers and HIV-1-infected patients. "( An LC-MS-MS method for quantitative determination of maraviroc (UK-427,857) in human plasma, urine and cerebrospinal fluid.
Brewer, E; Clarke, P; Edgington, A; Felix, T; Muirhead, D, 2010
)
2.05
"Maraviroc is a nonpeptidic small molecule human immunodeficiency virus type 1 (HIV-1) entry inhibitor that has just entered the therapeutic arsenal for the treatment of patients. "( Allosteric model of maraviroc binding to CC chemokine receptor 5 (CCR5).
Alcami, J; Arenzana-Seisdedos, F; Garcia-Perez, J; Kellenberger, E; Lagane, B; Rognan, D; Rueda, P, 2011
)
2.14
"Maraviroc proved to be an effective antiretroviral agent for a combination therapy of HIV infection."( [Our experience with maraviroc treatment in HIV positive patients].
Machala, L; Perlík, F; Slanar, O; Stanková, M; Vondracková, H, 2011
)
1.41
"Maraviroc is a generally safe and well-tolerated medication for the treatment of HIV-1 infection with a unique mechanism of action. "( Drug safety evaluation of maraviroc for the treatment of HIV infection.
Hardy, WD; Rockstroh, JK; Wasmuth, JC, 2012
)
2.12
"Maraviroc is an anti-human immunodeficiency virus drug that acts by blocking viral entry into target cells. "( Cytochrome P450 3A5 plays a prominent role in the oxidative metabolism of the anti-human immunodeficiency virus drug maraviroc.
Bumpus, NN; Hendrix, CW; Lu, Y, 2012
)
2.03
"Maraviroc (MVC) is a CCR5 antagonist that inhibits HIV-1 entry by binding to the coreceptor and inducing structural alterations in the extracellular loops. "( HIV-1 resistance to maraviroc conferred by a CD4 binding site mutation in the envelope glycoprotein gp120.
Arts, EJ; Ratcliff, AN; Shi, W, 2013
)
2.16
"Maraviroc (MVC) is a drug approved for use as part of HAART in treatment-experienced HIV-1 patients with CCR5-tropic virus. "( Maraviroc reduces cytokine expression and secretion in human adipose cells without altering adipogenic differentiation.
Díaz-Delfín, J; Domingo, P; Giralt, M; Villarroya, F, 2013
)
3.28
"Maraviroc (UK-427,857) is a selective CCR5 antagonist with potent anti-human immunodeficiency virus type 1 (HIV-1) activity and favorable pharmacological properties. "( Maraviroc (UK-427,857), a potent, orally bioavailable, and selective small-molecule inhibitor of chemokine receptor CCR5 with broad-spectrum anti-human immunodeficiency virus type 1 activity.
Armour, D; Dobbs, S; Dorr, P; Griffin, P; Irvine, B; Macartney, M; Mori, J; Napier, C; Perros, M; Price, D; Rickett, G; Smith-Burchnell, C; Stammen, B; Webster, R; Westby, M; Wood, A, 2005
)
3.21
"Maraviroc is a CCR5 antagonist in clinical development as one of a new class of antiretrovirals targeting human immunodeficiency virus type 1 (HIV-1) coreceptor binding. "( Reduced maximal inhibition in phenotypic susceptibility assays indicates that viral strains resistant to the CCR5 antagonist maraviroc utilize inhibitor-bound receptor for entry.
Ciaramella, G; Dorr, P; Lewis, M; Mori, J; Mosley, M; Perros, M; Smith-Burchnell, C; Stockdale, M; Westby, M, 2007
)
1.99
"Maraviroc is a specific, slowly reversible, noncompetitive, small-molecule antagonist of the CCR5 chemokine receptor, which also serves as an HIV-1 coreceptor. "( Maraviroc.
Carter, NJ; Keating, GM, 2007
)
3.23
"Maraviroc (Celsentri) is an orally administered drug available as 150 and 300 mg film-coated tablets."( [Clinical pharmacokinetic of maraviroc].
Peytavin, G, 2008
)
1.36

Effects

Maraviroc (MVC) has shown good efficacy and tolerability in treatment-naive and treatment-experienced HIV-1-infected patients with CCR5-tropic virus. It has two dose formulations (150- and 300-mg tablets) and can be taken with or without food. Marviroc has received regulatory approvals, and vicriviroc is in phase 3 trials.

ExcerptReferenceRelevance
"Maraviroc has an in vitro activity against most HIV-1C viruses and could be considered as an alternative regimen in individuals infected with CCR5-tropic HIV-1C viruses."( Coreceptor Tropism and Maraviroc Sensitivity of Clonally Derived Ethiopian HIV-1C Strains Using an in-house Phenotypic Assay and Commonly Used Genotypic Methods.
Aralaguppe, SG; Fekade, D; Gebre-Selassie, S; Kalu, AW; Marrone, G; Sonnerborg, A; Telele, NF, 2018
)
1.51
"Maraviroc has an in vitro activity against most HIV-1C viruses and could be considered as an alternative regimen in individuals infected with CCR5-tropic HIV-1C viruses."( Coreceptor Tropism and Maraviroc Sensitivity of Clonally Derived Ethiopian HIV-1C Strains Using an in-house Phenotypic Assay and Commonly Used Genotypic Methods.
Aralaguppe, SG; Fekade, D; Gebre-Selassie, S; Kalu, AW; Marrone, G; Sonnerborg, A; Telele, NF, 2018
)
1.51
"Maraviroc has six impurities."( A validated stability-indicating UPLC method for the determination of impurities in Maraviroc.
Chilukuri, M; Hussainreddy, K; Narayanareddy, P; Venkataramana, M, 2014
)
1.35
"Maraviroc has been proved to be effective for anti-HIV-1 by targeting CCR5."( Computational study on the interaction between CCR5 and HIV-1 entry inhibitor maraviroc: insight from accelerated molecular dynamics simulation and free energy calculation.
Bai, Q; Chen, W; Li, X; Liu, H; Yao, X; Zhang, Y, 2014
)
1.35
"Maraviroc has been shown to have potent anti-inflammatory effects."( Maraviroc-Mediated Lung Protection following Trauma-Hemorrhagic Shock.
Liu, FC; Yu, HP; Zheng, CW, 2016
)
2.6
"Maraviroc has received regulatory approvals, and vicriviroc is in phase 3 trials."( A pièce de resistance: how HIV-1 escapes small molecule CCR5 inhibitors.
Kuritzkes, DR; Moore, JP, 2009
)
1.07
"Maraviroc has two dose formulations (150- and 300-mg tablets) and can be taken with or without food."( Maraviroc: a new CCR5 antagonist.
Khanlou, H; Sayana, S, 2009
)
2.52
"Maraviroc (MVC) has shown good efficacy and tolerability in treatment-naive and treatment-experienced HIV-1-infected patients with CCR5-tropic virus. "( Responses to switching to maraviroc-based antiretroviral therapy in treated patients with suppressed plasma HIV-1-RNA load.
Brudney, D; Garcia, A; Geretti, AM; Johnson, M; MacCartney, M; Marshall, N; Smith, C; Vitiello, P, 2012
)
2.12
"Maraviroc (MVC) use has trailed that of other post-2006 antiretroviral therapy (ART) options for treatment-experienced patients. "( Maraviroc observational study: the impact of expanded resistance testing and clinical considerations for antiretroviral regimen selection in treatment-experienced patients.
Mugavero, MJ; Napolitano, LA; Nevin, CR; Raper, JL; Saag, MS; Tamhane, A; Wilkins, SA; Willig, JH, 2013
)
3.28
"Maraviroc has no detectable in vitro cytotoxicity and is highly selective for CCR5, as confirmed against a wide range of receptors and enzymes, including the hERG ion channel (50% inhibitory concentration, >10 microM), indicating potential for an excellent clinical safety profile."( Maraviroc (UK-427,857), a potent, orally bioavailable, and selective small-molecule inhibitor of chemokine receptor CCR5 with broad-spectrum anti-human immunodeficiency virus type 1 activity.
Armour, D; Dobbs, S; Dorr, P; Griffin, P; Irvine, B; Macartney, M; Mori, J; Napier, C; Perros, M; Price, D; Rickett, G; Smith-Burchnell, C; Stammen, B; Webster, R; Westby, M; Wood, A, 2005
)
2.49
"Maraviroc has demonstrated in vitro activity against a wide range of CCR5 tropic clinical isolates, including those resistant to the four currently existing drug classes of antiretroviral agents."( Maraviroc.
Fadel, H; Temesgen, Z, 2007
)
2.5
"Maraviroc has shown its clinical efficacy in patients failing other antiretroviral classes."( [Maraviroc: clinical trials results].
Chidiac, C; Katlama, C; Yeni, P, 2008
)
1.98

Actions

Maraviroc induced an increase in cell-associated HIV RNA during the administration of the drug. The drug does not inhibit any of the major CYP450 enzymes at clinically relevant doses and it has not shown any clinically relevant effects on plasma concentrations of other agents.

ExcerptReferenceRelevance
"Maraviroc induced an increase in cell-associated HIV RNA during the administration of the drug."( Prolonged administration of maraviroc reactivates latent HIV in vivo but it does not prevent antiretroviral-free viral rebound.
Delgado, R; Gutiérrez, C; Hernández-Novoa, B; López-Huertas, MR; Madrid-Elena, N; Moreno, S; Muñoz-Fernández, MÁ; Olalla-Sierra, J; Plana, M; Rubio, R, 2020
)
1.57
"Maraviroc does not inhibit any of the major CYP450 enzymes at clinically relevant doses and it has not shown any clinically relevant effects on plasma concentrations of other agents; hence, no dose adjustments of coadministered agents are required."( Maraviroc: pharmacokinetics and drug interactions.
Abel, S; Back, DJ; Vourvahis, M, 2009
)
2.52

Treatment

Maraviroc treatment reduced 2B11.3-induced endocapillary hypercellularity and improved serum blood urea nitrogen levels. Treatment for HIV-1 infected patients results in larger CD4(+) T cell rises than are attributable to its antiviral activity alone.

ExcerptReferenceRelevance
"Maraviroc treatment attenuated oxycodone-seeking in abstinent rats and reduced functional coupling in the reward circuitry. "( The utility of maraviroc, an antiretroviral agent used to treat HIV, as treatment for opioid abuse? Data from MRI and behavioural testing in rats.
Borges, C; Cai, X; Ferris, CF; Iriah, SC; Kulkarni, PP; Madularu, D; Shalev, U, 2021
)
2.42
"Maraviroc treatment reduced 2B11.3-induced endocapillary hypercellularity and improved serum blood urea nitrogen levels."( CCR2- and CCR5-mediated macrophage infiltration contributes to glomerular endocapillary hypercellularity in antibody-induced lupus nephritis.
Baba, T; Ishida, Y; Kawano, M; Mukaida, N; Nagata, M; Nakatani, K; Tanabe, Y; Zoshima, T, 2022
)
1.44
"Maraviroc pre-treatment (CCR5 antagonist, 40uM) blunted CCL5-induced Nox1 expression."( CCR5 antagonist treatment inhibits vascular injury by regulating NADPH oxidase 1.
Belin de Chantemele, EJ; Bruder-Nascimento, A; Bruder-Nascimento, T; Singh, S, 2022
)
1.44
"Maraviroc treatment reduced tumor volume, CAF recruitment, and ERK1/2 signaling in vivo, thus, mimicking the effects observed with genetic loss of CCL5."( Tumor-Derived CCL5 Recruits Cancer-Associated Fibroblasts and Promotes Tumor Cell Proliferation in Esophageal Squamous Cell Carcinoma.
Diehl, JA; Dunbar, KJ; Efe, G; Harris, M; Henick, B; Hu, J; Karakasheva, TA; Klein-Szanto, AJ; Lin, EW; Nakagawa, H; Rustgi, AK; Sachdeva, UM; Sahu, V; Tang, Q, 2023
)
1.63
"Maraviroc treatment also increased apoptosis and modified cytoskeletal organization."( CR5/CCL5 axis is linked to a poor outcome, and inhibition reduces metastasis in oral squamous cell carcinoma.
Alcayaga-Miranda, F; Coletta, RD; García, C; García, IE; Godínez-Pacheco, B; González-Arriagada, WA; Lozano-Burgos, C; Maripillán, J; Martínez-Flores, R; Oyarce-Pezoa, S, 2023
)
1.63
"Maraviroc treatment should not be interrupted if alcohol is consumed."( Interactions between alcohol and the HIV entry inhibitor Maraviroc.
Aweeka, FT; Beatty, GW; Gruber, VA; Lum, PJ; McCance-Katz, EF; Rainey, PM,
)
1.82
"Maraviroc treatment also increased hepatic PPAR(γ) expression compared with vehicle-treated trauma-hemorrhaged rats."( Maraviroc attenuates trauma-hemorrhage-induced hepatic injury through PPAR gamma-dependent pathway in rats.
Liu, FC; Tsai, YF; Yu, HP, 2013
)
2.55
"Maraviroc-treated patients had a greater increase from baseline in mean CD4(+) cell count than efavirenz-treated patients at week 240 (293 vs."( Efficacy and safety of maraviroc vs. efavirenz in treatment-naive patients with HIV-1: 5-year findings.
Botes, M; Burnside, R; Clumeck, N; Cooper, DA; Dejesus, E; Heera, J; Ive, P; Lazzarin, A; Mukwaya, G; Saag, M; van Der Ryst, E; Walmsley, S, 2014
)
1.43
"Maraviroc treatment also decreased lung tissue damage as compared to the vehicle-treated trauma-hemorrhaged rats."( Maraviroc-Mediated Lung Protection following Trauma-Hemorrhagic Shock.
Liu, FC; Yu, HP; Zheng, CW, 2016
)
2.6
"Maraviroc treatment for HIV-1 infected patients results in larger CD4(+) T cell rises than are attributable to its antiviral activity alone. "( Effects of maraviroc and efavirenz on markers of immune activation and inflammation and associations with CD4+ cell rises in HIV-infected patients.
Eason, J; Funderburg, N; Goodrich, J; Heera, J; Kalinowska, M; Lederman, MM; Mayer, H; Rajicic, N; Valdez, H, 2010
)
2.19
"Maraviroc treatment tended to result in more rapid decreases in CD38 expression on CD4(+) T cells and in plasma D-dimer concentrations than did treatment with efavirenz."( Effects of maraviroc and efavirenz on markers of immune activation and inflammation and associations with CD4+ cell rises in HIV-infected patients.
Eason, J; Funderburg, N; Goodrich, J; Heera, J; Kalinowska, M; Lederman, MM; Mayer, H; Rajicic, N; Valdez, H, 2010
)
1.47
"Maraviroc-treated patients had earlier, modest decreases in certain markers of immune activation and inflammation, although in this small study, many of the differences were not statistically significant. "( Effects of maraviroc and efavirenz on markers of immune activation and inflammation and associations with CD4+ cell rises in HIV-infected patients.
Eason, J; Funderburg, N; Goodrich, J; Heera, J; Kalinowska, M; Lederman, MM; Mayer, H; Rajicic, N; Valdez, H, 2010
)
2.19
"Pretreatment with maraviroc (1, 2.5, 5 mg/kg, IP) reduced hyperlocomotion induced by acute cocaine (10 mg/kg) without affecting spontaneous locomotor activity."( Chemokine CCR5 and cocaine interactions in the brain: Cocaine enhances mesolimbic CCR5 mRNA levels and produces place preference and locomotor activation that are reduced by a CCR5 antagonist.
Cicalese, S; Nayak, SU; Oliver, CF; Rawls, SM; Tallarida, C, 2020
)
0.88
"Treatment with maraviroc significantly inhibited CRC liver metastasis in the animal model."( Antineoplastic effects of targeting CCR5 and its therapeutic potential for colorectal cancer liver metastasis.
Adwan, H; Berger, MR; Bergmann, F; Faiza, S; Georges, R; Mahmood, S; Pervaiz, A; Zepp, M, 2021
)
0.96
"Pretreatment with maraviroc blocked the effects of Tat, reinstating morphine potency in non-tolerant mice and restoring withdrawal symptomology in morphine-tolerant mice."( CCR5 mediates HIV-1 Tat-induced neuroinflammation and influences morphine tolerance, dependence, and reward.
Akbarali, HI; Dewey, WL; Gonek, M; Hauser, KF; Knapp, PE; Lippold, K; McLane, VD; Paris, JJ; Stevens, DL, 2018
)
0.8
"Treatment with Maraviroc resulted in a block in S phase of LX-2 cells with increased expression levels of cyclin D1 and p21 while the expression of p53 was reduced."( Effects of treatment with Maraviroc a CCR5 inhibitor on a human hepatic stellate cell line.
Carleo, MA; Coppola, N; De Luca, A; Esposito, V; Guerra, G; Lucariello, A; Macera, M; Martini, S; Perna, A, 2018
)
1.12

Toxicity

Maraviroc is a generally safe and well-tolerated medication for the treatment of HIV-1 infection with a unique mechanism of action. The adverse event (AE) profile was compared with the active and placebo arms of the maraviroC phase III clinical trials MOTIVATE 1 and 2.

ExcerptReferenceRelevance
" Finally, the third CCR5 antagonist vicriviroc also showed long-term potent viral activity in phase II studies as long as it was boosted with low-dose ritonavir, with no significant differences in grade 3 and grade 4 adverse effects compared to placebo."( CCR5 antagonists: comparison of efficacy, side effects, pharmacokinetics and interactions--review of the literature.
Emmelkamp, JM; Rockstroh, JK, 2007
)
0.34
" There were no serious adverse events in any of these studies, and doses up to 900 mg were generally well tolerated, with postural hypotension being the dose-limiting event."( Assessment of the pharmacokinetics, safety and tolerability of maraviroc, a novel CCR5 antagonist, in healthy volunteers.
Abel, S; Medhurst, CG; Muirhead, GJ; Ridgway, CE; Rosario, MC; Taylor-Worth, RJ; van der Ryst, E, 2008
)
0.59
" It does not significantly influence the activity of major drug-metabolizing enzymes and is well tolerated at clinically relevant doses, with most adverse events being mild or moderate."( Assessment of the pharmacokinetics, safety and tolerability of maraviroc, a novel CCR5 antagonist, in healthy volunteers.
Abel, S; Medhurst, CG; Muirhead, GJ; Ridgway, CE; Rosario, MC; Taylor-Worth, RJ; van der Ryst, E, 2008
)
0.59
" Exposure-adjusted rates of adverse events were similar with maraviroc or placebo."( Two-year safety and virologic efficacy of maraviroc in treatment-experienced patients with CCR5-tropic HIV-1 infection: 96-week combined analysis of MOTIVATE 1 and 2.
Fätkenheuer, G; Goodrich, J; Gulick, RM; Hardy, WD; Heera, J; Mayer, H; Nelson, M; Rajicic, N, 2010
)
0.87
" Maraviroc recipients had greater CD4 increases (+ 212 vs + 171 cells/mm(3)) and fewer adverse event discontinuations (6."( Efficacy and safety of maraviroc versus efavirenz, both with zidovudine/lamivudine: 96-week results from the MERIT study.
Burnside, R; Craig, C; Di Perri, G; Frank, I; Goodrich, J; Heera, J; Mayer, H; McCracken, J; Pontani, D; Saag, M; Sierra-Madero, J; Wood, R,
)
1.35
" It is generally well tolerated, with a similar side-effect profile to placebo in controlled studies."( Hepatic safety and tolerability in the maraviroc clinical development program.
Alston, S; Ayoub, A; Goodrich, J; Heera, J; Hoepelman, AI; Lalezari, J; Mayer, H; Mchale, M; Nelson, M; van der Ryst, E, 2010
)
0.63
"Maraviroc is a generally safe and well-tolerated medication for the treatment of HIV-1 infection with a unique mechanism of action."( Drug safety evaluation of maraviroc for the treatment of HIV infection.
Hardy, WD; Rockstroh, JK; Wasmuth, JC, 2012
)
2.12
" The adverse event (AE) profile was compared with the active and placebo arms of the maraviroc phase III clinical trials MOTIVATE 1 and 2, where the OBT did not include these agents."( Safety profile of maraviroc in patients coinfected with HIV-1 and hepatitis B or C included in the maraviroc expanded access program.
Heera, J; Lazzarin, A; Mukwaya, G; Than, S; Valluri, SR,
)
0.69
" 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
" Adverse event (AE) and treatment-related AE incidence was 91."( Safety and immunovirologic outcomes with maraviroc combination regimens in patients with a history of past treatment failures and virologic resistance in Brazil: an open-label, multicenter phase 3b study.
Bicudo, EL; Cassoli, LM; da Eira, M; de Andrade Neto, JL; Furtado, J; Leite, OH; Lewi, DS; Lima, MP; Lopes, MI; Machado, AA; Madruga, JV; Miranda, AF; Netto, EM; Pedro, Rde J; Portsmouth, S; Santini-Oliveira, M; Santos, BR; Tupinambas, U; Wajsbrot, DB, 2013
)
0.66
"Maraviroc was generally safe in treatment-experienced participants for >5 years."( Five-year safety evaluation of maraviroc in HIV-1-infected treatment-experienced patients.
Burnside, R; Fatkenheuer, G; Goodrich, J; Gulick, RM; Hardy, WD; Heera, JR; Mukwaya, G; Nelson, MR; Portsmouth, S, 2014
)
2.13
" The frequency of serious adverse effects was investigated."( Efficacy and safety of once-daily maraviroc plus ritonavir-boosted darunavir in pretreated HIV-infected patients in a real-life setting.
Bancalero, P; Chueca, N; García, C; Jiménez, P; Macías, J; Márquez, M; Merino, D; Muñoz, L; Ojeda, G; Pasquau, J; Pineda, JA; Recio, E, 2014
)
0.68
" Reasons for starting MVC plus DRV/r were: adverse effects in 38 individuals (63%), simplification in 15 (25%) and virological failure in seven (12%)."( Efficacy and safety of once-daily maraviroc plus ritonavir-boosted darunavir in pretreated HIV-infected patients in a real-life setting.
Bancalero, P; Chueca, N; García, C; Jiménez, P; Macías, J; Márquez, M; Merino, D; Muñoz, L; Ojeda, G; Pasquau, J; Pineda, JA; Recio, E, 2014
)
0.68
"Most individuals starting MVC plus DRV/r qd because of simplification or adverse effects maintained HIV suppression after 48 weeks of follow-up."( Efficacy and safety of once-daily maraviroc plus ritonavir-boosted darunavir in pretreated HIV-infected patients in a real-life setting.
Bancalero, P; Chueca, N; García, C; Jiménez, P; Macías, J; Márquez, M; Merino, D; Muñoz, L; Ojeda, G; Pasquau, J; Pineda, JA; Recio, E, 2014
)
0.68
" MVC was well tolerated with no grade 3/4 adverse events; all subjects maintained viral suppression to the end of the study."( Switching safely: pharmacokinetics, efficacy and safety of switching efavirenz to maraviroc twice daily in patients on suppressive antiretroviral therapy.
Back, D; Boffito, M; Else, L; Gazzard, B; Jackson, A; Nelson, M; Newell, S; Rockwood, N; Waters, L, 2015
)
0.64
" efavirenz experienced treatment-related adverse events (68."( Efficacy and safety of maraviroc vs. efavirenz in treatment-naive patients with HIV-1: 5-year findings.
Botes, M; Burnside, R; Clumeck, N; Cooper, DA; Dejesus, E; Heera, J; Ive, P; Lazzarin, A; Mukwaya, G; Saag, M; van Der Ryst, E; Walmsley, S, 2014
)
0.71
"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,
)
0.69
" No subjects met protocol-defined liver stopping criteria and there were no cases of Hy's law or treatment-related hepatobiliary serious adverse events."( Hepatic safety in subjects with HIV-1 and hepatitis C and/or B virus: a randomized, double-blind study of maraviroc versus placebo in combination with antiretroviral agents.
Asmuth, DM; Bansal, M; Fätkenheuer, G; Heera, J; Jagannatha, S; Mukwaya, G; Pialoux, G; Pineda, JA; Plonski, F; Rockstroh, JK; Small, CB; Soriano, V,
)
0.34
" Safety was assessed by adverse events."( Phase 1 Safety, Pharmacokinetics, and Pharmacodynamics of Dapivirine and Maraviroc Vaginal Rings: A Double-Blind Randomized Trial.
Chen, BA; Dezzutti, CS; Hendrix, CW; Hoesley, CJ; Husnik, MJ; Johnson, S; Marzinke, MA; Nel, A; Panther, L; Rabe, LK; Richardson-Harman, N; Soto-Torres, L; van der Straten, A, 2015
)
0.65
"There was no difference in related genitourinary adverse events between treatment arms compared with placebo."( Phase 1 Safety, Pharmacokinetics, and Pharmacodynamics of Dapivirine and Maraviroc Vaginal Rings: A Double-Blind Randomized Trial.
Chen, BA; Dezzutti, CS; Hendrix, CW; Hoesley, CJ; Husnik, MJ; Johnson, S; Marzinke, MA; Nel, A; Panther, L; Rabe, LK; Richardson-Harman, N; Soto-Torres, L; van der Straten, A, 2015
)
0.65
"In this first study of a combination microbicide vaginal ring, all 4 rings were safe and well tolerated."( Phase 1 Safety, Pharmacokinetics, and Pharmacodynamics of Dapivirine and Maraviroc Vaginal Rings: A Double-Blind Randomized Trial.
Chen, BA; Dezzutti, CS; Hendrix, CW; Hoesley, CJ; Husnik, MJ; Johnson, S; Marzinke, MA; Nel, A; Panther, L; Rabe, LK; Richardson-Harman, N; Soto-Torres, L; van der Straten, A, 2015
)
0.65
" We recorded the ORs for dual-therapy versus control groups for serious adverse events (1·16 [0·92-1·48]), adverse events (0·82 [0·52-1·28]), and mutations (2·11 [1·32-3·36])."( Efficacy and safety of contemporary dual-drug antiretroviral regimens as first-line treatment or as a simplification strategy: a systematic review and meta-analysis.
Achhra, AC; Amin, J; Boyd, MA; Mwasakifwa, G, 2016
)
0.43
"Dual therapy, especially with regimens excluding maraviroc, could be safe and efficacious, particularly in patients with baseline viral loads of less than 100 000 copies per mL."( Efficacy and safety of contemporary dual-drug antiretroviral regimens as first-line treatment or as a simplification strategy: a systematic review and meta-analysis.
Achhra, AC; Amin, J; Boyd, MA; Mwasakifwa, G, 2016
)
0.69
" Rates of grade 3-4 adverse events did not differ among regimens (P = ."( Phase 2 Study of the Safety and Tolerability of Maraviroc-Containing Regimens to Prevent HIV Infection in Men Who Have Sex With Men (HPTN 069/ACTG A5305).
Amico, KR; Andrade, A; Andrew, P; Chege, W; Chen, YQ; Cottle, LM; Eshleman, SH; Farley, JE; Frank, I; Gulick, RM; Hendrix, CW; Ho, K; Klingman, KL; Landovitz, RJ; Liu, A; Magnus, M; Marcus, C; Marzinke, MA; Mayer, KH; McCauley, M; McGowan, I; Richardson, P; Rinehart, AR; Rooney, JF; Salata, RA; Santana, J; Stekler, JD; Wilkin, TJ; Young, AM, 2017
)
0.71
"MVC-containing regimens were safe and well tolerated compared with TDF + FTC; this study was not powered for efficacy."( Phase 2 Study of the Safety and Tolerability of Maraviroc-Containing Regimens to Prevent HIV Infection in Men Who Have Sex With Men (HPTN 069/ACTG A5305).
Amico, KR; Andrade, A; Andrew, P; Chege, W; Chen, YQ; Cottle, LM; Eshleman, SH; Farley, JE; Frank, I; Gulick, RM; Hendrix, CW; Ho, K; Klingman, KL; Landovitz, RJ; Liu, A; Magnus, M; Marcus, C; Marzinke, MA; Mayer, KH; McCauley, M; McGowan, I; Richardson, P; Rinehart, AR; Rooney, JF; Salata, RA; Santana, J; Stekler, JD; Wilkin, TJ; Young, AM, 2017
)
0.71
" One (maraviroc) and two (placebo) patients experienced treatment-related hepatobiliary adverse events (AEs)."( Hepatic safety of maraviroc in patients with HIV-1 and hepatitis C and/or B virus: 144-week results from a randomized, placebo-controlled trial.
Asmuth, DM; Bansal, M; Fätkenheuer, G; Heera, J; Pialoux, G; Pineda, JA; Plonski, F; Rockstroh, JK; Small, CB; Wang, R; Zhang-Roper, R, 2017
)
1.27
" Primary outcomes were grade 3 and 4 adverse events and time to permanent discontinuation of the study regimen."( Safety and Tolerability of Maraviroc-Containing Regimens to Prevent HIV Infection in Women: A Phase 2 Randomized Trial.
Amico, KR; Andrade, A; Andrew, P; Chege, W; Chen, YQ; Cottle, LM; Eshleman, SH; Frank, I; Gulick, RM; Hendrix, CW; Ho, K; Hodder, S; Klingman, KL; Landovitz, RJ; Manabe, YC; Marcus, C; Marzinke, MA; Mayer, KH; McCauley, M; McGowan, I; Richardson, P; Rinehart, AR; Rooney, JF; Salata, RA; Santana, J; Siegel, M; Stekler, JD; Swaminathan, S; Wilkin, TJ; Young, AM, 2017
)
0.75
" Grade 3 or 4 adverse events occurred in 5 (MVC), 13 (MVC-FTC), 9 (MVC-TDF), and 8 (TDF-FTC) participants; rates did not differ among regimens."( Safety and Tolerability of Maraviroc-Containing Regimens to Prevent HIV Infection in Women: A Phase 2 Randomized Trial.
Amico, KR; Andrade, A; Andrew, P; Chege, W; Chen, YQ; Cottle, LM; Eshleman, SH; Frank, I; Gulick, RM; Hendrix, CW; Ho, K; Hodder, S; Klingman, KL; Landovitz, RJ; Manabe, YC; Marcus, C; Marzinke, MA; Mayer, KH; McCauley, M; McGowan, I; Richardson, P; Rinehart, AR; Rooney, JF; Salata, RA; Santana, J; Siegel, M; Stekler, JD; Swaminathan, S; Wilkin, TJ; Young, AM, 2017
)
0.75
" Safety was assessed by adverse events (AEs), colposcopy, and culture-independent analysis of the vaginal microbiome (VMB)."( Safety and pharmacokinetics of single, dual, and triple antiretroviral drug formulations delivered by pod-intravaginal rings designed for HIV-1 prevention: A Phase I trial.
Anton, PA; Baum, MM; Butkyavichene, I; Churchman, SA; Cortez, JM; Dawson, L; Fanter, R; Gunawardana, M; Guthrie, KM; Hendrix, CW; Marzinke, MA; Miller, CS; Moss, JA; Olive, TJ; Pyles, RB; Rosen, RK; Vargas, SE; Vincent, KL; Yang, F, 2018
)
0.48
"An innovative pod-IVR delivery device with 3 different formulations delivering different regimens of ARV drugs vaginally appeared to be safe and acceptable and provided drug concentrations in CVFs and tissues exceeding concentrations achieved by highly protective oral dosing, suggesting that efficacy for vaginal HIV PrEP is achievable."( Safety and pharmacokinetics of single, dual, and triple antiretroviral drug formulations delivered by pod-intravaginal rings designed for HIV-1 prevention: A Phase I trial.
Anton, PA; Baum, MM; Butkyavichene, I; Churchman, SA; Cortez, JM; Dawson, L; Fanter, R; Gunawardana, M; Guthrie, KM; Hendrix, CW; Marzinke, MA; Miller, CS; Moss, JA; Olive, TJ; Pyles, RB; Rosen, RK; Vargas, SE; Vincent, KL; Yang, F, 2018
)
0.48
" Triple combinations resulted toxic and were rejected."( Maraviroc reactivates HIV with potency similar to that of other latency reversing drugs without inducing toxicity in CD8 T cells.
Gutiérrez, C; Jiménez-Tormo, L; López-Huertas, MR; Luna, L; Madrid-Elena, N; Moreno, S; Vivancos, MJ, 2020
)
2
" There were 25 adverse events, of which 24 were Grade 1 (G1) and one was G2 (unrelated)."( A Randomized, Open-Label, Crossover Phase 1 Safety and Pharmacokinetic Study of Oral Maraviroc and Maraviroc 1% Gel (the CHARM-03 Study).
Abebe, KZ; Achilles, SL; Anton, PA; Brand, RM; Chawki, S; Chen, B; Cranston, RD; Engstrom, JC; Hendrix, CW; Jacobson, CE; Khanukova, E; Marzinke, MA; McGowan, IM; Reinhart, A; Richardson-Harman, N; Rohan, LC; Siegel, A; Spiegel, H; Stall, R; Steytler, J, 2022
)
0.95

Pharmacokinetics

The Combination HIV Antiretroviral Rectal Microbicide-3 (CHARM-03) study was a randomized, open-label, phase 1 study. The study aimed to develop a population pharmacokinetic model to characterize the disposition of maraviroc in neonates.

ExcerptReferenceRelevance
" The integration of these models in a pharmacokinetic (PK)-pharmacodynamic (PD)-disease model can help toward a better understanding of the complexity of the interactions, as well as in the identification and clarification of the current model assumptions."( A pharmacokinetic-pharmacodynamic disease model to predict in vivo antiviral activity of maraviroc.
Dorr, P; Hitchcock, C; Jacqmin, P; Rosario, MC; van der Ryst, E, 2005
)
0.55
" An integrated pharmacokinetic-pharmacodynamic model was developed using the mixed effects modeling approach with patients' pharmacokinetic profiles on the last day of treatment, HIV-1 RNA levels over time, and the individual viral susceptibility."( A pharmacokinetic-pharmacodynamic model to optimize the phase IIa development program of maraviroc.
Poland, B; Rosario, MC; Sullivan, J; van der Ryst, E; Westby, M, 2006
)
0.56
"To assess the potential of known CYP3A4 inducers, with and without CYP3A4 inhibitors, to alter the pharmacokinetic profile of maraviroc."( Effects of CYP3A4 inducers with and without CYP3A4 inhibitors on the pharmacokinetics of maraviroc in healthy volunteers.
Abel, S; Jenkins, TM; Muirhead, GJ; Ridgway, CE; Whitlock, LA, 2008
)
0.77
" No pharmacokinetic data are reported for cohort 3 because all subjects were discontinued during period 1 due to poor toleration of the drug regimen."( Effects of CYP3A4 inducers with and without CYP3A4 inhibitors on the pharmacokinetics of maraviroc in healthy volunteers.
Abel, S; Jenkins, TM; Muirhead, GJ; Ridgway, CE; Whitlock, LA, 2008
)
0.57
"To assess the potential of cotrimoxazole and tenofovir, drugs which are inhibitors and/or substrates of renal transporters, to alter the pharmacokinetic profile of maraviroc."( The effects of cotrimoxazole or tenofovir co-administration on the pharmacokinetics of maraviroc in healthy volunteers.
Abel, S; Muirhead, GJ; Ridgway, CE; Russell, D; Whitlock, LA, 2008
)
0.76
" Plasma pharmacokinetic parameters from this study were compared with historical data generated in HIV-positive subjects receiving maraviroc monotherapy in a Phase IIa study."( A novel probe drug interaction study to investigate the effect of selected antiretroviral combinations on the pharmacokinetics of a single oral dose of maraviroc in HIV-positive subjects.
Boffito, M; Muirhead, GJ; Pozniak, AL; Ridgway, CE; Russell, D, 2008
)
0.75
"To develop a population pharmacokinetic model for maraviroc, a noncompetitive CCR5 antagonist, after oral administration of tablets to healthy volunteers and asymptomatic HIV-infected subjects and to quantify the inherent variability and influence of covariates on the parameters of the model."( A population pharmacokinetic meta-analysis of maraviroc in healthy volunteers and asymptomatic HIV-infected subjects.
Chan, PL; McFadyen, L; Weatherley, B, 2008
)
0.86
"Rich pharmacokinetic data available from 15 studies in healthy volunteers (n = 365) and two studies in asymptomatic HIV-infected subjects (n = 48) were analysed using NONMEM."( A population pharmacokinetic meta-analysis of maraviroc in healthy volunteers and asymptomatic HIV-infected subjects.
Chan, PL; McFadyen, L; Weatherley, B, 2008
)
0.6
" AIMS To develop a population pharmacokinetic (PK)-pharmacodynamic (PD) model that describes CCR5 receptor occupancy by maraviroc after oral administration at different doses in healthy volunteers and HIV-positive patients and to assess the relevance of receptor occupancy in predicting the decrease in viral load (HIV-1 RNA copies ml(-1)) in HIV-positive patients."( Population pharmacokinetic/pharmacodynamic analysis of CCR5 receptor occupancy by maraviroc in healthy subjects and HIV-positive patients.
Abel, S; Dorr, P; Jacqmin, P; James, I; Jenkins, TM; Rosario, MC; van der Ryst, E, 2008
)
0.78
"To model the basic pharmacokinetic (PK) characteristics of maraviroc to construct an integrated semi-mechanistic PK model for use in later population PK analyses."( Maraviroc modelling strategy: use of early phase 1 data to support a semi-mechanistic population pharmacokinetic model.
McFadyen, L; Weatherley, B, 2009
)
2.04
"The pharmacokinetic (PK) interaction between ritonavir-boosted elvitegravir (elvitegravir/r) and maraviroc was evaluated."( Pharmacokinetic interaction of ritonavir-boosted elvitegravir and maraviroc.
Abel, S; Hui, J; Kearney, BP; Ramanathan, S; Tweedy, S; West, S, 2010
)
0.82
" Lack of PK alteration was defined as 90% confidence intervals for ratio of geometric least squares means ratio (coadministration:alone) between 70% and 143% for elvitegravir and ritonavir Cmax (maximum concentration), Ctau (trough), and AUCtau (area under plasma concentration-time curve; 0-24 hours); for maraviroc, given a 100% increase in Cmax and AUCtau (0-12 hours); the predicted 90% confidence intervals were 162% to 247% and 136% to 295%, respectively."( Pharmacokinetic interaction of ritonavir-boosted elvitegravir and maraviroc.
Abel, S; Hui, J; Kearney, BP; Ramanathan, S; Tweedy, S; West, S, 2010
)
0.77
" * As both these drugs are likely to be used in combination, this study evaluated the pharmacokinetic interaction between them."( Assessment of the pharmacokinetics of co-administered maraviroc and raltegravir.
Andrews, E; Crownover, P; Damle, B; Fang, J; Glue, P; Tressler, R, 2010
)
0.61
"To assess the two-way pharmacokinetic interaction between maraviroc and raltegravir."( Assessment of the pharmacokinetics of co-administered maraviroc and raltegravir.
Andrews, E; Crownover, P; Damle, B; Fang, J; Glue, P; Tressler, R, 2010
)
0.85
" Test/reference ratios and 95% confidence intervals (CIs) were determined for pharmacokinetic parameters."( Assessment of the pharmacokinetics of co-administered maraviroc and raltegravir.
Andrews, E; Crownover, P; Damle, B; Fang, J; Glue, P; Tressler, R, 2010
)
0.61
" The pharmacokinetic profile of the HIV entry inhibitor maraviroc after a single intrapartum dose in rhesus macaques was studied to determine whether maraviroc could serve as an alternative to NVP in a single-dose strategy."( Maternal-fetal pharmacokinetics and dynamics of a single intrapartum dose of maraviroc in rhesus macaques.
Holodniy, M; Kashuba, AD; Shulman, NS; Van Rompay, KK; Winters, MA, 2010
)
0.84
" For the given maraviroc monotherapy sampling design, it was difficult to separate the viral dynamics system delay from a pharmacokinetic distributional delay or delay due to receptor binding and subsequent cellular signalling."( The use of the SAEM algorithm in MONOLIX software for estimation of population pharmacokinetic-pharmacodynamic-viral dynamics parameters of maraviroc in asymptomatic HIV subjects.
Chan, PL; Jacqmin, P; Lavielle, M; McFadyen, L; Weatherley, B, 2011
)
0.92
"A pharmacokinetic study was performed in 12 HIV-negative men receiving maraviroc 300 mg twice daily for 8 days."( Single and multiple dose pharmacokinetics of maraviroc in saliva, semen, and rectal tissue of healthy HIV-negative men.
Brown, KC; Cohen, MS; Dumond, JB; Heidt, PE; Kashuba, AD; Malone, SA; Patterson, KB; Prince, HM; Shaheen, NJ; Spacek, MB, 2011
)
0.86
"We conducted a pharmacokinetic and in vivo cerebral (1)H magnetic resonance spectroscopy ((1)H-MRS) study to assess CSF exposure and cerebral metabolite ratios (CMRs) following maraviroc intensification."( CNS effects of a CCR5 inhibitor in HIV-infected subjects: a pharmacokinetic and cerebral metabolite study.
Allsop, JM; Back, D; Erlwein, OW; Garvey, L; Kaye, S; Latch, N; Mitchell, A; Nelson, M; Taylor-Robinson, SD; Watson, V; Winston, A, 2012
)
0.57
" More comprehensive in vivo pharmacokinetic data are required to justify the potential use of these agents as safe and effective options during pregnancy."( Pharmacokinetics of antiretroviral drugs in anatomical sanctuary sites: the fetal compartment (placenta and amniotic fluid).
Back, DJ; Else, LJ; Khoo, SH; Taylor, S, 2011
)
0.37
" However, the pharmacokinetic profiles of such regimens are often not established."( Pharmacokinetic profile and safety of 150 mg of maraviroc dosed with 800/100 mg of darunavir/ritonavir all once daily, with and without nucleoside analogues, in HIV-infected subjects.
Back, D; Croucher, A; Else, LJ; Khoo, S; Mora-Peris, B; Scullard, G; Vera, JH; Winston, A, 2013
)
0.65
" At steady-state (days 10 and 20), intensive pharmacokinetic sampling was undertaken."( Pharmacokinetic profile and safety of 150 mg of maraviroc dosed with 800/100 mg of darunavir/ritonavir all once daily, with and without nucleoside analogues, in HIV-infected subjects.
Back, D; Croucher, A; Else, LJ; Khoo, S; Mora-Peris, B; Scullard, G; Vera, JH; Winston, A, 2013
)
0.65
"To address the need for nucleos(t)ide reverse transcriptase inhibitor (NRTI)-sparing regimens, we explored the virologic and pharmacokinetic characteristics of maraviroc plus ritonavir-boosted darunavir in a single-arm, open-label, 96-week study."( Virologic response, early HIV-1 decay, and maraviroc pharmacokinetics with the nucleos(t)ide-free regimen of maraviroc plus darunavir/ritonavir in a pilot study.
Acosta, EP; Adeyemi, O; Berzins, B; Castro, J; Eron, JJ; Kuritzkes, DR; Lalezari, J; Lu, D; Ryscavage, P; Swindells, S; Taiwo, B; Tsibris, A, 2013
)
0.85
"This open-label, fixed-sequence, phase 1 study evaluated the pharmacokinetic interaction between maraviroc (MVC) and ritonavir-boosted fosamprenavir (FPV/r) in healthy subjects."( Pharmacokinetic interaction between maraviroc and fosamprenavir-ritonavir: an open-label, fixed-sequence study in healthy subjects.
Fang, A; Heera, J; Mendes da Costa, L; Plotka, A; Vourvahis, M, 2013
)
0.88
" This study evaluated whether a pharmacokinetic interaction exists between these agents."( Lack of a significant pharmacokinetic interaction between maraviroc and tacrolimus in allogeneic HSCT recipients.
Ganetsky, A; Hughes, ME; Miano, TA; Porter, DL; Reshef, R; Vonderheide, RH, 2015
)
0.66
"HIV-infected pregnant women receiving maraviroc as part of clinical care had intensive steady-state 12-hour pharmacokinetic profiles performed during the third trimester and ≥2 weeks after delivery."( Maraviroc Pharmacokinetics in HIV-1-Infected Pregnant Women.
Best, B; Burchett, S; Burger, D; Capparelli, E; Colbers, A; Haberl, A; Hawkins, D; Hidalgo Tenorio, C; Kabeya, K; Kreitchmann, R; Mirochnick, M; Schalkwijk, S; Smith, E; Stek, A; Taylor, G; van Kasteren, M; Wang, J, 2015
)
2.13
" Fresh and cryopreserved tissues were evaluated for human immunodeficiency virus (HIV) infection and pharmacokinetic (PK)/pharmacodynamic (PD) relationships."( Pharmacodynamic correlations using fresh and cryopreserved tissue following use of vaginal rings containing dapivirine and/or maraviroc in a randomized, placebo controlled trial.
Chen, BA; Dezzutti, CS; Hoesley, CJ; Johnson, S; Marzinke, MA; Nel, A; Nuttall, JP; Panther, L; Richardson-Harman, N; Rohan, LC, 2016
)
0.64
" The goal of this study was to establish feasibility and pharmacokinetic and pharmacodynamic profiles of maraviroc in pediatric HSCT recipients."( A Pharmacokinetic and Pharmacodynamic Study of Maraviroc as Acute Graft-versus-Host Disease Prophylaxis in Pediatric Allogeneic Stem Cell Transplant Recipients with Nonmalignant Diagnoses.
Davies, SM; Fukuda, T; Kashuba, ADM; Khandelwal, P; Marsh, RA; Mehta, PA; Mizuno, K; Teusink-Cross, A; Vinks, AA, 2016
)
0.91
" Maraviroc Cmax occurred within 4 hours in most compartments."( Brief Report: Pharmacokinetic/Pharmacodynamic Investigation of Single-Dose Oral Maraviroc in the Context of HIV-1 Pre-exposure Prophylaxis.
Back, D; Boffito, M; Dickinson, L; Egan, D; Else, L; Fox, J; Herrera, C; Jackson, A; Khoo, S; Olejniczak, N; Shattock, R; Tiraboschi, JM, 2016
)
1.57
" Dose adjustment and pharmacokinetic reevaluation occurred if the average concentrations (Cavg) at Week 2 were <100 ng/mL (Stage 1-dose finding)."( Pharmacokinetics, Safety and Efficacy of Maraviroc in Treatment-experienced Pediatric Patients Infected With CCR5-Tropic HIV-1.
Chokephaibulkit, K; Clark, A; Craig, C; Fang, A; Fourie, J; Giaquinto, C; Heera, J; Mawela, MP; McFadyen, L; Mitha, IH; Negra, MD; Valluri, SR; van der Ryst, E; Vourvahis, M; Zhang-Roper, RY, 2018
)
0.75
" Our results demonstrate that pharmacodynamic monitoring of CCR5 blockade unravels interpatient variability in the response to therapy and may serve as a clinically informative biomarker."( Pharmacodynamic Monitoring Predicts Outcomes of CCR5 Blockade as Graft-versus-Host Disease Prophylaxis.
Crisalli, L; Ganetsky, A; Huffman, AP; Porter, DL; Reshef, R; Richman, LP; Vonderheide, RH, 2018
)
0.48
" Our results indicate that neither ABCG2 nor ABCC2 influence maraviroc pharmacokinetic but that ABCB1/Abcb1 may be partly responsible for the decreased transplacental permeability of maraviroc to the fetus."( Interactions between Maraviroc and the ABCB1, ABCG2, and ABCC2 Transporters: An Important Role in Transplacental Pharmacokinetics.
Ambrus, C; Ceckova, M; Gaborik, Z; Ptackova, Z; Sorf, A; Staud, F; Tupova, L, 2019
)
1.07
" Formal pharmacokinetic (PK) sampling was performed on days five and fifteen."( A drug interaction study investigating the effect of Rifabutin on the pharmacokinetics of Maraviroc in healthy subjects.
Cameron, DW; Dennehy, M; Ghannad, M; Kanji, S; la Porte, C; Mallick, R; Sabri, E; Seguin, I; Tardiff, D; Zhang, G, 2019
)
0.74
" Cohort 1 participants received two single 8 mg/kg maraviroc doses 1 week apart with pharmacokinetic sampling after each dose."( Pharmacokinetics and safety of maraviroc in neonates.
Acosta, EP; Aziz, M; Best, BM; Bradford, S; Butler, K; Chadwick, EG; Hanley, S; Hayward, K; Homans, J; Mathiba, SR; Mcfadyen, L; Mirochnick, M; Moye, J; Murtaugh, W; Reding, CA; Rosebush, JC; Samson, P; Smith, E; Vourvahis, M, 2021
)
1.16
" All 13 evaluable Cohort 1 infants met the pharmacokinetic target."( Pharmacokinetics and safety of maraviroc in neonates.
Acosta, EP; Aziz, M; Best, BM; Bradford, S; Butler, K; Chadwick, EG; Hanley, S; Hayward, K; Homans, J; Mathiba, SR; Mcfadyen, L; Mirochnick, M; Moye, J; Murtaugh, W; Reding, CA; Rosebush, JC; Samson, P; Smith, E; Vourvahis, M, 2021
)
0.91
"The Combination HIV Antiretroviral Rectal Microbicide-3 (CHARM-03) study was a randomized, open-label, crossover Phase 1 safety and pharmacokinetic (PK) study of oral maraviroc (MVC) and MVC 1% gel."( A Randomized, Open-Label, Crossover Phase 1 Safety and Pharmacokinetic Study of Oral Maraviroc and Maraviroc 1% Gel (the CHARM-03 Study).
Abebe, KZ; Achilles, SL; Anton, PA; Brand, RM; Chawki, S; Chen, B; Cranston, RD; Engstrom, JC; Hendrix, CW; Jacobson, CE; Khanukova, E; Marzinke, MA; McGowan, IM; Reinhart, A; Richardson-Harman, N; Rohan, LC; Siegel, A; Spiegel, H; Stall, R; Steytler, J, 2022
)
1.14
" This study aimed to develop a population pharmacokinetic model to characterize the disposition of maraviroc in neonates to inform dosing regimens and expand available options."( Maraviroc Population Pharmacokinetics Within the First 6 Weeks of Life.
Best, BM; Bradford, S; Capparelli, EV; Chadwick, EG; Jean-Philippe, P; Liyanage, M; McFadyen, L; Mirochnick, M; Moye, J; Nikanjam, M; Rogg, L; Vourvahis, M; Whitson, K, 2022
)
2.38
"Using maraviroc concentrations from neonates who received either a single dose or multiple doses of 8 mg/kg of maraviroc in the first 6 weeks of life, a population pharmacokinetic model was developed to determine the effects of age, sex, maternal efavirenz exposure and concomitant ARV therapy on maraviroc disposition."( Maraviroc Population Pharmacokinetics Within the First 6 Weeks of Life.
Best, BM; Bradford, S; Capparelli, EV; Chadwick, EG; Jean-Philippe, P; Liyanage, M; McFadyen, L; Mirochnick, M; Moye, J; Nikanjam, M; Rogg, L; Vourvahis, M; Whitson, K, 2022
)
2.64

Compound-Compound Interactions

To characterize the cytochrome P450 enzyme(s) responsible for the N-dealkylation of maraviroc in vitro. To predict the extent of clinical drug-drug interactions (DDIs)

ExcerptReferenceRelevance
"To characterize the cytochrome P450 enzyme(s) responsible for the N-dealkylation of maraviroc in vitro, and predict the extent of clinical drug-drug interactions (DDIs)."( Maraviroc: in vitro assessment of drug-drug interaction potential.
Collins, C; Dickins, M; Hyland, R; Jones, B; Jones, H, 2008
)
2.01
" In this study we used nonhuman primates to examine the effect and mechanism of MVC alone or in combination with cyclosporine (CsA) to prolong cardiac allograft survivals."( CCR5 blockade in combination with cyclosporine increased cardiac graft survival and generated alternatively activated macrophages in primates.
Chen, G; Chen, W; Li, J; Stepkowski, SM; Wang, S; Xia, J; Ye, P; Zhang, K; Zhong, S, 2011
)
0.37
"To evaluate the hepatic safety of maraviroc in combination with other antiretroviral agents in HIV-1-infected subjects co-infected with HCV and/or HBV."( Hepatic safety in subjects with HIV-1 and hepatitis C and/or B virus: a randomized, double-blind study of maraviroc versus placebo in combination with antiretroviral agents.
Asmuth, DM; Bansal, M; Fätkenheuer, G; Heera, J; Jagannatha, S; Mukwaya, G; Pialoux, G; Pineda, JA; Plonski, F; Rockstroh, JK; Small, CB; Soriano, V,
)
0.62
"In this 148-week randomized, double-blind, placebo-controlled, multicentre study (NCT01327547), subjects received maraviroc twice daily (n = 70) or placebo (n = 67) in combination with other antiretroviral agents."( Hepatic safety in subjects with HIV-1 and hepatitis C and/or B virus: a randomized, double-blind study of maraviroc versus placebo in combination with antiretroviral agents.
Asmuth, DM; Bansal, M; Fätkenheuer, G; Heera, J; Jagannatha, S; Mukwaya, G; Pialoux, G; Pineda, JA; Plonski, F; Rockstroh, JK; Small, CB; Soriano, V,
)
0.56
"Our preview study found that CCR5 blockade combined with cyclosporine A could attenuate the severity of liver GVHD."( CCR5 blockade combined with cyclosporine A attenuates liver GVHD by impairing T cells function.
Dong, Y; Liu, H; Liu, W; Miao, S; Ren, H; Shi, Y; Tang, B; Yin, Y, 2016
)
0.43
"Data showed that MVC combined with CsA reduced donor T cells migration to target organs in vivo."( CCR5 blockade combined with cyclosporine A attenuates liver GVHD by impairing T cells function.
Dong, Y; Liu, H; Liu, W; Miao, S; Ren, H; Shi, Y; Tang, B; Yin, Y, 2016
)
0.43
" Latently infected primary rCD4+ or central memory T cells were stimulated with MVC alone or in combination with Bryostatin-1, a PKC agonist known to reverse HIV-1 latency."( The CCR5-antagonist Maraviroc reverses HIV-1 latency in vitro alone or in combination with the PKC-agonist Bryostatin-1.
Alcamí, J; Coiras, M; Gutiérrez, C; Jiménez-Tormo, L; López-Huertas, MR; Madrid-Elena, N; Moreno, S; Rodríguez-Mora, S, 2017
)
0.78
"" The present studies tested the hypothesis that in evoked pain, certain chemokine receptor antagonists (CRAs), given with a submaximal dose of morphine, would result in enhanced morphine potency."( Chemokine Receptor Antagonists in Combination with Morphine as a Novel Strategy for Opioid Dose Reduction in Pain Management.
Adler, MW; Chen, X; Cowan, A; Eisenstein, TK; Geller, EB; Inan, S; Meissler, JJ; Rawls, SM; Tallarida, CS, 2020
)
0.56
" In the cold-water tail flick and formalin tests, significant increases of the antinociceptive effects of morphine were also observed when combined with CRAs."( Chemokine Receptor Antagonists in Combination with Morphine as a Novel Strategy for Opioid Dose Reduction in Pain Management.
Adler, MW; Chen, X; Cowan, A; Eisenstein, TK; Geller, EB; Inan, S; Meissler, JJ; Rawls, SM; Tallarida, CS, 2020
)
0.56

Bioavailability

The absolute bioavailability of 100 mg oral maraviroc is 23%.

ExcerptReferenceRelevance
" The compound was incompletely absorbed in rat (approximately 20-30%) but well absorbed in dog (>70%)."( Species differences in the disposition of the CCR5 antagonist, UK-427,857, a new potential treatment for HIV.
Abel, S; Comby, P; Muirhead, GJ; Nedderman, AN; Smith, DA; Walker, DK, 2005
)
0.33
") maraviroc and determine the absolute bioavailability of oral maraviroc."( Assessment of the absorption, metabolism and absolute bioavailability of maraviroc in healthy male subjects.
Abel, S; Nedderman, AN; Ridgway, CE; Russell, D; Walker, DK; Whitlock, LA, 2008
)
1.3
" The absolute bioavailability of 100 mg oral maraviroc is 23%."( Assessment of the absorption, metabolism and absolute bioavailability of maraviroc in healthy male subjects.
Abel, S; Nedderman, AN; Ridgway, CE; Russell, D; Walker, DK; Whitlock, LA, 2008
)
0.84
"A two-compartment model parameterized to separate out absorption and clearance components on bioavailability was used."( A population pharmacokinetic meta-analysis of maraviroc in healthy volunteers and asymptomatic HIV-infected subjects.
Chan, PL; McFadyen, L; Weatherley, B, 2008
)
0.6
"For the typical non-Asian subject, fasted bioavailability increased asymptotically with dose from 24% at 100 mg to 33% at 600 mg."( A population pharmacokinetic meta-analysis of maraviroc in healthy volunteers and asymptomatic HIV-infected subjects.
Chan, PL; McFadyen, L; Weatherley, B, 2008
)
0.6
" MVC has an oral bioavailability of approximately 33%, which is limited by poor permeability as well as affinity for CYP3A and several drug transporters."( Improving maraviroc oral bioavailability by formation of solid drug nanoparticles.
Clark, A; Owen, A; Rannard, SP; Savage, AC; Scott, T; Siccardi, M; Tatham, LM; Vourvahis, M, 2019
)
0.92
"The ATP-binding cassette transporter P-glycoprotein (P-gp) is known to limit both brain penetration and oral bioavailability of many chemotherapy drugs."( A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
Ambudkar, SV; Brimacombe, KR; Chen, L; Gottesman, MM; Guha, R; Hall, MD; Klumpp-Thomas, C; Lee, OW; Lee, TD; Lusvarghi, S; Robey, RW; Shen, M; Tebase, BG, 2019
)
0.51

Dosage Studied

The potential of maraviroc (MVC), a small-molecule CCR5 antagonist, as a candidate to prevent HIV-1 sexual transmission by oral or topical dosing has not yet been completely established. Dosing followed recommendations for HIV therapy in the Summary of Product Characteristics for marviroc.

ExcerptRelevanceReference
" Studies in preclinical in vitro and in vivo models predicted maraviroc to have human pharmacokinetics consistent with once- or twice-daily dosing following oral administration."( Maraviroc (UK-427,857), a potent, orally bioavailable, and selective small-molecule inhibitor of chemokine receptor CCR5 with broad-spectrum anti-human immunodeficiency virus type 1 activity.
Armour, D; Dobbs, S; Dorr, P; Griffin, P; Irvine, B; Macartney, M; Mori, J; Napier, C; Perros, M; Price, D; Rickett, G; Smith-Burchnell, C; Stammen, B; Webster, R; Westby, M; Wood, A, 2005
)
2.01
"To use a viral dynamics model to compare the effectiveness of in vivo viral inhibition of several doses of maraviroc (MVC;UK-427,857) and to use a modeling approach to support design decisions for a monotherapy study using various dosing regimens of maraviroc given with and without food."( A pharmacokinetic-pharmacodynamic model to optimize the phase IIa development program of maraviroc.
Poland, B; Rosario, MC; Sullivan, J; van der Ryst, E; Westby, M, 2006
)
0.77
" The mean total recovery of dosed radioactivity was 96%, with the majority of radioactivity being recovered within 96 h postdose."( Assessment of the absorption, metabolism and absolute bioavailability of maraviroc in healthy male subjects.
Abel, S; Nedderman, AN; Ridgway, CE; Russell, D; Walker, DK; Whitlock, LA, 2008
)
0.58
" The current approved daily dosage of maraviroc is 300 mg bid in combination with other antiretroviral medications."( [Clinical pharmacokinetic of maraviroc].
Peytavin, G, 2008
)
0.91
" From a practical point of view, standardised tests are currently unavailable to assess susceptibility to MVC, although in dose-response phenotype tests a maximum percentage inhibition (MPI) < 95% would be indicative of resistance to the compound."( [Mechanisms of resistance and failure of treatment with maraviroc].
Delgado, R, 2008
)
0.59
"The mechanism of action, pharmacology, pharmacokinetics, clinical efficacy, drug interactions, adverse effects, dosage and administration, cost, and role in therapy of maraviroc are reviewed."( Maraviroc: a coreceptor CCR5 antagonist for management of HIV infection.
Pasquale, TR; Sahloff, EG; Yost, R, 2009
)
1.99
"In this exploratory study involving extensively treatment-experienced patients with advanced, non-R5 HIV-1 infection, neither superiority nor noninferiority was statistically demonstrated for either maraviroc dosage compared with placebo at 24 weeks of treatment."( A double-blind, placebo-controlled trial of maraviroc in treatment-experienced patients infected with non-R5 HIV-1.
Clotet, B; Clumeck, N; Fätkenheuer, G; Goodrich, J; Mayer, H; Saag, M; Sullivan, J; van der Ryst, E; Westby, M, 2009
)
0.8
" The dosing recommendations are based on concomitant medications due to drug interactions."( Maraviroc: a new CCR5 antagonist.
Khanlou, H; Sayana, S, 2009
)
1.8
" Secondly, a sigmoid E(max)vs dose model of dose-normalized non-compartmental AUC from oral dosing in 134 healthy young males and females across five phase 1 studies was constructed."( Maraviroc modelling strategy: use of early phase 1 data to support a semi-mechanistic population pharmacokinetic model.
McFadyen, L; Weatherley, B, 2009
)
1.8
" Providers should be aware of the need for tropism determination and dosing requirements for maraviroc."( Novel drug classes: entry inhibitors [enfuvirtide, chemokine (C-C motif) receptor 5 antagonists].
McKinnell, JA; Saag, MS, 2009
)
0.57
" We proposed a systematic classification scheme using FDA-approved drug labeling to assess the DILI potential of drugs, which yielded a benchmark dataset with 287 drugs representing a wide range of therapeutic categories and daily dosage amounts."( FDA-approved drug labeling for the study of drug-induced liver injury.
Chen, M; Fang, H; Liu, Z; Shi, Q; Tong, W; Vijay, V, 2011
)
0.37
" Minor, clinically nonsignificant effects on the pharmacokinetics of raltegravir coadministered with lersivirine were observed at steady state for raltegravir, with estimated mean changes of -15%, -29%, and +25% in the area under the concentration-time profile from time zero to the end of the dosing interval (AUC(tau)), maximum plasma concentration (C(max)), and concentration observed 12 h postdose (C(12)), respectively."( Pharmacokinetic effects of coadministration of lersivirine with raltegravir or maraviroc in healthy subjects.
Banerjee, S; Davis, J; Labadie, RR; Langdon, G; Layton, G; Ndongo, MN; Vourvahis, M, 2012
)
0.61
"To describe the pharmacokinetics of maraviroc when dosed at 150 or 300 mg once daily with 800/100 mg of darunavir/ritonavir."( Once daily maraviroc 300 mg or 150 mg in combination with ritonavir-boosted darunavir 800/100 mg.
Ainsworth, J; Cook, R; Dufty, N; Gandhi, K; Hickinbottom, G; Khonyongwa, K; Okoli, C; Owen, A; Siccardi, M; Taylor, S; Thomas-William, S; Watson, J, 2012
)
1.04
" The 50% inhibitory concentration (IC(50)), 90% inhibitory concentration (IC(90)) and dose-response curve slopes were determined for each drug."( Baseline susceptibility of primary HIV-2 to entry inhibitors.
Antunes, F; Barroso, H; Bártolo, I; Borrego, P; Caixas, U; Calado, R; Cavaco-Silva, P; Doroana, M; Maltez, F; Marcelino, JM; Rocha, C; Taveira, N, 2012
)
0.38
"Current product labels for maraviroc and raltegravir provide no dosing guidance for patients with end-stage liver disease and worsening renal function."( Impaired maraviroc and raltegravir clearance in a human immunodeficiency virus-infected patient with end-stage liver disease and renal impairment: a management dilemma.
Boyd, SD; McLaughlin, M; Morse, CG; Pau, AK; Penzak, SR, 2012
)
1.09
" This is apparently due to MVC's twice-daily dosing schedule."( CCR5 inhibitors: emergence, success, and challenges.
Das, D; Maeda, K; Mitsuya, H; Nakata, H, 2012
)
0.38
" 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
" MVC was added to cART for 24 weeks, at the recommended dosage per drug-drug interactions."( Maraviroc intensification of stable antiviral therapy in HIV-1-infected patients with poor immune restoration: MARIMUNO-ANRS 145 study.
Allavena, C; Barbuat, C; Corbeau, P; Cuzin, L; Delobel, P; Flandre, P; Ghosn, J; Lascoux-Combe, C; Peytavin, G; Psomas, C; Reynes, J; Trabelsi, S, 2012
)
1.82
" MVC dosing was informed by the pharmacokinetic profile seen in blood and rectal tissues and consisted of a human-equivalent dose given 24 h before virus exposure, followed by a booster postexposure dose."( Lack of prophylactic efficacy of oral maraviroc in macaques despite high drug concentrations in rectal tissues.
Aubert, R; Aung, W; Bachman, S; García-Lerma, JG; Heneine, W; Kersh, E; Martin, A; Massud, I; Mitchell, J; Pau, CP; Solomon Tsegaye, T, 2013
)
0.66
"With SQV/r, geometric mean steady-state maraviroc area under the plasma concentration-time curve for the dosing interval (AUCtau) was 5,341 (coefficient of variation [CV], 27%), 8,119 (35%), and 6,193 (27%) h•ng/mL, in normal function, mild, and moderate impairment groups, respectively."( Pharmacokinetics, safety, and tolerability of maraviroc in HIV-negative subjects with impaired renal function.
Checchio, T; Fang, J; Heera, J; McFadyen, L; Milton, A; Vourvahis, M; Weatherley, B,
)
0.66
"The data suggest that no dosing interval adjustments are required in subjects with renal impairment when maraviroc is administered alone."( Pharmacokinetics, safety, and tolerability of maraviroc in HIV-negative subjects with impaired renal function.
Checchio, T; Fang, J; Heera, J; McFadyen, L; Milton, A; Vourvahis, M; Weatherley, B,
)
0.6
" Pharmacokinetic parameters, assessed on day 5 of period 1 and on days 10 and 20 of period 2, included the maximum plasma concentration (Cmax), the concentration at end of dosing interval (Cτ), and the area under the curve over dosing interval (AUCτ)."( Pharmacokinetic interaction between maraviroc and fosamprenavir-ritonavir: an open-label, fixed-sequence study in healthy subjects.
Fang, A; Heera, J; Mendes da Costa, L; Plotka, A; Vourvahis, M, 2013
)
0.66
"MVC exposures were significantly increased with BOC or TVR, therefore MVC should be dosed at 150 mg twice daily when coadministered with these newly approved hepatitis C protease inhibitors."( The effects of boceprevir and telaprevir on the pharmacokinetics of maraviroc: an open-label, fixed-sequence study in healthy volunteers.
Fang, A; Heera, J; Kantaridis, C; Plotka, A; Vourvahis, M, 2014
)
0.64
"Despite improvements in access to antiretroviral therapy and the use of simplified dosing regimens, HIV infection is still an important global public health problem."( An overview of antiretroviral pre-exposure prophylaxis of HIV infection.
McGowan, I, 2014
)
0.4
" While solid dosage forms like films and tablets increase retention, they often require more than 15 min to fully dissolve, potentially increasing the risk of inducing epithelial abrasions during sex."( Electrospun solid dispersions of Maraviroc for rapid intravaginal preexposure prophylaxis of HIV.
Ball, C; Woodrow, KA, 2014
)
0.68
" This modelling technique is able to inform the design of clinical studies, and allows assessment of pragmatic dosing strategies under complex therapeutic scenarios."( Use of in vitro to in vivo extrapolation to predict the optimal strategy for patients switching from efavirenz to maraviroc or nevirapine.
Back, D; Davies, G; Khoo, S; Owen, A; Schipani, A; Siccardi, M, 2015
)
0.63
" These results provide a framework for future early investigations of antiretroviral efficacy in HIV prevention to optimize dosing strategies in clinical investigations."( Models for predicting effective HIV chemoprevention in women.
Cohen, MS; Emerson, CW; Fedoriw, Y; Geller, EJ; Kashuba, AD; Nelson, JA; Nicol, MR; Patterson, KB; Prince, HM; Sykes, C, 2015
)
0.42
"The potential of maraviroc (MVC), a small-molecule CCR5 antagonist, as a candidate to prevent HIV-1 sexual transmission by oral or topical dosing has not yet been completely established."( Short Communication: Limited Anti-HIV-1 Activity of Maraviroc in Mucosal Tissues.
Armanasco, N; Fletcher, P; Herrera, C; Nuttall, J; Shattock, RJ, 2016
)
1.02
" Using an in vitro model of Ebola Zaire replication with transcription-competent virus like particles (trVLPs), requiring only level 2 biosafety containment, we compared the activities of the type I interferons (IFNs) IFN-α and IFN-ß, a panel of viral polymerase inhibitors (lamivudine (3TC), zidovudine (AZT) tenofovir (TFV), favipiravir (FPV), the active metabolite of brincidofovir, cidofovir (CDF)), and the estrogen receptor modulator, toremifene (TOR), in inhibiting viral replication in dose-response and time course studies."( A Rapid Screening Assay Identifies Monotherapy with Interferon-ß and Combination Therapies with Nucleoside Analogs as Effective Inhibitors of Ebola Virus.
Baker, DP; Branch, DR; Fish, EN; Hoenen, T; Kobinger, GP; Kozlowski, HN; Majchrzak-Kita, B; McCarthy, SD; Racine, T, 2016
)
0.43
" A significant research thrust in the field has been to characterize different dosage forms for formulation of physicochemically diverse antiretroviral drugs."( Biophysical characterization of small molecule antiviral-loaded nanolipogels for HIV-1 chemoprophylaxis and topical mucosal application.
Golan-Paz, S; Jiang, Y; Ramanathan, R; Read, B; Woodrow, KA, 2016
)
0.43
" New drugs that offer new mechanisms of action, improvements in potency and activity even against multidrug-resistant viruses, dosing convenience, and tolerability have been approved."( [Companion Diagnostics for Selecting Antiretroviral Drugs against HIV-1].
Fukutake, K, 2015
)
0.42
" Data were log-transformed and analyzed by linear least squared regression, nonlinear Emax dose-response model and Satterthwaite t test."( Pharmacodynamic correlations using fresh and cryopreserved tissue following use of vaginal rings containing dapivirine and/or maraviroc in a randomized, placebo controlled trial.
Chen, BA; Dezzutti, CS; Hoesley, CJ; Johnson, S; Marzinke, MA; Nel, A; Nuttall, JP; Panther, L; Richardson-Harman, N; Rohan, LC, 2016
)
0.64
" 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
)
0.96
" One-hundred and three participants were enrolled into 4 age/formulation cohorts and dosed twice daily."( Pharmacokinetics, Safety and Efficacy of Maraviroc in Treatment-experienced Pediatric Patients Infected With CCR5-Tropic HIV-1.
Chokephaibulkit, K; Clark, A; Craig, C; Fang, A; Fourie, J; Giaquinto, C; Heera, J; Mawela, MP; McFadyen, L; Mitha, IH; Negra, MD; Valluri, SR; van der Ryst, E; Vourvahis, M; Zhang-Roper, RY, 2018
)
0.75
"The maraviroc dosing strategy resulted in participants achieving the target Cavg, with exposure ranges similar to those observed in adults on approved doses."( Pharmacokinetics, Safety and Efficacy of Maraviroc in Treatment-experienced Pediatric Patients Infected With CCR5-Tropic HIV-1.
Chokephaibulkit, K; Clark, A; Craig, C; Fang, A; Fourie, J; Giaquinto, C; Heera, J; Mawela, MP; McFadyen, L; Mitha, IH; Negra, MD; Valluri, SR; van der Ryst, E; Vourvahis, M; Zhang-Roper, RY, 2018
)
1.3
" All patients completed full maraviroc dosing and follow-up."( Maraviroc Is Associated with Latent HIV-1 Reactivation through NF-κB Activation in Resting CD4
Coronel Díaz, M; Díaz-de Santiago, A; Domínguez, E; Dronda, F; García-Bermejo, ML; Gutiérrez, C; Hernández-Novoa, B; López-Huertas, MR; Madrid-Elena, N; Moreno, S; Sastre, B; Serrano-Villar, S, 2018
)
2.21
" Maraviroc (MVC) is an orally dosed CCR5 antagonist approved for use in patients infected with CCR5-trophic HIV-1."( Towards a Maraviroc long-acting injectable nanoformulation.
Clark, A; Dwyer, A; Owen, A; Rannard, SP; Savage, AC; Scott, T; Siccardi, M; Tatham, LM; Vourvahis, M, 2019
)
1.83
"Intravaginal rings (IVRs) for HIV pre-exposure prophylaxis (PrEP) theoretically overcome some adherence concerns associated with frequent dosing that can occur with oral or vaginal film/gel regimens."( Safety and pharmacokinetics of single, dual, and triple antiretroviral drug formulations delivered by pod-intravaginal rings designed for HIV-1 prevention: A Phase I trial.
Anton, PA; Baum, MM; Butkyavichene, I; Churchman, SA; Cortez, JM; Dawson, L; Fanter, R; Gunawardana, M; Guthrie, KM; Hendrix, CW; Marzinke, MA; Miller, CS; Moss, JA; Olive, TJ; Pyles, RB; Rosen, RK; Vargas, SE; Vincent, KL; Yang, F, 2018
)
0.48
" We found that standard dosing strategies in preclinical species closely mimicked tissue concentrations in humans for some, but not all, ARVs."( Antiretroviral Penetration across Three Preclinical Animal Models and Humans in Eight Putative HIV Viral Reservoirs.
Adamson, L; Akkina, R; Blake, K; Burgunder, EM; Devanathan, AS; Garcia, JV; Kashuba, ADM; Kovarova, M; Luciw, P; Pirone, JR; Remling-Mulder, L; Rosen, EP; Schauer, AP; Srinivas, N; Sykes, C; White, NR, 2019
)
0.51
" Non-standard maraviroc use also comprised reported once daily dosing (20."( Clinical use, efficacy, and durability of maraviroc for antiretroviral therapy in routine care: A European survey.
Boucher, C; De Luca, A; Döring, M; Geretti, AM; Incardona, F; Kaiser, R; Lengauer, T; Pezzotti, P; Pfeifer, N; Schülter, E; Vandamme, AM; Zazzi, M, 2019
)
1.14
" However, dosing is usually two times per day, unlike most currently recommended antiretroviral therapies."( Protocol for a phase IV, open-label feasibility study investigating non-invasive markers of hepatic fibrosis in people living with HIV-1 and non-alcoholic fatty liver disease randomised to receiving optimised background therapy (OBT) plus maraviroc or OBT
Abramowicz, I; Bradshaw, D; Bremner, S; Gilleece, Y; Perry, N; Verma, S, 2020
)
0.74
" At a single site, healthy HIV-uninfected men and women were enrolled and randomized to an open label crossover sequence of eight consecutive daily exposures to MVC 300 mg dosed orally, MCV 1% gel dosed rectally, and MVC 1% gel dosed vaginally."( A Randomized, Open-Label, Crossover Phase 1 Safety and Pharmacokinetic Study of Oral Maraviroc and Maraviroc 1% Gel (the CHARM-03 Study).
Abebe, KZ; Achilles, SL; Anton, PA; Brand, RM; Chawki, S; Chen, B; Cranston, RD; Engstrom, JC; Hendrix, CW; Jacobson, CE; Khanukova, E; Marzinke, MA; McGowan, IM; Reinhart, A; Richardson-Harman, N; Rohan, LC; Siegel, A; Spiegel, H; Stall, R; Steytler, J, 2022
)
0.95
" Application of maraviroc with morphine can reduce effective dosage of morphine 2,3 fold."( [Potential application of maraviroc in the therapy of neuropathic pain].
Olakowska, E; Sojka, P; Właszczuk, A, 2021
)
1.27
" This study aimed to develop a population pharmacokinetic model to characterize the disposition of maraviroc in neonates to inform dosing regimens and expand available options."( Maraviroc Population Pharmacokinetics Within the First 6 Weeks of Life.
Best, BM; Bradford, S; Capparelli, EV; Chadwick, EG; Jean-Philippe, P; Liyanage, M; McFadyen, L; Mirochnick, M; Moye, J; Nikanjam, M; Rogg, L; Vourvahis, M; Whitson, K, 2022
)
2.38
" The final model was used in Monte Carlo simulations to generate expected exposures with recommended dosing regimens."( Maraviroc Population Pharmacokinetics Within the First 6 Weeks of Life.
Best, BM; Bradford, S; Capparelli, EV; Chadwick, EG; Jean-Philippe, P; Liyanage, M; McFadyen, L; Mirochnick, M; Moye, J; Nikanjam, M; Rogg, L; Vourvahis, M; Whitson, K, 2022
)
2.16
" Monte Carlo simulations with FDA-approved weight band dosing resulted in the majority of simulated patients (84."( Maraviroc Population Pharmacokinetics Within the First 6 Weeks of Life.
Best, BM; Bradford, S; Capparelli, EV; Chadwick, EG; Jean-Philippe, P; Liyanage, M; McFadyen, L; Mirochnick, M; Moye, J; Nikanjam, M; Rogg, L; Vourvahis, M; Whitson, K, 2022
)
2.16
"While maraviroc apparent clearance is decreased in the first few days of life, the current FDA-approved maraviroc weight band dosing provides maraviroc exposures for neonates in the first 6 weeks of life, which were consistent with adult maraviroc exposure range."( Maraviroc Population Pharmacokinetics Within the First 6 Weeks of Life.
Best, BM; Bradford, S; Capparelli, EV; Chadwick, EG; Jean-Philippe, P; Liyanage, M; McFadyen, L; Mirochnick, M; Moye, J; Nikanjam, M; Rogg, L; Vourvahis, M; Whitson, K, 2022
)
2.64
" An MSI threshold for classifying daily adherence was established using clinical samples from healthy volunteers following directly observed dosing of 1 to 7 doses MVC/week."( Mass spectrometry imaging of hair identifies daily maraviroc adherence in HPTN 069/ACTG A5305.
Amico, KR; Gandhi, M; Gerona, RR; Gilliland, WM; Gulick, RM; Kashuba, ADM; Mayer, KH; Rosen, EP; White, N, 2023
)
1.16
" Dosing followed recommendations for HIV therapy in the Summary of Product Characteristics for maraviroc."( Hepmarc: A 96 week randomised controlled feasibility trial of add-on maraviroc in people with HIV and non-alcoholic fatty liver disease.
Abramowicz, I; Bradshaw, D; Bremner, S; Chadwick, D; Clarke, E; Curnock, M; Fox, A; Gilleece, Y; Gompels, M; Housman, R; Jennings, L; Kirk, S; Lambert, P; Miras, H; Nelson, M; Orkin, C; Perry, N; Robinson, R; Verma, S, 2023
)
1.36
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Drug Classes (1)

ClassDescription
tropane alkaloid
[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 (22)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
EWS/FLI fusion proteinHomo sapiens (human)Potency32.47970.001310.157742.8575AID1259252; AID1259253; AID1259255; AID1259256
estrogen nuclear receptor alphaHomo sapiens (human)Potency22.59780.000229.305416,493.5996AID743069; AID743075
[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)
Solute carrier family 22 member 2Homo sapiens (human)IC50 (µMol)255.00000.40003.10009.7000AID721751
ATP-binding cassette sub-family C member 3Homo sapiens (human)IC50 (µMol)133.00000.63154.45319.3000AID1473740
Multidrug resistance-associated protein 4Homo sapiens (human)IC50 (µMol)133.00000.20005.677410.0000AID1473741
Bile salt export pumpHomo sapiens (human)IC50 (µMol)110.00000.11007.190310.0000AID1473738
Cytochrome P450 1A2Homo sapiens (human)IC50 (µMol)25.00000.00011.774010.0000AID1897799
Cytochrome P450 3A4Homo sapiens (human)IC50 (µMol)9.00000.00011.753610.0000AID1897803; AID1897804
Cytochrome P450 2D6Homo sapiens (human)IC50 (µMol)25.00000.00002.015110.0000AID1897802
Cytochrome P450 2C9 Homo sapiens (human)IC50 (µMol)14.40000.00002.800510.0000AID1897800
Potassium voltage-gated channel subfamily E member 1Homo sapiens (human)IC50 (µMol)63.09570.12004.048010.0000AID1207375
Cytochrome P450 2C19Homo sapiens (human)IC50 (µMol)25.00000.00002.398310.0000AID1897801
Delta-type opioid receptorRattus norvegicus (Norway rat)IC50 (µMol)0.00140.00030.38877.0000AID465010
C-C chemokine receptor type 5Homo sapiens (human)IC50 (µMol)0.00440.00020.25679.0000AID1062970; AID1065004; AID1065005; AID1065006; AID1065007; AID1121816; AID1191659; AID1370975; AID1400606; AID1713967; AID1808908; AID1820204; AID1897751; AID393050; AID395483; AID465010; AID479402; AID554132; AID707904
C-C chemokine receptor type 5Mus musculus (house mouse)IC50 (µMol)0.00520.00030.00170.0052AID1353545
Potassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)IC50 (µMol)63.09570.12004.048010.0000AID1207375
C-C chemokine receptor type 5Macaca mulatta (Rhesus monkey)Ki0.00020.00020.00020.0002AID1121818; AID1713968
Potassium voltage-gated channel subfamily H member 2Homo sapiens (human)IC50 (µMol)81.71220.00091.901410.0000AID1207469; AID1207499; AID1207529
Sodium channel protein type 5 subunit alphaHomo sapiens (human)IC50 (µMol)815.47850.00033.64849.2000AID1207315; AID1207345
Multidrug and toxin extrusion protein 2Homo sapiens (human)IC50 (µMol)297.00000.16003.95718.6000AID721752
Canalicular multispecific organic anion transporter 1Homo sapiens (human)IC50 (µMol)133.00002.41006.343310.0000AID1473739
Multidrug and toxin extrusion protein 1Homo sapiens (human)IC50 (µMol)17.30000.01002.765610.0000AID721754
[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)
C-C chemokine receptor type 5Homo sapiens (human)EC50 (µMol)0.01800.00702.09365.0119AID1820206
C-C chemokine receptor type 5Homo sapiens (human)Kd0.02500.00300.01400.0250AID652438
[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)
C-C chemokine receptor type 5Homo sapiens (human)IC90 (µMol)0.01000.01000.01000.0100AID249028
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (228)

Processvia Protein(s)Taxonomy
activation of cysteine-type endopeptidase activity involved in apoptotic processSolute carrier family 22 member 2Homo sapiens (human)
positive regulation of gene expressionSolute carrier family 22 member 2Homo sapiens (human)
organic cation transportSolute carrier family 22 member 2Homo sapiens (human)
monoatomic cation transportSolute carrier family 22 member 2Homo sapiens (human)
neurotransmitter transportSolute carrier family 22 member 2Homo sapiens (human)
serotonin transportSolute carrier family 22 member 2Homo sapiens (human)
body fluid secretionSolute carrier family 22 member 2Homo sapiens (human)
organic cation transportSolute carrier family 22 member 2Homo sapiens (human)
quaternary ammonium group transportSolute carrier family 22 member 2Homo sapiens (human)
prostaglandin transportSolute carrier family 22 member 2Homo sapiens (human)
amine transportSolute carrier family 22 member 2Homo sapiens (human)
putrescine transportSolute carrier family 22 member 2Homo sapiens (human)
spermidine transportSolute carrier family 22 member 2Homo sapiens (human)
acetylcholine transportSolute carrier family 22 member 2Homo sapiens (human)
choline transportSolute carrier family 22 member 2Homo sapiens (human)
dopamine transportSolute carrier family 22 member 2Homo sapiens (human)
norepinephrine transportSolute carrier family 22 member 2Homo sapiens (human)
xenobiotic transportSolute carrier family 22 member 2Homo sapiens (human)
epinephrine transportSolute carrier family 22 member 2Homo sapiens (human)
histamine transportSolute carrier family 22 member 2Homo sapiens (human)
serotonin uptakeSolute carrier family 22 member 2Homo sapiens (human)
histamine uptakeSolute carrier family 22 member 2Homo sapiens (human)
norepinephrine uptakeSolute carrier family 22 member 2Homo sapiens (human)
thiamine transmembrane transportSolute carrier family 22 member 2Homo sapiens (human)
purine-containing compound transmembrane transportSolute carrier family 22 member 2Homo sapiens (human)
amino acid import across plasma membraneSolute carrier family 22 member 2Homo sapiens (human)
dopamine uptakeSolute carrier family 22 member 2Homo sapiens (human)
L-arginine import across plasma membraneSolute carrier family 22 member 2Homo sapiens (human)
export across plasma membraneSolute carrier family 22 member 2Homo sapiens (human)
transport across blood-brain barrierSolute carrier family 22 member 2Homo sapiens (human)
L-alpha-amino acid transmembrane transportSolute carrier family 22 member 2Homo sapiens (human)
spermidine transmembrane transportSolute carrier family 22 member 2Homo sapiens (human)
L-arginine transmembrane transportSolute carrier family 22 member 2Homo sapiens (human)
cellular detoxificationSolute carrier family 22 member 2Homo sapiens (human)
xenobiotic transport across blood-brain barrierSolute carrier family 22 member 2Homo sapiens (human)
xenobiotic metabolic processATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
bile acid and bile salt transportATP-binding cassette sub-family C member 3Homo sapiens (human)
glucuronoside transportATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transportATP-binding cassette sub-family C member 3Homo sapiens (human)
transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
leukotriene transportATP-binding cassette sub-family C member 3Homo sapiens (human)
monoatomic anion transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
transport across blood-brain barrierATP-binding cassette sub-family C member 3Homo sapiens (human)
prostaglandin secretionMultidrug resistance-associated protein 4Homo sapiens (human)
cilium assemblyMultidrug resistance-associated protein 4Homo sapiens (human)
platelet degranulationMultidrug resistance-associated protein 4Homo sapiens (human)
xenobiotic metabolic processMultidrug resistance-associated protein 4Homo sapiens (human)
xenobiotic transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
bile acid and bile salt transportMultidrug resistance-associated protein 4Homo sapiens (human)
prostaglandin transportMultidrug resistance-associated protein 4Homo sapiens (human)
urate transportMultidrug resistance-associated protein 4Homo sapiens (human)
glutathione transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
cAMP transportMultidrug resistance-associated protein 4Homo sapiens (human)
leukotriene transportMultidrug resistance-associated protein 4Homo sapiens (human)
monoatomic anion transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
export across plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
transport across blood-brain barrierMultidrug resistance-associated protein 4Homo sapiens (human)
guanine nucleotide transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
fatty acid metabolic processBile salt export pumpHomo sapiens (human)
bile acid biosynthetic processBile salt export pumpHomo sapiens (human)
xenobiotic metabolic processBile salt export pumpHomo sapiens (human)
xenobiotic transmembrane transportBile salt export pumpHomo sapiens (human)
response to oxidative stressBile salt export pumpHomo sapiens (human)
bile acid metabolic processBile salt export pumpHomo sapiens (human)
response to organic cyclic compoundBile salt export pumpHomo sapiens (human)
bile acid and bile salt transportBile salt export pumpHomo sapiens (human)
canalicular bile acid transportBile salt export pumpHomo sapiens (human)
protein ubiquitinationBile salt export pumpHomo sapiens (human)
regulation of fatty acid beta-oxidationBile salt export pumpHomo sapiens (human)
carbohydrate transmembrane transportBile salt export pumpHomo sapiens (human)
bile acid signaling pathwayBile salt export pumpHomo sapiens (human)
cholesterol homeostasisBile salt export pumpHomo sapiens (human)
response to estrogenBile salt export pumpHomo sapiens (human)
response to ethanolBile salt export pumpHomo sapiens (human)
xenobiotic export from cellBile salt export pumpHomo sapiens (human)
lipid homeostasisBile salt export pumpHomo sapiens (human)
phospholipid homeostasisBile salt export pumpHomo sapiens (human)
positive regulation of bile acid secretionBile salt export pumpHomo sapiens (human)
regulation of bile acid metabolic processBile salt export pumpHomo sapiens (human)
transmembrane transportBile salt export pumpHomo sapiens (human)
steroid catabolic processCytochrome P450 1A2Homo sapiens (human)
porphyrin-containing compound metabolic processCytochrome P450 1A2Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 1A2Homo sapiens (human)
cholesterol metabolic processCytochrome P450 1A2Homo sapiens (human)
estrogen metabolic processCytochrome P450 1A2Homo sapiens (human)
toxin biosynthetic processCytochrome P450 1A2Homo sapiens (human)
post-embryonic developmentCytochrome P450 1A2Homo sapiens (human)
alkaloid metabolic processCytochrome P450 1A2Homo sapiens (human)
regulation of gene expressionCytochrome P450 1A2Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 1A2Homo sapiens (human)
dibenzo-p-dioxin metabolic processCytochrome P450 1A2Homo sapiens (human)
epoxygenase P450 pathwayCytochrome P450 1A2Homo sapiens (human)
lung developmentCytochrome P450 1A2Homo sapiens (human)
methylationCytochrome P450 1A2Homo sapiens (human)
monocarboxylic acid metabolic processCytochrome P450 1A2Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 1A2Homo sapiens (human)
retinol metabolic processCytochrome P450 1A2Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 1A2Homo sapiens (human)
cellular respirationCytochrome P450 1A2Homo sapiens (human)
aflatoxin metabolic processCytochrome P450 1A2Homo sapiens (human)
hydrogen peroxide biosynthetic processCytochrome P450 1A2Homo sapiens (human)
oxidative demethylationCytochrome P450 1A2Homo sapiens (human)
cellular response to cadmium ionCytochrome P450 1A2Homo sapiens (human)
omega-hydroxylase P450 pathwayCytochrome P450 1A2Homo sapiens (human)
lipid hydroxylationCytochrome P450 3A4Homo sapiens (human)
lipid metabolic processCytochrome P450 3A4Homo sapiens (human)
steroid catabolic processCytochrome P450 3A4Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 3A4Homo sapiens (human)
steroid metabolic processCytochrome P450 3A4Homo sapiens (human)
cholesterol metabolic processCytochrome P450 3A4Homo sapiens (human)
androgen metabolic processCytochrome P450 3A4Homo sapiens (human)
estrogen metabolic processCytochrome P450 3A4Homo sapiens (human)
alkaloid catabolic processCytochrome P450 3A4Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 3A4Homo sapiens (human)
calcitriol biosynthetic process from calciolCytochrome P450 3A4Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 3A4Homo sapiens (human)
vitamin D metabolic processCytochrome P450 3A4Homo sapiens (human)
vitamin D catabolic processCytochrome P450 3A4Homo sapiens (human)
retinol metabolic processCytochrome P450 3A4Homo sapiens (human)
retinoic acid metabolic processCytochrome P450 3A4Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 3A4Homo sapiens (human)
aflatoxin metabolic processCytochrome P450 3A4Homo sapiens (human)
oxidative demethylationCytochrome P450 3A4Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 2D6Homo sapiens (human)
steroid metabolic processCytochrome P450 2D6Homo sapiens (human)
cholesterol metabolic processCytochrome P450 2D6Homo sapiens (human)
estrogen metabolic processCytochrome P450 2D6Homo sapiens (human)
coumarin metabolic processCytochrome P450 2D6Homo sapiens (human)
alkaloid metabolic processCytochrome P450 2D6Homo sapiens (human)
alkaloid catabolic processCytochrome P450 2D6Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 2D6Homo sapiens (human)
isoquinoline alkaloid metabolic processCytochrome P450 2D6Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 2D6Homo sapiens (human)
retinol metabolic processCytochrome P450 2D6Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 2D6Homo sapiens (human)
negative regulation of bindingCytochrome P450 2D6Homo sapiens (human)
oxidative demethylationCytochrome P450 2D6Homo sapiens (human)
negative regulation of cellular organofluorine metabolic processCytochrome P450 2D6Homo sapiens (human)
arachidonic acid metabolic processCytochrome P450 2D6Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 2C9 Homo sapiens (human)
steroid metabolic processCytochrome P450 2C9 Homo sapiens (human)
cholesterol metabolic processCytochrome P450 2C9 Homo sapiens (human)
estrogen metabolic processCytochrome P450 2C9 Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 2C9 Homo sapiens (human)
epoxygenase P450 pathwayCytochrome P450 2C9 Homo sapiens (human)
urea metabolic processCytochrome P450 2C9 Homo sapiens (human)
monocarboxylic acid metabolic processCytochrome P450 2C9 Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 2C9 Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 2C9 Homo sapiens (human)
amide metabolic processCytochrome P450 2C9 Homo sapiens (human)
icosanoid biosynthetic processCytochrome P450 2C9 Homo sapiens (human)
oxidative demethylationCytochrome P450 2C9 Homo sapiens (human)
omega-hydroxylase P450 pathwayCytochrome P450 2C9 Homo sapiens (human)
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)
long-chain fatty acid metabolic processCytochrome P450 2C19Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 2C19Homo sapiens (human)
steroid metabolic processCytochrome P450 2C19Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 2C19Homo sapiens (human)
epoxygenase P450 pathwayCytochrome P450 2C19Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 2C19Homo sapiens (human)
omega-hydroxylase P450 pathwayCytochrome P450 2C19Homo sapiens (human)
MAPK cascadeC-C chemokine receptor type 5Homo sapiens (human)
dendritic cell chemotaxisC-C chemokine receptor type 5Homo sapiens (human)
calcium ion transportC-C chemokine receptor type 5Homo sapiens (human)
chemotaxisC-C chemokine receptor type 5Homo sapiens (human)
cellular defense responseC-C chemokine receptor type 5Homo sapiens (human)
cell surface receptor signaling pathwayC-C chemokine receptor type 5Homo sapiens (human)
G protein-coupled receptor signaling pathwayC-C chemokine receptor type 5Homo sapiens (human)
cell-cell signalingC-C chemokine receptor type 5Homo sapiens (human)
release of sequestered calcium ion into cytosol by sarcoplasmic reticulumC-C chemokine receptor type 5Homo sapiens (human)
calcium-mediated signalingC-C chemokine receptor type 5Homo sapiens (human)
signalingC-C chemokine receptor type 5Homo sapiens (human)
symbiont entry into host cellC-C chemokine receptor type 5Homo sapiens (human)
chemokine-mediated signaling pathwayC-C chemokine receptor type 5Homo sapiens (human)
response to cholesterolC-C chemokine receptor type 5Homo sapiens (human)
cellular response to lipopolysaccharideC-C chemokine receptor type 5Homo sapiens (human)
negative regulation of macrophage apoptotic processC-C chemokine receptor type 5Homo sapiens (human)
inflammatory responseC-C chemokine receptor type 5Homo sapiens (human)
positive regulation of cytosolic calcium ion concentrationC-C chemokine receptor type 5Homo sapiens (human)
immune responseC-C chemokine receptor type 5Homo sapiens (human)
cell chemotaxisC-C chemokine receptor type 5Homo 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)
organic cation transportMultidrug and toxin extrusion protein 2Homo sapiens (human)
transmembrane transportMultidrug and toxin extrusion protein 2Homo sapiens (human)
proton transmembrane transportMultidrug and toxin extrusion protein 2Homo sapiens (human)
xenobiotic detoxification by transmembrane export across the plasma membraneMultidrug and toxin extrusion protein 2Homo sapiens (human)
xenobiotic metabolic processCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
negative regulation of gene expressionCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bile acid and bile salt transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bilirubin transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
heme catabolic processCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic export from cellCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transepithelial transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
leukotriene transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
monoatomic anion transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transport across blood-brain barrierCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transport across blood-brain barrierCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transmembrane transportMultidrug and toxin extrusion protein 1Homo sapiens (human)
organic cation transportMultidrug and toxin extrusion protein 1Homo sapiens (human)
putrescine transportMultidrug and toxin extrusion protein 1Homo sapiens (human)
xenobiotic transportMultidrug and toxin extrusion protein 1Homo sapiens (human)
transmembrane transportMultidrug and toxin extrusion protein 1Homo sapiens (human)
thiamine transmembrane transportMultidrug and toxin extrusion protein 1Homo sapiens (human)
amino acid import across plasma membraneMultidrug and toxin extrusion protein 1Homo sapiens (human)
L-arginine import across plasma membraneMultidrug and toxin extrusion protein 1Homo sapiens (human)
L-alpha-amino acid transmembrane transportMultidrug and toxin extrusion protein 1Homo sapiens (human)
proton transmembrane transportMultidrug and toxin extrusion protein 1Homo sapiens (human)
L-arginine transmembrane transportMultidrug and toxin extrusion protein 1Homo sapiens (human)
xenobiotic detoxification by transmembrane export across the plasma membraneMultidrug and toxin extrusion protein 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (115)

Processvia Protein(s)Taxonomy
amine transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
acetylcholine transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
neurotransmitter transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
monoamine transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
organic anion transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
organic cation transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
prostaglandin transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
L-amino acid transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
pyrimidine nucleoside transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
choline transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
thiamine transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
putrescine transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
efflux transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
spermidine transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
quaternary ammonium group transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
toxin transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
xenobiotic transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
L-arginine transmembrane transporter activitySolute carrier family 22 member 2Homo sapiens (human)
ATP bindingATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type xenobiotic transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
glucuronoside transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type bile acid transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATP hydrolysis activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATPase-coupled transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
icosanoid transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
guanine nucleotide transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
protein bindingMultidrug resistance-associated protein 4Homo sapiens (human)
ATP bindingMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type xenobiotic transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
prostaglandin transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
urate transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
purine nucleotide transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type bile acid transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
efflux transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
15-hydroxyprostaglandin dehydrogenase (NAD+) activityMultidrug resistance-associated protein 4Homo sapiens (human)
ATP hydrolysis activityMultidrug resistance-associated protein 4Homo sapiens (human)
glutathione transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ATPase-coupled transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
xenobiotic transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
protein bindingBile salt export pumpHomo sapiens (human)
ATP bindingBile salt export pumpHomo sapiens (human)
ABC-type xenobiotic transporter activityBile salt export pumpHomo sapiens (human)
bile acid transmembrane transporter activityBile salt export pumpHomo sapiens (human)
canalicular bile acid transmembrane transporter activityBile salt export pumpHomo sapiens (human)
carbohydrate transmembrane transporter activityBile salt export pumpHomo sapiens (human)
ABC-type bile acid transporter activityBile salt export pumpHomo sapiens (human)
ATP hydrolysis activityBile salt export pumpHomo sapiens (human)
monooxygenase activityCytochrome P450 1A2Homo sapiens (human)
iron ion bindingCytochrome P450 1A2Homo sapiens (human)
protein bindingCytochrome P450 1A2Homo sapiens (human)
electron transfer activityCytochrome P450 1A2Homo sapiens (human)
oxidoreductase activityCytochrome P450 1A2Homo sapiens (human)
oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen, reduced flavin or flavoprotein as one donor, and incorporation of one atom of oxygenCytochrome P450 1A2Homo sapiens (human)
enzyme bindingCytochrome P450 1A2Homo sapiens (human)
heme bindingCytochrome P450 1A2Homo sapiens (human)
demethylase activityCytochrome P450 1A2Homo sapiens (human)
caffeine oxidase activityCytochrome P450 1A2Homo sapiens (human)
aromatase activityCytochrome P450 1A2Homo sapiens (human)
estrogen 16-alpha-hydroxylase activityCytochrome P450 1A2Homo sapiens (human)
estrogen 2-hydroxylase activityCytochrome P450 1A2Homo sapiens (human)
hydroperoxy icosatetraenoate dehydratase activityCytochrome P450 1A2Homo sapiens (human)
monooxygenase activityCytochrome P450 3A4Homo sapiens (human)
steroid bindingCytochrome P450 3A4Homo sapiens (human)
iron ion bindingCytochrome P450 3A4Homo sapiens (human)
protein bindingCytochrome P450 3A4Homo sapiens (human)
steroid hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
retinoic acid 4-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
oxidoreductase activityCytochrome P450 3A4Homo sapiens (human)
oxygen bindingCytochrome P450 3A4Homo sapiens (human)
enzyme bindingCytochrome P450 3A4Homo sapiens (human)
heme bindingCytochrome P450 3A4Homo sapiens (human)
vitamin D3 25-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
caffeine oxidase activityCytochrome P450 3A4Homo sapiens (human)
quinine 3-monooxygenase activityCytochrome P450 3A4Homo sapiens (human)
testosterone 6-beta-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
1-alpha,25-dihydroxyvitamin D3 23-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
anandamide 8,9 epoxidase activityCytochrome P450 3A4Homo sapiens (human)
anandamide 11,12 epoxidase activityCytochrome P450 3A4Homo sapiens (human)
anandamide 14,15 epoxidase activityCytochrome P450 3A4Homo sapiens (human)
aromatase activityCytochrome P450 3A4Homo sapiens (human)
vitamin D 24-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
estrogen 16-alpha-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
estrogen 2-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
1,8-cineole 2-exo-monooxygenase activityCytochrome P450 3A4Homo sapiens (human)
monooxygenase activityCytochrome P450 2D6Homo sapiens (human)
iron ion bindingCytochrome P450 2D6Homo sapiens (human)
oxidoreductase activityCytochrome P450 2D6Homo sapiens (human)
oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen, reduced flavin or flavoprotein as one donor, and incorporation of one atom of oxygenCytochrome P450 2D6Homo sapiens (human)
heme bindingCytochrome P450 2D6Homo sapiens (human)
anandamide 8,9 epoxidase activityCytochrome P450 2D6Homo sapiens (human)
anandamide 11,12 epoxidase activityCytochrome P450 2D6Homo sapiens (human)
anandamide 14,15 epoxidase activityCytochrome P450 2D6Homo sapiens (human)
monooxygenase activityCytochrome P450 2C9 Homo sapiens (human)
iron ion bindingCytochrome P450 2C9 Homo sapiens (human)
arachidonic acid epoxygenase activityCytochrome P450 2C9 Homo sapiens (human)
steroid hydroxylase activityCytochrome P450 2C9 Homo sapiens (human)
arachidonic acid 14,15-epoxygenase activityCytochrome P450 2C9 Homo sapiens (human)
arachidonic acid 11,12-epoxygenase activityCytochrome P450 2C9 Homo sapiens (human)
oxidoreductase activityCytochrome P450 2C9 Homo sapiens (human)
(S)-limonene 6-monooxygenase activityCytochrome P450 2C9 Homo sapiens (human)
(S)-limonene 7-monooxygenase activityCytochrome P450 2C9 Homo sapiens (human)
caffeine oxidase activityCytochrome P450 2C9 Homo sapiens (human)
(R)-limonene 6-monooxygenase activityCytochrome P450 2C9 Homo sapiens (human)
aromatase activityCytochrome P450 2C9 Homo sapiens (human)
heme bindingCytochrome P450 2C9 Homo sapiens (human)
oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen, reduced flavin or flavoprotein as one donor, and incorporation of one atom of oxygenCytochrome P450 2C9 Homo sapiens (human)
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)
monooxygenase activityCytochrome P450 2C19Homo sapiens (human)
iron ion bindingCytochrome P450 2C19Homo sapiens (human)
steroid hydroxylase activityCytochrome P450 2C19Homo sapiens (human)
oxidoreductase activityCytochrome P450 2C19Homo sapiens (human)
(S)-limonene 6-monooxygenase activityCytochrome P450 2C19Homo sapiens (human)
(S)-limonene 7-monooxygenase activityCytochrome P450 2C19Homo sapiens (human)
oxygen bindingCytochrome P450 2C19Homo sapiens (human)
enzyme bindingCytochrome P450 2C19Homo sapiens (human)
heme bindingCytochrome P450 2C19Homo sapiens (human)
(R)-limonene 6-monooxygenase activityCytochrome P450 2C19Homo sapiens (human)
aromatase activityCytochrome P450 2C19Homo sapiens (human)
long-chain fatty acid omega-1 hydroxylase activityCytochrome P450 2C19Homo sapiens (human)
arachidonic acid epoxygenase activityCytochrome P450 2C19Homo sapiens (human)
oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen, reduced flavin or flavoprotein as one donor, and incorporation of one atom of oxygenCytochrome P450 2C19Homo sapiens (human)
virus receptor activityC-C chemokine receptor type 5Homo sapiens (human)
actin bindingC-C chemokine receptor type 5Homo sapiens (human)
phosphatidylinositol phospholipase C activityC-C chemokine receptor type 5Homo sapiens (human)
chemokine receptor activityC-C chemokine receptor type 5Homo sapiens (human)
protein bindingC-C chemokine receptor type 5Homo sapiens (human)
coreceptor activityC-C chemokine receptor type 5Homo sapiens (human)
C-C chemokine receptor activityC-C chemokine receptor type 5Homo sapiens (human)
C-C chemokine bindingC-C chemokine receptor type 5Homo sapiens (human)
identical protein bindingC-C chemokine receptor type 5Homo sapiens (human)
chemokine (C-C motif) ligand 5 bindingC-C chemokine receptor type 5Homo 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)
organic cation transmembrane transporter activityMultidrug and toxin extrusion protein 2Homo sapiens (human)
antiporter activityMultidrug and toxin extrusion protein 2Homo sapiens (human)
transmembrane transporter activityMultidrug and toxin extrusion protein 2Homo sapiens (human)
xenobiotic transmembrane transporter activityMultidrug and toxin extrusion protein 2Homo sapiens (human)
polyspecific organic cation:proton antiporter activityMultidrug and toxin extrusion protein 2Homo sapiens (human)
protein bindingCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATP bindingCanalicular multispecific organic anion transporter 1Homo sapiens (human)
organic anion transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type xenobiotic transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bilirubin transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATP hydrolysis activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATPase-coupled transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
protein bindingMultidrug and toxin extrusion protein 1Homo sapiens (human)
organic cation transmembrane transporter activityMultidrug and toxin extrusion protein 1Homo sapiens (human)
L-amino acid transmembrane transporter activityMultidrug and toxin extrusion protein 1Homo sapiens (human)
thiamine transmembrane transporter activityMultidrug and toxin extrusion protein 1Homo sapiens (human)
antiporter activityMultidrug and toxin extrusion protein 1Homo sapiens (human)
putrescine transmembrane transporter activityMultidrug and toxin extrusion protein 1Homo sapiens (human)
transmembrane transporter activityMultidrug and toxin extrusion protein 1Homo sapiens (human)
xenobiotic transmembrane transporter activityMultidrug and toxin extrusion protein 1Homo sapiens (human)
L-arginine transmembrane transporter activityMultidrug and toxin extrusion protein 1Homo sapiens (human)
polyspecific organic cation:proton antiporter activityMultidrug and toxin extrusion protein 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (47)

Processvia Protein(s)Taxonomy
plasma membraneSolute carrier family 22 member 2Homo sapiens (human)
basal plasma membraneSolute carrier family 22 member 2Homo sapiens (human)
membraneSolute carrier family 22 member 2Homo sapiens (human)
basolateral plasma membraneSolute carrier family 22 member 2Homo sapiens (human)
apical plasma membraneSolute carrier family 22 member 2Homo sapiens (human)
extracellular exosomeSolute carrier family 22 member 2Homo sapiens (human)
presynapseSolute carrier family 22 member 2Homo sapiens (human)
plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
basal plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
basolateral plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
nucleolusMultidrug resistance-associated protein 4Homo sapiens (human)
Golgi apparatusMultidrug resistance-associated protein 4Homo sapiens (human)
plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
membraneMultidrug resistance-associated protein 4Homo sapiens (human)
basolateral plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
apical plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
platelet dense granule membraneMultidrug resistance-associated protein 4Homo sapiens (human)
external side of apical plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
basolateral plasma membraneBile salt export pumpHomo sapiens (human)
Golgi membraneBile salt export pumpHomo sapiens (human)
endosomeBile salt export pumpHomo sapiens (human)
plasma membraneBile salt export pumpHomo sapiens (human)
cell surfaceBile salt export pumpHomo sapiens (human)
apical plasma membraneBile salt export pumpHomo sapiens (human)
intercellular canaliculusBile salt export pumpHomo sapiens (human)
intracellular canaliculusBile salt export pumpHomo sapiens (human)
recycling endosomeBile salt export pumpHomo sapiens (human)
recycling endosome membraneBile salt export pumpHomo sapiens (human)
extracellular exosomeBile salt export pumpHomo sapiens (human)
membraneBile salt export pumpHomo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 1A2Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 1A2Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 1A2Homo sapiens (human)
cytoplasmCytochrome P450 3A4Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 3A4Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 3A4Homo sapiens (human)
mitochondrionCytochrome P450 2D6Homo sapiens (human)
endoplasmic reticulumCytochrome P450 2D6Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 2D6Homo sapiens (human)
cytoplasmCytochrome P450 2D6Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2D6Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 2C9 Homo sapiens (human)
plasma membraneCytochrome P450 2C9 Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2C9 Homo sapiens (human)
cytoplasmCytochrome P450 2C9 Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2C9 Homo sapiens (human)
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)
endoplasmic reticulum membraneCytochrome P450 2C19Homo sapiens (human)
plasma membraneCytochrome P450 2C19Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2C19Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2C19Homo sapiens (human)
cytoplasmCytochrome P450 2C19Homo sapiens (human)
cell surfaceC-C chemokine receptor type 5Homo sapiens (human)
endosomeC-C chemokine receptor type 5Homo sapiens (human)
plasma membraneC-C chemokine receptor type 5Homo sapiens (human)
external side of plasma membraneC-C chemokine receptor type 5Homo sapiens (human)
cell surfaceC-C chemokine receptor type 5Homo sapiens (human)
external side of plasma membraneC-C chemokine receptor type 5Homo sapiens (human)
cytoplasmC-C chemokine receptor type 5Homo sapiens (human)
early endosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
endoplasmic reticulumPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cytoplasmPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
lysosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
early endosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
late endosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
endoplasmic reticulumPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
basolateral plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
apical plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
transport vesiclePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cytoplasmic vesicle membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
neuron projectionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
neuronal cell bodyPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane raftPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
ciliary basePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
lumenal side of membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
basolateral part of cellPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
monoatomic ion channel complexPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cell surfacePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
perinuclear region of cytoplasmPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
inward rectifier potassium channel complexPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
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)
plasma membraneMultidrug and toxin extrusion protein 2Homo sapiens (human)
apical plasma membraneMultidrug and toxin extrusion protein 2Homo sapiens (human)
membraneMultidrug and toxin extrusion protein 2Homo sapiens (human)
plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
cell surfaceCanalicular multispecific organic anion transporter 1Homo sapiens (human)
apical plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
intercellular canaliculusCanalicular multispecific organic anion transporter 1Homo sapiens (human)
apical plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
plasma membraneMultidrug and toxin extrusion protein 1Homo sapiens (human)
basolateral plasma membraneMultidrug and toxin extrusion protein 1Homo sapiens (human)
apical plasma membraneMultidrug and toxin extrusion protein 1Homo sapiens (human)
membraneMultidrug and toxin extrusion protein 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (316)

Assay IDTitleYearJournalArticle
AID1713963Antiviral activity against HIV-1 BAL infected in human macrophages assessed as reduction in p24 production at 500 nM treated for 1 hr prior to viral infection and measured after 5 days by ELISA2016Bioorganic & medicinal chemistry, 11-15, Volume: 24, Issue:22
Exploration of bivalent ligands targeting putative mu opioid receptor and chemokine receptor CCR5 dimerization.
AID1400647Tmax in Sprague-Dawley rat at 10 mg/kg, po by LC-MS/MS analysis2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400690Half life in dog at 0.5 mg/kg, iv by LC-MS/MS analysis2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400622Antiviral activity against CCR5-dependent HIV1 SF162 infected in human HOS cells assessed as reduction in virus infection measured after 5 days by luciferase reporter gene assay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400623Cytotoxicity against human HOS cells after 48 hrs by MTT assay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400616Antiviral activity against CCR5-dependent HIV1 SF162 infected in human TZM-bl cells assessed as reduction in virus infection measured after 48 hrs post infection by luciferase reporter gene assay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400608Drug metabolism in dog liver cells assessed as CYP450-mediated 4,4-difluoro-N-((1S)-1-(4-hydroxyphenyl)-3-(3-(3-isopropyl-5-methyl-4H-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]octan-8-yl)propyl)cyclohexanecarboxamide metabolite levels by Q-TOF mass spectrome2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400618Therapeutic index, ratio of CC50 for human TZM-bl cells to EC50 for CCR5-dependent HIV1 SF162 infected in human TZM-bl cells2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400632Cytotoxicity against human PM1 cells after 48 hrs by MTT assay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400665Metabolic stability in rat hepatocytes assessed as compound remaining by UPLC/Q-TOF mass spectrometric analysis2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400650Half life in Sprague-Dawley rat at 1 mg/kg, iv by LC-MS/MS analysis2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400605Oral bioavailability in rat at 10 mg/kg administered as single dose2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400620Cytotoxicity against human PBMC after 48 hrs by MTT assay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1713975Antiviral activity against HIV-1 SF162 infected in human astrocytes assessed as reduction in viral invasion by measuring decrease in Tat protein expression at 100 nM co-treated with morphine prior to viral infection and measured after 18 hrs by luciferase2016Bioorganic & medicinal chemistry, 11-15, Volume: 24, Issue:22
Exploration of bivalent ligands targeting putative mu opioid receptor and chemokine receptor CCR5 dimerization.
AID1400613Drug metabolism in rat liver cells assessed as CYP450-mediated 3-(3-isopropyl-5-methyl-4H-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]octane metabolite levels by Q-TOF mass spectrometric analysis2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400612Drug metabolism in human liver cells assessed as CYP450-mediated 4,4-difluoro-N-((1S)-3-(3-(3-(hydroxymethyl)-5-isopropyl-4H-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]octan-8-yl)-1-(4-hydroxyphenyl)propyl)cyclohexanecarboxamide metabolite levels by Q-TOF mas2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1713969Antiviral activity against HIV-1 SF162 infected in human astrocytes assessed as reduction in viral invasion by measuring decrease in Tat protein expression at 100 nM treated prior to viral infection and measured after 18 hrs by luciferase based assay2016Bioorganic & medicinal chemistry, 11-15, Volume: 24, Issue:22
Exploration of bivalent ligands targeting putative mu opioid receptor and chemokine receptor CCR5 dimerization.
AID1713979Antagonist activity at CCR5 receptor in CHO cells co-expressing hMOR, CD4 and luciferase reporter assessed as inhibition of CD4-gp120 complex interaction with receptor dimer by measuring inhibition of cell-cell fusion process at 100 nM measured after co-i2016Bioorganic & medicinal chemistry, 11-15, Volume: 24, Issue:22
Exploration of bivalent ligands targeting putative mu opioid receptor and chemokine receptor CCR5 dimerization.
AID1400648AUC (0 to infinity) in Sprague-Dawley rat at 10 mg/kg, po by LC-MS/MS analysis2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400692AUC (0 to infinity) in dog at 2 mg/kg, po by LC-MS/MS analysis2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400633Therapeutic index, ratio of CC50 for human PM1 cells to IC50 for CCR5-dependent HIV1 Ba-L infected in human PM1 cells2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1713967Antagonist activity at CCR5 receptor in human MOLT4 cells transfected with Gqi5 assessed as inhibition of RANTES-induced calcium mobilization incubated for 15 mins prior to RANTES addition and measured for 120 secs by Fluo-4AM dye based fluorescence analy2016Bioorganic & medicinal chemistry, 11-15, Volume: 24, Issue:22
Exploration of bivalent ligands targeting putative mu opioid receptor and chemokine receptor CCR5 dimerization.
AID1400629Antiviral activity against CCR5-dependent HIV1 Ba-L infected in human HOS cells assessed as reduction in virus infection measured after 5 days by luciferase reporter gene assay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400621Therapeutic index, ratio of CC50 for human PBMC to IC50 for CCR5-dependent HIV1 SF162 infected in human PBMC cells2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400625Antiviral activity against CCR5-dependent HIV1 KM018 infected in human PBMC assessed as reduction in virus infection measured after 7 days by luciferase reporter gene assay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1713964Antiviral activity against HIV-1 BAL infected in human PBMC assessed as reduction in p24 production at 500 nM treated for 1 hr prior to viral infection and measured after 5 days by ELISA2016Bioorganic & medicinal chemistry, 11-15, Volume: 24, Issue:22
Exploration of bivalent ligands targeting putative mu opioid receptor and chemokine receptor CCR5 dimerization.
AID1400688Cmax in dog at 2 mg/kg, po by LC-MS/MS analysis2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400662Metabolic stability in human hepatocytes assessed as compound remaining by UPLC/Q-TOF mass spectrometric analysis2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400651Clearance in Sprague-Dawley rat at 1 mg/kg, iv by LC-MS/MS analysis2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400631Antiviral activity against CCR5-dependent HIV1 Ba-L infected in human PM1 cells assessed as reduction in virus infection measured after 5 days by luciferase reporter gene assay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400626Therapeutic index, ratio of CC50 for human PBMC to IC50 for CCR5-dependent HIV1 KM018 infected in human PBMC2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400627Antiviral activity against CCR5-dependent HIV1 Ba-L infected in human PBMC assessed as reduction in virus infection measured 7 days by luciferase reporter gene assay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400624Therapeutic index, ratio of CC50 for human HOS cells to IC50 for CCR5-dependent HIV1 SF162 infected in human HOS cells2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400611Drug metabolism in dog liver cells assessed as CYP450-mediated 4,4-difluoro-N-((1S)-3-(3-(3-(hydroxymethyl)-5-isopropyl-4H-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]octan-8-yl)-1-(4-hydroxyphenyl)propyl)cyclohexanecarboxamide metabolite levels by Q-TOF mass 2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1713980Antagonist activity at CCR5 receptor in CHO cells co-expressing hMOR, CD4 and luciferase reporter assessed as inhibition of CD4-gp120 complex interaction with receptor dimer by measuring inhibition of cell-cell fusion process at 100 nM measured after co-i2016Bioorganic & medicinal chemistry, 11-15, Volume: 24, Issue:22
Exploration of bivalent ligands targeting putative mu opioid receptor and chemokine receptor CCR5 dimerization.
AID1400691Clearance in dog at 0.5 mg/kg, iv by LC-MS/MS analysis2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400603Cmax in rat at 10 mg/kg, po administered as single dose2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400609Drug metabolism in human liver cells assessed as CYP450-mediated 4,4-difluoro-N-((1S)-1-(4-hydroxyphenyl)-3-(3-(3-isopropyl-5-methyl-4H-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]octan-8-yl)propyl)cyclohexanecarboxamide metabolite levels by Q-TOF mass spectro2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400615Drug metabolism in human liver cells assessed as CYP450-mediated 3-(3-isopropyl-5-methyl-4H-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]octane metabolite levels by Q-TOF mass spectrometric analysis2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1713968Displacement of [125I]MIP-1alpha from CCR5 receptor in rhesus monkey membrane incubated for 120 mins by liquid scintillation counting analysis2016Bioorganic & medicinal chemistry, 11-15, Volume: 24, Issue:22
Exploration of bivalent ligands targeting putative mu opioid receptor and chemokine receptor CCR5 dimerization.
AID1400617Cytotoxicity against human TZM-bl cells after 48 hrs by MTT assay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400607Drug metabolism in rat liver cells assessed as CYP450-mediated 4,4-difluoro-N-((1S)-1-(4-hydroxyphenyl)-3-(3-(3-isopropyl-5-methyl-4H-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]octan-8-yl)propyl)cyclohexanecarboxamide metabolite levels by Q-TOF mass spectrome2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400649Oral bioavailability in Sprague-Dawley rat at 10 mg/kg by LC-MS/MS analysis2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400630Therapeutic index, ratio of CC50 for human HOS cells to IC50 for CCR5-dependent HIV1 Ba-L infected in human HOS cells2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400610Drug metabolism in rat liver cells assessed as CYP450-mediated 4,4-difluoro-N-((1S)-3-(3-(3-(hydroxymethyl)-5-isopropyl-4H-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]octan-8-yl)-1-(4-hydroxyphenyl)propyl)cyclohexanecarboxamide metabolite levels by Q-TOF mass 2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400614Drug metabolism in dog liver cells assessed as CYP450-mediated 3-(3-isopropyl-5-methyl-4H-1,2,4-triazol-4-yl)-8-azabicyclo[3.2.1]octane metabolite levels by Q-TOF mass spectrometric analysis2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400693Oral bioavailability in dog at 2 mg/kg by LC-MS/MS analysis2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400606Antagonist activity at CCR5 (unknown origin) expressed in HEK293 cells co-expressing Galpha16 assessed as inhibition of RANTES-induced calcium flux preincubated for 10 mins followed by RANTES addition by fluo-4AM-based fluorescence assay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400604AUC in rat at 10 mg/kg, po administered as single dose2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400628Therapeutic index, ratio of CC50 for human PBMC to IC50 for CCR5-dependent HIV1 Ba-L infected in human PBMC2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400689Tmax in dog at 2 mg/kg, po by LC-MS/MS analysis2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400619Antiviral activity against CCR5-dependent HIV1 SF162 infected in human PBMC assessed as reduction in virus infection measured after 7 days by luciferase reporter gene assay2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400664Metabolic stability in dog hepatocytes assessed as compound remaining by UPLC/Q-TOF mass spectrometric analysis2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1400646Cmax in Sprague-Dawley rat at 10 mg/kg, po by LC-MS/MS analysis2018Journal of medicinal chemistry, 11-08, Volume: 61, Issue:21
Structure-Based Design of 1-Heteroaryl-1,3-propanediamine Derivatives as a Novel Series of CC-Chemokine Receptor 5 Antagonists.
AID1654459Drug metabolism in human liver microsomes assessed as CYP3A5*1/*1-mediated formation of (1S,2S)-2-OH-maraviroc at 8 uM incubated for 15 mins in presence of ketoconazole by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID554142Unbound clearance in rat at 1 mg/kg, iv and 10 mg/kg, po2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID554144Oral bioavailability in rat at 10 mg/kg2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID393050Antagonist activity at human recombinant CCR5 expressed in human HeLa-P4 cells assessed as inhibition of human HeLa-P4 cells binding to HIV1 gp160 expressing CHO cells by cell-cell fusion assay2009Bioorganic & medicinal chemistry letters, Feb-15, Volume: 19, Issue:4
1-Amido-1-phenyl-3-piperidinylbutanes - CCR5 antagonists for the treatment of HIV. Part 1.
AID320905Antiviral activity against HIV1 RU570 by viral entry assay2008Bioorganic & medicinal chemistry letters, Feb-15, Volume: 18, Issue:4
Anti-HIV-1 entry optimization of novel imidazopiperidine-tropane CCR5 antagonists.
AID1897764Therapeutic index, ratio of CC50 for human PM1 cells to EC50 for antiviral activity against HIV-1 YU-22022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID393051Antiviral activity against HIV1 Bal infected in human PM1 cells assessed as inhibition of viral replication2009Bioorganic & medicinal chemistry letters, Feb-15, Volume: 19, Issue:4
1-Amido-1-phenyl-3-piperidinylbutanes - CCR5 antagonists for the treatment of HIV. Part 1.
AID776010Antiviral activity against Human immunodeficiency virus 1 clade O isolate I-2478B infected in PHA-stimulated human PBMC after 10 days by ELISA2013Journal of medicinal chemistry, Oct-24, Volume: 56, Issue:20
Design of substituted imidazolidinylpiperidinylbenzoic acids as chemokine receptor 5 antagonists: potent inhibitors of R5 HIV-1 replication.
AID776011Antiviral activity against Human immunodeficiency virus 1 clade G isolate BZ163 infected in PHA-stimulated human PBMC after 10 days by ELISA2013Journal of medicinal chemistry, Oct-24, Volume: 56, Issue:20
Design of substituted imidazolidinylpiperidinylbenzoic acids as chemokine receptor 5 antagonists: potent inhibitors of R5 HIV-1 replication.
AID554137Intrinsic clearance in human liver microsomes assessed per mg of protein2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID776013Antiviral activity against Human immunodeficiency virus 1 clade C isolate ETH2220 infected in PHA-stimulated human PBMC after 10 days by ELISA2013Journal of medicinal chemistry, Oct-24, Volume: 56, Issue:20
Design of substituted imidazolidinylpiperidinylbenzoic acids as chemokine receptor 5 antagonists: potent inhibitors of R5 HIV-1 replication.
AID554163Intrinsic clearance in dog liver microsomes assessed per mg of protein2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID698308Antiviral activity against HIV1 clade C (SM145) infected in human PBMCs assessed as inhibition of p24 antigen production by ELISA2012Journal 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.
AID1897758Antiviral activity against HIV-1 SF162 infected in human TZM-bl cells measured after 48 hrs incubation by luciferase based analysis2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID1897772Therapeutic index, ratio of CC50 for human PBMC to EC50 for antiviral activity against HIV-1 KIZ006 clinical isolate2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID776061Antiviral activity against Human immunodeficiency virus 1 clade A isolate UG273 infected in PHA-stimulated human PBMC after 10 days by ELISA2013Journal of medicinal chemistry, Oct-24, Volume: 56, Issue:20
Design of substituted imidazolidinylpiperidinylbenzoic acids as chemokine receptor 5 antagonists: potent inhibitors of R5 HIV-1 replication.
AID776012Antiviral activity against Human immunodeficiency virus 1 clade E isolate ID12 infected in PHA-stimulated human PBMC after 10 days by ELISA2013Journal of medicinal chemistry, Oct-24, Volume: 56, Issue:20
Design of substituted imidazolidinylpiperidinylbenzoic acids as chemokine receptor 5 antagonists: potent inhibitors of R5 HIV-1 replication.
AID320902Antiviral activity against HIV1 ASM80 by viral entry assay2008Bioorganic & medicinal chemistry letters, Feb-15, Volume: 18, Issue:4
Anti-HIV-1 entry optimization of novel imidazopiperidine-tropane CCR5 antagonists.
AID395488Apparent permeability from apical to basolateral side of human Caco-2 cells2009Bioorganic & medicinal chemistry letters, Mar-01, Volume: 19, Issue:5
1-Amido-1-phenyl-3-piperidinylbutanes--CCR5 antagonists for the treatment of HIV: part 2.
AID499099Antiviral activity against HIV1 UG273 clade A, R5 in human PBMC assessed as p24 antigen level by ELISA2010Antimicrobial agents and chemotherapy, Jan, Volume: 54, Issue:1
Long-lasting enfuvirtide carrier pentasaccharide conjugates with potent anti-human immunodeficiency virus type 1 activity.
AID1897792Half life in Sprague-Dawley rat at 2 mg/kg, iv measured upto 24 hrs2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID499095Antiviral activity against HIV1 NL4.3 in human PBMC assessed as p24 antigen level by ELISA2010Antimicrobial agents and chemotherapy, Jan, Volume: 54, Issue:1
Long-lasting enfuvirtide carrier pentasaccharide conjugates with potent anti-human immunodeficiency virus type 1 activity.
AID554143Half life in rat at 1 mg/kg, iv and 10 mg/kg, po2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID1654488AUC(0 to infinity) in human carrying CYP3A5*3/*3 allele at 300 mg, po measured up to 32 hrs2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID776007Antiviral activity against Human immunodeficiency virus 1 clade B isolate CI#15 infected in PHA-stimulated human PBMC after 10 days by ELISA2013Journal of medicinal chemistry, Oct-24, Volume: 56, Issue:20
Design of substituted imidazolidinylpiperidinylbenzoic acids as chemokine receptor 5 antagonists: potent inhibitors of R5 HIV-1 replication.
AID698307Antiviral activity against HIV1 clade A/E (ID12) infected in human PBMCs assessed as inhibition of p24 antigen production by ELISA2012Journal 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.
AID698152Antiviral activity against HIV1 clade A (UG273) infected in human PBMCs assessed as inhibition of p24 antigen production by ELISA2012Journal 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.
AID721752Inhibition of human MATE2K-mediated ASP+ uptake expressed in HEK293 cells after 1.5 mins by fluorescence assay2013Journal of medicinal chemistry, Feb-14, Volume: 56, Issue:3
Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling.
AID776005Antiviral activity against Human immunodeficiency virus 1 clade B isolate BaL infected in PHA-stimulated human PBMC after 10 days by ELISA2013Journal of medicinal chemistry, Oct-24, Volume: 56, Issue:20
Design of substituted imidazolidinylpiperidinylbenzoic acids as chemokine receptor 5 antagonists: potent inhibitors of R5 HIV-1 replication.
AID250740Mean decrease in viral load in humans after administration of 100 mg twice a day; expressed as log10 reduction2005Journal of medicinal chemistry, Mar-10, Volume: 48, Issue:5
New approaches toward anti-HIV chemotherapy.
AID1207345Inhibition of fast sodium current (INa) in HEK293 cells transfected with human Nav1.5 measured using IonWorks Quattro automated patch clamp platform
AID1654469Drug metabolism in human liver microsomes assessed as formation of (1S,3S)-3-OH-maraviroc at 8 uM incubated for 15 mins in presence of ketoconazole by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID776006Antiviral activity against Human immunodeficiency virus 1 clade B isolate CI#19 infected in PHA-stimulated human PBMC after 10 days by ELISA2013Journal of medicinal chemistry, Oct-24, Volume: 56, Issue:20
Design of substituted imidazolidinylpiperidinylbenzoic acids as chemokine receptor 5 antagonists: potent inhibitors of R5 HIV-1 replication.
AID1897765Therapeutic index, ratio of CC50 for human PM1 cells to EC50 for antiviral activity against HIV-1 SF1622022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID1207529Inhibition of rapid delayed inward rectifying potassium current (IKr) measured using manual patch clamp assay
AID397744Inhibition of human ERG channel at 300 uM2009Journal of medicinal chemistry, Jul-23, Volume: 52, Issue:14
Side chain flexibilities in the human ether-a-go-go related gene potassium channel (hERG) together with matched-pair binding studies suggest a new binding mode for channel blockers.
AID1897757Cytotoxicity against human PBMC cells incubated for 48 hrs by MTT assay2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID1473741Inhibition of human MRP4 overexpressed in Sf9 cell membrane vesicles assessed as uptake of [3H]-estradiol-17beta-D-glucuronide in presence of ATP and GSH measured after 20 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1191661Antiviral activity against HIV1 infected in human SupT1 cells exposed to supernatant from HIV1 infected human 293T cells incubated for 48 hrs by firefly luciferase assay based single-cycle HIV1 replication assay2015Bioorganic & medicinal chemistry, Mar-01, Volume: 23, Issue:5
Design, synthesis, and biological evaluation of novel 2-methylpiperazine derivatives as potent CCR5 antagonists.
AID1897762Antiviral activity against HIV-1 Ba-L infected in human PM1 cells measured after 48 hrs incubation by ELISA based p24 antigen assay2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID1654478Drug metabolism in CYP3cide-treated human liver microsomes assessed as CYP3A5*1/*1-mediated formation of (1S,3R)-3-OH-MVC-maraviroc at 8 uM incubated for 15 mins by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1654486Drug metabolism in CYP3cide-treated human liver microsomes assessed as CYP3A5*3/*3-mediated formation of hydroxymethyl-maraviroc at 8 uM incubated for 15 mins by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID499097Antiviral activity against HIV1 BZ167 in human PBMC assessed as p24 antigen level by ELISA2010Antimicrobial agents and chemotherapy, Jan, Volume: 54, Issue:1
Long-lasting enfuvirtide carrier pentasaccharide conjugates with potent anti-human immunodeficiency virus type 1 activity.
AID465010Displacement of [128I]RANTES from human CCR5 receptor coexpressed with Galphai6 in CHO cells after 2 hrs by scintillation counting2010Bioorganic & medicinal chemistry letters, Mar-01, Volume: 20, Issue:5
Evaluation of a 3-amino-8-azabicyclo[3.2.1]octane replacement in the CCR5 antagonist maraviroc.
AID393053Metabolic stability in human liver microsomes assessed as clearance measured per mg of protein2009Bioorganic & medicinal chemistry letters, Feb-15, Volume: 19, Issue:4
1-Amido-1-phenyl-3-piperidinylbutanes - CCR5 antagonists for the treatment of HIV. Part 1.
AID710734Antagonist activity against CXCR4 expressed in U87.CD.CXCR4 cells assessed as inhibition of CXCL12-induced calcium signaling incubated for 10 mins by FLIPR2012Journal of medicinal chemistry, Dec-13, Volume: 55, Issue:23
HIV-1 X4 activities of polycationic "viologen" based dendrimers by interaction with the chemokine receptor CXCR4: study of structure-activity relationship.
AID479416Antiviral activity against HIV1 NL4-3 infected in human JC53-BL cells assessed as luciferase activity after 3 days post infection2010Bioorganic & medicinal chemistry letters, May-15, Volume: 20, Issue:10
Novel hexahydropyrrolo[3,4-c]pyrrole CCR5 antagonists.
AID776014Antiviral activity against Human immunodeficiency virus 1 clade B isolate US2 infected in PHA-stimulated human PBMC after 10 days by ELISA2013Journal of medicinal chemistry, Oct-24, Volume: 56, Issue:20
Design of substituted imidazolidinylpiperidinylbenzoic acids as chemokine receptor 5 antagonists: potent inhibitors of R5 HIV-1 replication.
AID554152Absorption in dog at 0.5 mg/kg, iv and 1 mg/kg, po2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID1897759Antiviral activity against HIV-1 YU-2 infected in human TZM-bl cells measured after 48 hrs incubation by luciferase based analysis2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID1353545Displacement of [125I]-RANTES from CCR5 in mouse NIH/3T3 cells after 1 hr2018European journal of medicinal chemistry, Mar-10, Volume: 147Recent updates for designing CCR5 antagonists as anti-retroviral agents.
AID707904Binding affinity to CCR52012Journal of medicinal chemistry, Nov-26, Volume: 55, Issue:22
Chemokine receptor antagonists.
AID625276FDA Liver Toxicity Knowledge Base Benchmark Dataset (LTKB-BD) drugs of most concern for DILI2011Drug discovery today, Aug, Volume: 16, Issue:15-16
FDA-approved drug labeling for the study of drug-induced liver injury.
AID1654470Drug metabolism in CYP3cide-treated human liver microsomes assessed as formation of (1S,3S)-3-OH-maraviroc at 8 uM incubated for 15 mins by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1897767Antiviral activity against HIV-1 KM018 clinical isolate infected in human PBMC measured after 48 hrs incubation by luciferase based analysis2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID393056Displacement of [3H]dofetilide from human ERG expressed in HEK293 cells at 300 nM2009Bioorganic & medicinal chemistry letters, Feb-15, Volume: 19, Issue:4
1-Amido-1-phenyl-3-piperidinylbutanes - CCR5 antagonists for the treatment of HIV. Part 1.
AID1654457Drug metabolism in human liver microsomes assessed as formation of (1S,2S)-2-OH-maraviroc at 8 uM incubated for 15 mins in presence of ketoconazole by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1207315Inhibition 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
AID1654483Drug metabolism in human liver microsomes assessed as CYP3A5*1/*1-mediated formation of hydroxymethyl-maraviroc at 8 uM incubated for 15 mins in presence of ketoconazole by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID554139Antiviral activity against HIV Ba-L infected in PBMC2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID1897789AUC (0 to infinitive) in Sprague-Dawley rat at 10 mg/kg, po measured upto 24 hrs2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID270504Distribution coefficient, log D of the compound2006Bioorganic & medicinal chemistry letters, Sep-01, Volume: 16, Issue:17
Overcoming HERG affinity in the discovery of the CCR5 antagonist maraviroc.
AID465011Antiviral activity against CCR5-tropic recombinant HIV1 NLBal virus infected in JC53-BL cells assessed as inhibition of viral replication after 3 days by luciferase reporter gene assay2010Bioorganic & medicinal chemistry letters, Mar-01, Volume: 20, Issue:5
Evaluation of a 3-amino-8-azabicyclo[3.2.1]octane replacement in the CCR5 antagonist maraviroc.
AID1654474Drug metabolism in CYP3cide-treated human liver microsomes assessed as CYP3A5*3/*3-mediated formation of (1S,3S)-3-OH-maraviroc at 8 uM incubated for 15 mins by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1207435Inhibition 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
AID1654485Drug metabolism in human liver microsomes assessed as CYP3A5*3/*3-mediated formation of hydroxymethyl-maraviroc at 8 uM incubated for 15 mins in presence of ketoconazole by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1654489AUC(0 to infinity) in human carrying CYP3A5*1/*3 allele at 300 mg, po measured up to 32 hrs2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1654458Drug metabolism in CYP3cide-treated human liver microsomes assessed as formation of (1S,2S)-2-OH-maraviroc at 8 uM incubated for 15 mins by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1897803Inhibition of CYP3A4 (unknown origin) using midazolam as substrate2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID320903Antiviral activity against HIV1 BaL by viral entry assay2008Bioorganic & medicinal chemistry letters, Feb-15, Volume: 18, Issue:4
Anti-HIV-1 entry optimization of novel imidazopiperidine-tropane CCR5 antagonists.
AID1897761Therapeutic index, ratio of CC50 for human HOS-CD4-CCR5 cells to EC50 for antiviral activity against HIV-1 Ba-L2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID554141Clearance in rat at 1 mg/kg, iv and 10 mg/kg, po2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID270500Inhibition of [3H]dofetilide binding to HERG expressed in HEK293 cells at 300 nM2006Bioorganic & medicinal chemistry letters, Sep-01, Volume: 16, Issue:17
Overcoming HERG affinity in the discovery of the CCR5 antagonist maraviroc.
AID776009Antiviral activity against Human immunodeficiency virus 1 clade B isolate SF162 infected in PHA-stimulated human PBMC after 10 days by ELISA2013Journal of medicinal chemistry, Oct-24, Volume: 56, Issue:20
Design of substituted imidazolidinylpiperidinylbenzoic acids as chemokine receptor 5 antagonists: potent inhibitors of R5 HIV-1 replication.
AID721751Inhibition of human OCT2-mediated ASP+ uptake expressed in HEK293 cells after 3 mins by fluorescence assay2013Journal of medicinal chemistry, Feb-14, Volume: 56, Issue:3
Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling.
AID1654472Drug metabolism in CYP3cide-treated human liver microsomes assessed as CYP3A5*1/*1-mediated formation of (1S,3S)-3-OH-maraviroc at 8 uM incubated for 15 mins by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1897802Inhibition of CYP2D6 (unknown origin)2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID1473738Inhibition of human BSEP overexpressed in Sf9 cell membrane vesicles assessed as uptake of [3H]-taurocholate in presence of ATP measured after 15 to 20 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID776008Antiviral activity against Human immunodeficiency virus 1 clade B isolate Ada infected in PHA-stimulated human PBMC after 10 days by ELISA2013Journal of medicinal chemistry, Oct-24, Volume: 56, Issue:20
Design of substituted imidazolidinylpiperidinylbenzoic acids as chemokine receptor 5 antagonists: potent inhibitors of R5 HIV-1 replication.
AID1065006Antagonist activity against CCR5 receptor in human TZM-bl cells assessed as inhibition of HIV-1 JR-FL-induced cell-cell fusion between viral envelope protein expressing human HEK293 cells to TZM-bl cells after 48 hrs by luciferase reporter gene assay2014ACS medicinal chemistry letters, Feb-13, Volume: 5, Issue:2
Preparation and activities of macromolecule conjugates of the CCR5 antagonist Maraviroc.
AID554058Tmax in rat at 10 mg/kg, po2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID1897771Antiviral activity against HIV-1 KIZ006 clinical isolate infected in human PBMC measured after 48 hrs incubation by luciferase based analysis2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID1594898Half life in human liver microsomes2019Journal of medicinal chemistry, 06-13, Volume: 62, Issue:11
Applications of Deuterium in Medicinal Chemistry.
AID1654475Drug metabolism in human liver microsomes assessed as formation of (1S,3R)-3-OH-MVC-maraviroc at 8 uM incubated for 15 mins in presence of ketoconazole by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1897786Tmax in Sprague-Dawley rat at 10 mg/kg, po measured upto 24 hrs2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID652438Binding affinity to CCR52011ACS medicinal chemistry letters, Jul-14, Volume: 2, Issue:7
Screening for GPCR Ligands Using Surface Plasmon Resonance.
AID1654463Drug metabolism in human liver microsomes assessed as formation of (1R,2R)-2-OH-maraviroc at 8 uM incubated for 15 mins in presence of ketoconazole by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID320904Antiviral activity against HIV1 97-ZA-003 by viral entry assay2008Bioorganic & medicinal chemistry letters, Feb-15, Volume: 18, Issue:4
Anti-HIV-1 entry optimization of novel imidazopiperidine-tropane CCR5 antagonists.
AID270503Antiviral activity against HIV BaL in PM1 cell2006Bioorganic & medicinal chemistry letters, Sep-01, Volume: 16, Issue:17
Overcoming HERG affinity in the discovery of the CCR5 antagonist maraviroc.
AID1207375Inhibition 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
AID395483Antagonist activity at human recombinant CCR5 expressed in human HeLa-P4 cells assessed as inhibition of human HeLa-P4 cells binding to HIV1 gp160 expressing CHO cells by cell-cell fusion assay2009Bioorganic & medicinal chemistry letters, Mar-01, Volume: 19, Issue:5
1-Amido-1-phenyl-3-piperidinylbutanes--CCR5 antagonists for the treatment of HIV: part 2.
AID499101Antiviral activity against HIV1 ETH2220 in human PBMC assessed as p24 antigen level by ELISA2010Antimicrobial agents and chemotherapy, Jan, Volume: 54, Issue:1
Long-lasting enfuvirtide carrier pentasaccharide conjugates with potent anti-human immunodeficiency virus type 1 activity.
AID1897751Antagonist activity at CCR5 (unknown origin) expressed in HEK293 cells co-expressing Galpha16 assessed as intracellular calcium change incubated for 15 mins by Fluo-4 AM calcium flux assay2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID1654481Drug metabolism in human liver microsomes assessed as formation of hydroxymethyl-maraviroc at 8 uM incubated for 15 mins in presence of ketoconazole by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1897797Half life in Sprague-Dawley rat at 1 mg/kg, iv measured upto 24 hrs2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID393054Dissociation constant, pKa of the compound2009Bioorganic & medicinal chemistry letters, Feb-15, Volume: 19, Issue:4
1-Amido-1-phenyl-3-piperidinylbutanes - CCR5 antagonists for the treatment of HIV. Part 1.
AID1654482Drug metabolism in CYP3cide-treated human liver microsomes assessed as formation of hydroxymethyl-maraviroc at 8 uM incubated for 15 mins by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1207563Inhibition of long-lasting type calcium current (hICa) in Chinese Hamster Ovary (CHO) cells expressing hCav1.2 measured using IonWorks Quattro automated patch clamp platform
AID554132Antagonist activity at CCR5 receptor expressed in HeLa-P4 cells co-expressing CD4 assessed as inhibition of infusion to HIV gp120 expressed in CHO-tat10 cells after 20 hrs by cell-cell fusion assay2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID698149Antiviral activity against HIV1 clade G (BCF-DIOUM) infected in human PBMCs assessed as inhibition of p24 antigen production by ELISA2012Journal 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.
AID1191659Antagonist activity against CCR5 (unknown origin) expressed in CHO cells co-expressing Galpha16 incubated for 10 mins assessed as inhibition of RANTES-induced calcium mobilization2015Bioorganic & medicinal chemistry, Mar-01, Volume: 23, Issue:5
Design, synthesis, and biological evaluation of novel 2-methylpiperazine derivatives as potent CCR5 antagonists.
AID554060fCmax in rat at 10 mg/kg, po2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID270506Inhibition of [3H]dofetilide binding to HERG expressed in HEK293 cells at 1000 nM2006Bioorganic & medicinal chemistry letters, Sep-01, Volume: 16, Issue:17
Overcoming HERG affinity in the discovery of the CCR5 antagonist maraviroc.
AID1897773Antiviral activity against HIV-1 YU-2 (G140S/Q148H) infected in human TZM-bl cells measured after 48 hrs incubation by luciferase based analysis2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID395486Antiviral activity against HIV1 BAL assessed as inhibition of viral replication2009Bioorganic & medicinal chemistry letters, Mar-01, Volume: 19, Issue:5
1-Amido-1-phenyl-3-piperidinylbutanes--CCR5 antagonists for the treatment of HIV: part 2.
AID1897763Therapeutic index, ratio of CC50 for human PM1 cells to EC50 for antiviral activity against HIV-1 Ba-L2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID1207469Inhibition 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
AID1207285Inhibition of long-lasting type calcium current (ICaL) in HEK293 cells (alpha1C/beta2a/alpha2delta1) cells measured using IonWorks Barracuda automated patch clamp platform
AID1897760Antiviral activity against HIV-1 Ba-L infected in human HOS-CD4-CCR5 cells measured after 48 hrs incubation by ELISA based p24 antigen assay2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID395489Apparent permeability from basolateral to apical side of human Caco-2 cells2009Bioorganic & medicinal chemistry letters, Mar-01, Volume: 19, Issue:5
1-Amido-1-phenyl-3-piperidinylbutanes--CCR5 antagonists for the treatment of HIV: part 2.
AID249028Inhibitory concentration against human immunodeficiency virus isolates utilizing C-C chemokine receptor type 5 for cell entry2005Journal of medicinal chemistry, Mar-10, Volume: 48, Issue:5
New approaches toward anti-HIV chemotherapy.
AID698150Antiviral activity against HIV1 clade F (BZ162) infected in human PBMCs assessed as inhibition of p24 antigen production by ELISA2012Journal 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.
AID1654465Drug metabolism in human liver microsomes assessed as CYP3A5*1/*1-mediated formation of (1R,2R)-2-OH-maraviroc at 8 uM incubated for 15 mins in presence of ketoconazole by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID499096Antiviral activity against HIV1 Ba-L in human PBMC assessed as p24 antigen level by ELISA2010Antimicrobial agents and chemotherapy, Jan, Volume: 54, Issue:1
Long-lasting enfuvirtide carrier pentasaccharide conjugates with potent anti-human immunodeficiency virus type 1 activity.
AID1654464Drug metabolism in CYP3cide-treated human liver microsomes assessed as formation of (1R,2R)-2-OH-maraviroc at 8 uM incubated for 15 mins by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID554145Absorption in rat at 1 mg/kg, iv and 10 mg/kg, po2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID554149Volume of distribution in dog at 0.5 mg/kg, iv and 1 mg/kg, po2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID698310Antiviral activity against HIV1 clade D (UG270) infected in human PBMCs assessed as inhibition of p24 antigen production by ELISA2012Journal 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.
AID1897769Antiviral activity against HIV-1 KIZ001 clinical isolate infected in human PBMC measured after 48 hrs incubation by luciferase based analysis2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID698309Antiviral activity against HIV1 clade C (DJ259) infected in human PBMCs assessed as inhibition of p24 antigen production by ELISA2012Journal 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.
AID1654471Drug metabolism in human liver microsomes assessed as CYP3A5*1/*1-mediated formation of (1S,3S)-3-OH-maraviroc at 8 uM incubated for 15 mins in presence of ketoconazole by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1065005Antagonist activity against CCR5 receptor in human TZM-bl cells assessed as inhibition of HIV-1 92RW-induced cell-cell fusion between viral envelope protein expressing human HEK293 cells to TZM-bl cells after 48 hrs by luciferase reporter gene assay2014ACS medicinal chemistry letters, Feb-13, Volume: 5, Issue:2
Preparation and activities of macromolecule conjugates of the CCR5 antagonist Maraviroc.
AID1370975Inhibition of CCR5 in human TZM-bl cells infected with HIV1 Bal R5 assessed as antiviral activity by measuring reduction in viral infection pre-incubated with cells followed by viral infection measured after 3 days by luciferase reporter gene assay2017Bioorganic & medicinal chemistry, 02-01, Volume: 25, Issue:3
Design, synthesis and biological evaluation of (E)-3,4-dihydroxystyryl 4-acylaminophenethyl sulfone, sulfoxide derivatives as dual inhibitors of HIV-1 CCR5 and integrase.
AID554151Oral bioavailability in dog at 1 mg/kg2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID1654480Drug metabolism in CYP3cide-treated human liver microsomes assessed as CYP3A5*3/*3-mediated formation of (1S,3R)-3-OH-MVC-maraviroc at 8 uM incubated for 15 mins by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID707884Antiviral activity against CCR5-tropic HIV12012Journal of medicinal chemistry, Nov-26, Volume: 55, Issue:22
Chemokine receptor antagonists.
AID554146Plasma protein binding in dog at 0.5 mg/kg, iv and 1 mg/kg, po2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID449003Antiviral activity against HIV1 Ba-L infected in HOS cells assessed as inhibition of viral infection2009Bioorganic & medicinal chemistry letters, Sep-01, Volume: 19, Issue:17
4,4-Disubstituted cyclohexylamine based CCR5 chemokine receptor antagonists as anti-HIV-1 agents.
AID554055Tmax in dog at 0.5 mg/kg, po2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID1897768Therapeutic index, ratio of CC50 for human PBMC to EC50 for antiviral activity against HIV-1 KM018 clinical isolate2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID1654473Drug metabolism in human liver microsomes assessed as CYP3A5*3/*3-mediated formation of (1S,3S)-3-OH-maraviroc at 8 uM incubated for 15 mins in presence of ketoconazole by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID710735Inhibition of CXCL-12'AF647 binding to CXCR4 in human SUPT1 cells incubated for 15 mins by FACS2012Journal of medicinal chemistry, Dec-13, Volume: 55, Issue:23
HIV-1 X4 activities of polycationic "viologen" based dendrimers by interaction with the chemokine receptor CXCR4: study of structure-activity relationship.
AID1808908Antagonist activity at human CCR5 receptor expressed in HEK293 cells co-expressing Galpha15 assessed as inhibition of RNATES-induced intracellular calcium flux measured after 10 mins by FLIPR calcium mobilization assay2021Journal of medicinal chemistry, 08-12, Volume: 64, Issue:15
A "Two-Birds-One-Stone" Approach toward the Design of Bifunctional Human Immunodeficiency Virus Type 1 Entry Inhibitors Targeting the CCR5 Coreceptor and gp41 N-Terminal Heptad Repeat Region.
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.
AID395485Metabolic stability in human liver microsomes assessed as clearance measured per mg of protein2009Bioorganic & medicinal chemistry letters, Mar-01, Volume: 19, Issue:5
1-Amido-1-phenyl-3-piperidinylbutanes--CCR5 antagonists for the treatment of HIV: part 2.
AID1897752Antiviral activity against HIV-1 Ba-L infected in human TZM-bl cells measured after 48 hrs incubation by luciferase based analysis2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID1897804Inhibition of CYP3A4 (unknown origin) using testosterone as substrate2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID1242368Inhibition of CXCR4 in human Jurkat cells assessed as reduction in HIV-Nef-M1-induced mitochondrial membrane depolarization at 0.01 to 100 uM by JC1 dye based fluorescence depolarization assay2015ACS medicinal chemistry letters, Jul-09, Volume: 6, Issue:7
Pyrazolo-Piperidines Exhibit Dual Inhibition of CCR5/CXCR4 HIV Entry and Reverse Transcriptase.
AID1207499Inhibition 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
AID625293Drug Induced Liver Injury Prediction System (DILIps) validation dataset; compound DILI positive/negative as observed in LTKB-BD2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID554147Clearance in dog at 0.5 mg/kg, iv and 1 mg/kg, po2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID1897756Cytotoxicity against human HOS-CD4-CCR5 cells incubated for 48 hrs by MTT assay2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID250714Mean decrease in viral load in humans after administration of 25 mg once a day; expressed as log 10 reduction2005Journal of medicinal chemistry, Mar-10, Volume: 48, Issue:5
New approaches toward anti-HIV chemotherapy.
AID1121816Antagonist activity at CCR5 in Gqi5 transfected human MOLT4 cells assessed as inhibition of RANTES-stimulated Ca2+ influx preincubated for 15 mins followed by DAMGO challenge2013MedChemComm, May-01, Volume: 4, Issue:5
A Bivalent Ligand Targeting the Putative Mu Opioid Receptor and Chemokine Receptor CCR5 Heterodimers: Binding Affinity versus Functional Activities.
AID1897766Antiviral activity against HIV-1 IIIB infected in human TZM-bl cells measured after 48 hrs incubation by luciferase based analysis2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID1897801Inhibition of CYP2C19 (unknown origin)2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID707908Lipophilicity, log P of the compound2012Journal of medicinal chemistry, Nov-26, Volume: 55, Issue:22
Chemokine receptor antagonists.
AID479402Binding affinity at CCR5 receptor by radiolabeled RANTES binding assay2010Bioorganic & medicinal chemistry letters, May-15, Volume: 20, Issue:10
Novel hexahydropyrrolo[3,4-c]pyrrole CCR5 antagonists.
AID395484Lipophilicity, log D at pH 7.42009Bioorganic & medicinal chemistry letters, Mar-01, Volume: 19, Issue:5
1-Amido-1-phenyl-3-piperidinylbutanes--CCR5 antagonists for the treatment of HIV: part 2.
AID554136Apparent permeability from basolateral to apical side of human Caco2 cells2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID1654468Drug metabolism in CYP3cide-treated human liver microsomes assessed as CYP3A5*3/*3-mediated formation of (1R,2R)-2-OH-maraviroc at 8 uM incubated for 15 mins by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1207405Inhibition 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
AID1654461Drug metabolism in CYP3cide-treated human liver microsomes assessed as CYP3A5*3/*3-mediated formation of (1S,2S)-2-OH-maraviroc at 8 uM incubated for 15 mins by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1121818Displacement of [125I]-MIP-1alpha from CCR5 in rhesus monkey Chem-1 cell membranes after 120 mins by liquid scintillation counting analysis2013MedChemComm, May-01, Volume: 4, Issue:5
A Bivalent Ligand Targeting the Putative Mu Opioid Receptor and Chemokine Receptor CCR5 Heterodimers: Binding Affinity versus Functional Activities.
AID554150Half life in dog at 0.5 mg/kg, iv and 1 mg/kg, po2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID1654479Drug metabolism in human liver microsomes assessed as CYP3A5*3/*3-mediated formation of (1S,3R)-3-OH-MVC-maraviroc at 8 uM incubated for 15 mins in presence of ketoconazole by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1654490AUC(0 to infinity) in human carrying CYP3A5*1/*1 allele at 300 mg, po measured up to 32 hrs2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID499100Antiviral activity against HIV1 US2 clade B, R5 in human PBMC assessed as p24 antigen level by ELISA2010Antimicrobial agents and chemotherapy, Jan, Volume: 54, Issue:1
Long-lasting enfuvirtide carrier pentasaccharide conjugates with potent anti-human immunodeficiency virus type 1 activity.
AID397746Lipophilicity, log D of the compound2009Journal of medicinal chemistry, Jul-23, Volume: 52, Issue:14
Side chain flexibilities in the human ether-a-go-go related gene potassium channel (hERG) together with matched-pair binding studies suggest a new binding mode for channel blockers.
AID1654466Drug metabolism in CYP3cide-treated human liver microsomes assessed as CYP3A5*1/*1-mediated formation of (1R,2R)-2-OH-maraviroc at 8 uM incubated for 15 mins by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1897785Cmax in Sprague-Dawley rat at 10 mg/kg, po measured upto 24 hrs2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID395491Volume of distribution in iv dosed Sprague-Dawley rat2009Bioorganic & medicinal chemistry letters, Mar-01, Volume: 19, Issue:5
1-Amido-1-phenyl-3-piperidinylbutanes--CCR5 antagonists for the treatment of HIV: part 2.
AID1897799Inhibition of CYP1A2 (unknown origin)2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID1897753Cytotoxicity against human TZM-bl cells incubated for 48 hrs by MTT assay2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID579722Oral bioavailability in Sprague-Dawley rat at 10 mg/kg2011Bioorganic & medicinal chemistry letters, Mar-01, Volume: 21, Issue:5
Synthesis and evaluation of 2-phenyl-1,4-butanediamine-based CCR5 antagonists for the treatment of HIV-1.
AID698151Antiviral activity against HIV1 clade B (BaL) infected in human PBMCs assessed as inhibition of p24 antigen production by ELISA2012Journal 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.
AID554140Plasma protein binding in rat at 1 mg/kg, iv and 10 mg/kg, po2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID395490Clearance in iv dosed Sprague-Dawley rat2009Bioorganic & medicinal chemistry letters, Mar-01, Volume: 19, Issue:5
1-Amido-1-phenyl-3-piperidinylbutanes--CCR5 antagonists for the treatment of HIV: part 2.
AID393052Lipophilicity, log D at pH 7.42009Bioorganic & medicinal chemistry letters, Feb-15, Volume: 19, Issue:4
1-Amido-1-phenyl-3-piperidinylbutanes - CCR5 antagonists for the treatment of HIV. Part 1.
AID1897800Inhibition of CYP2C9 (unknown origin)2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID554056fCmax in dog at 0.5 mg/kg, po2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID1062970Inhibition of CCR5 (unknown origin) expressed in CHO cells assessed as inhibition of RANTES-induced intracellular Ca2+ mobilization after 10 mins by Fluo-4 AM staining-based fluorescence assay2014European journal of medicinal chemistry, Jan, Volume: 71Design, synthesis, and biological evaluation of novel piperidine-4-carboxamide derivatives as potent CCR5 inhibitors.
AID1654484Drug metabolism in CYP3cide-treated human liver microsomes assessed as CYP3A5*1/*1-mediated formation of hydroxymethyl-maraviroc at 8 uM incubated for 15 mins by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1062969Antiviral activity against HIV-1 infected in human SupT1 cells assessed as inhibition of viral infectivity after 48 hrs by luciferase reporter gene assay2014European journal of medicinal chemistry, Jan, Volume: 71Design, synthesis, and biological evaluation of novel piperidine-4-carboxamide derivatives as potent CCR5 inhibitors.
AID710729Antagonist activity against CXCR5 expressed in U87.CD.CXCR5 cells assessed as inhibition of CCL3L1-induced calcium signaling incubated for 10 mins by FLIPR2012Journal of medicinal chemistry, Dec-13, Volume: 55, Issue:23
HIV-1 X4 activities of polycationic "viologen" based dendrimers by interaction with the chemokine receptor CXCR4: study of structure-activity relationship.
AID554162Intrinsic clearance in rat liver microsomes assessed per mg of protein2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID1654462Drug metabolism in human liver microsomes assessed as CYP3A5*3/*3-mediated formation of (1S,2S)-2-OH-maraviroc at 8 uM incubated for 15 mins in presence of ketoconazole by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1897755Cytotoxicity against human PM1 cells incubated for 48 hrs by MTT assay2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID499102Antiviral activity against HIV1 DJ259 in human PBMC assessed as p24 antigen level by ELISA2010Antimicrobial agents and chemotherapy, Jan, Volume: 54, Issue:1
Long-lasting enfuvirtide carrier pentasaccharide conjugates with potent anti-human immunodeficiency virus type 1 activity.
AID1473740Inhibition of human MRP3 overexpressed in Sf9 insect cell membrane vesicles assessed as uptake of [3H]-estradiol-17beta-D-glucuronide in presence of ATP and GSH measured after 10 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID395492Half life in iv dosed Sprague-Dawley rat2009Bioorganic & medicinal chemistry letters, Mar-01, Volume: 19, Issue:5
1-Amido-1-phenyl-3-piperidinylbutanes--CCR5 antagonists for the treatment of HIV: part 2.
AID1897798Clearance in Sprague-Dawley rat at 1 mg/kg, iv measured upto 24 hrs2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID1897770Therapeutic index, ratio of CC50 for human PBMC to EC50 for antiviral activity against HIV-1 KIZ001 clinical isolate2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID1370972Cytotoxicity against human TZM-bl cells assessed as reduction in cell viability after 2 days by CCK-8 assay2017Bioorganic & medicinal chemistry, 02-01, Volume: 25, Issue:3
Design, synthesis and biological evaluation of (E)-3,4-dihydroxystyryl 4-acylaminophenethyl sulfone, sulfoxide derivatives as dual inhibitors of HIV-1 CCR5 and integrase.
AID320901Antiviral activity against HIV1 JRCSF by viral entry assay2008Bioorganic & medicinal chemistry letters, Feb-15, Volume: 18, Issue:4
Anti-HIV-1 entry optimization of novel imidazopiperidine-tropane CCR5 antagonists.
AID1654460Drug metabolism in CYP3cide-treated human liver microsomes assessed as CYP3A5*1/*1-mediated formation of (1S,2S)-2-OH-maraviroc at 8 uM incubated for 15 mins by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID721754Inhibition of human MATE1-mediated ASP+ uptake expressed in HEK293 cells after 1.5 mins by fluorescence assay2013Journal of medicinal chemistry, Feb-14, Volume: 56, Issue:3
Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling.
AID499098Antiviral activity against HIV1 strain I-2496 in human PBMC assessed as p24 antigen level by ELISA2010Antimicrobial agents and chemotherapy, Jan, Volume: 54, Issue:1
Long-lasting enfuvirtide carrier pentasaccharide conjugates with potent anti-human immunodeficiency virus type 1 activity.
AID554134Displacement of [3H]-dofetilide from human ERG expressed in HEK293 cells at 300 nM2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID554148Unbound clearance in dog at 0.5 mg/kg, iv and 1 mg/kg, po2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID1654477Drug metabolism in human liver microsomes assessed as CYP3A5*1/*1-mediated formation of (1S,3R)-3-OH-MVC-maraviroc at 8 uM incubated for 15 mins in presence of ketoconazole by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1654467Drug metabolism in human liver microsomes assessed as CYP3A5*3/*3-mediated formation of (1R,2R)-2-OH-maraviroc at 8 uM incubated for 15 mins in presence of ketoconazole by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1473739Inhibition of human MRP2 overexpressed in Sf9 cell membrane vesicles assessed as uptake of [3H]-estradiol-17beta-D-glucuronide in presence of ATP and GSH measured after 20 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1654476Drug metabolism in CYP3cide-treated human liver microsomes assessed as formation of (1S,3R)-3-OH-MVC-maraviroc at 8 uM incubated for 15 mins by LC-MS/MS method2020Journal of medicinal chemistry, 06-25, Volume: 63, Issue:12
Effective Application of Metabolite Profiling in Drug Design and Discovery.
AID1897754Therapeutic index, ratio of CC50 for human TZM-bl cells to EC50 for antiviral activity against HIV-1 Ba-L2022Journal of medicinal chemistry, 12-22, Volume: 65, Issue:24
Structure-Based Design of Tropane Derivatives as a Novel Series of CCR5 Antagonists with Broad-Spectrum Anti-HIV-1 Activities and Improved Oral Bioavailability.
AID554135Apparent permeability from apical to basolateral side of human Caco2 cells2011Journal of medicinal chemistry, Jan-13, Volume: 54, Issue:1
An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]propyl}acetamide (PF-2
AID1065007Antagonist activity against CCR5 receptor in human TZM-bl cells assessed as inhibition of HIV-1 YU2-induced cell-cell fusion between viral envelope protein expressing human HEK293 cells to TZM-bl cells after 48 hrs by luciferase reporter gene assay2014ACS medicinal chemistry letters, Feb-13, Volume: 5, Issue:2
Preparation and activities of macromolecule conjugates of the CCR5 antagonist Maraviroc.
AID1065004Antagonist activity against CCR5 receptor in human TZM-bl cells assessed as inhibition of HIV-1 MGC26-induced cell-cell fusion between viral envelope protein expressing human HEK293 cells to TZM-bl cells after 48 hrs by luciferase reporter gene assay2014ACS medicinal chemistry letters, Feb-13, Volume: 5, Issue:2
Preparation and activities of macromolecule conjugates of the CCR5 antagonist Maraviroc.
AID1121815Binding affinity to CCR5/MOR in human astrocytes assessed as inhibition of R5 HIV-1 SF162 infection by measuring Tat protein expression at 100 nM preincubated for 30 to 60 mins followed by viral infection measured after 18 hrs by luciferase reporter gene 2013MedChemComm, May-01, Volume: 4, Issue:5
A Bivalent Ligand Targeting the Putative Mu Opioid Receptor and Chemokine Receptor CCR5 Heterodimers: Binding Affinity versus Functional Activities.
AID1347105qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for MG 63 (6-TG R) cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347128qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for OHS-50 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1346987P-glycoprotein substrates identified in KB-8-5-11 adenocarcinoma cell line, qHTS therapeutic library screen2019Molecular pharmacology, 11, Volume: 96, Issue:5
A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
AID1347114qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for DAOY cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347136qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Orthogonal 3D viability screen for SJ-GBM2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347141qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Orthogonal 3D viability screen for RD cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347112qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for BT-12 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347093qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for SK-N-MC cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347097qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Saos-2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347095qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for NB-EBc1 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347137qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Orthogonal 3D viability screen for Daoy cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1296008Cytotoxic Profiling of Annotated Libraries Using Quantitative High-Throughput Screening2020SLAS discovery : advancing life sciences R & D, 01, Volume: 25, Issue:1
Cytotoxic Profiling of Annotated and Diverse Chemical Libraries Using Quantitative High-Throughput Screening.
AID1347125qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for Rh18 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347103qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for OHS-50 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347119qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for MG 63 (6-TG R) cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347101qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for BT-12 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347092qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for A673 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347102qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Rh18 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347083qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lassa (LASV) Arenavirus: Viability assay - alamar blue signal for LASV Primary Screen2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID1347140qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Orthogonal 3D viability screen for MG 63 (6-TG R) cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347107qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Rh30 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347109qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for NB1643 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347099qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for NB1643 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1508630Primary qHTS for small molecule stabilizers of the endoplasmic reticulum resident proteome: Secreted ER Calcium Modulated Protein (SERCaMP) assay2021Cell reports, 04-27, Volume: 35, Issue:4
A target-agnostic screen identifies approved drugs to stabilize the endoplasmic reticulum-resident proteome.
AID1347138qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Orthogonal 3D caspase screen for TC32 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347126qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for Rh30 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347124qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for RD cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347096qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for U-2 OS cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID651635Viability Counterscreen for Primary qHTS for Inhibitors of ATXN expression
AID1347090qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for DAOY cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347082qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lassa (LASV) Arenavirus: LASV Primary Screen - GLuc reporter signal2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID1347116qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for SJ-GBM2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347127qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for Saos-2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347115qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for NB-EBc1 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347094qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for BT-37 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347123qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for Rh41 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347108qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Rh41 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347135qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Orthogonal 3D viability screen for SK-N-SH cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347117qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for BT-37 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347086qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lymphocytic Choriomeningitis Arenaviruses (LCMV): LCMV Primary Screen - GLuc reporter signal2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID1347091qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for SJ-GBM2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347100qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for LAN-5 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347089qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for TC32 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347098qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for SK-N-SH cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347129qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for SK-N-SH cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1346986P-glycoprotein substrates identified in KB-3-1 adenocarcinoma cell line, qHTS therapeutic library screen2019Molecular pharmacology, 11, Volume: 96, Issue:5
A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
AID1347122qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for U-2 OS cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347139qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Orthogonal 3D viability screen for TC32 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347113qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for LAN-5 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1745845Primary qHTS for Inhibitors of ATXN expression
AID1347106qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for control Hh wild type fibroblast cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347104qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for RD cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347111qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for SK-N-MC cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347118qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for TC32 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347110qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for A673 cells)2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347121qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for control Hh wild type fibroblast cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347154Primary screen GU AMC qHTS for Zika virus inhibitors2020Proceedings of the National Academy of Sciences of the United States of America, 12-08, Volume: 117, Issue:49
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors.
AID1820207Cytotoxicity against human GHOST CCR5 cells assessed as reduction in cell viability measured after 6 days by XTT assay2021Journal of medicinal chemistry, 06-10, Volume: 64, Issue:11
Structure-Based Design and Development of Chemical Probes Targeting Putative MOR-CCR5 Heterodimers to Inhibit Opioid Exacerbated HIV-1 Infectivity.
AID1820204Antagonist activity at CCR5 (unknown origin) expressed in HOS cells co-expressing Gqi-5 assessed as inhibition CCL5-stimulated Ca2+ mobilization by calcium mobilization assay2021Journal of medicinal chemistry, 06-10, Volume: 64, Issue:11
Structure-Based Design and Development of Chemical Probes Targeting Putative MOR-CCR5 Heterodimers to Inhibit Opioid Exacerbated HIV-1 Infectivity.
AID1820208Therapeutic index, ratio of TC50 for human GHOST CCR5 cells to EC50 for inhibition of CCR5-mediated HIV-1 Bal entry in human GHOST CCR5 cells2021Journal of medicinal chemistry, 06-10, Volume: 64, Issue:11
Structure-Based Design and Development of Chemical Probes Targeting Putative MOR-CCR5 Heterodimers to Inhibit Opioid Exacerbated HIV-1 Infectivity.
AID1820206Inhibition of CCR5-mediated HIV-1 Bal entry in human GHOST CCR5 cells assessed as decrease in viral reverse transcriptase activity by measuring [3H]thymidine triphosphate incorporation measured after 60 to 90 mins by scintillation counting analysis2021Journal of medicinal chemistry, 06-10, Volume: 64, Issue:11
Structure-Based Design and Development of Chemical Probes Targeting Putative MOR-CCR5 Heterodimers to Inhibit Opioid Exacerbated HIV-1 Infectivity.
AID1346815Human CCR5 (Chemokine receptors)2005Biochemical pharmacology, Dec-19, Volume: 71, Issue:1-2
Molecular cloning and radioligand binding characterization of the chemokine receptor CCR5 from rhesus macaque and human.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (742)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's0 (0.00)18.2507
2000's141 (19.00)29.6817
2010's515 (69.41)24.3611
2020's86 (11.59)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 65.61

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

MetricThis Compound (vs All)
Research Demand Index65.61 (24.57)
Research Supply Index6.81 (2.92)
Research Growth Index4.84 (4.65)
Search Engine Demand Index113.25 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (65.61)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials124 (15.80%)5.53%
Reviews93 (11.85%)6.00%
Case Studies30 (3.82%)4.05%
Observational9 (1.15%)0.25%
Other529 (67.39%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (139)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
Effect of Maraviroc on Metabolic Function in Obese Subjects (Phase I) [NCT01133210]Phase 127 participants (Actual)Interventional2011-01-31Completed
Proof-of-concept Trial to Evaluate the Safety and Efficacy of Maraviroc in Severe Acute Respiratory Syndrome (SARS) Coronavirus-2 (CoV-2) Infected Patients Hospitalized for COVID-19 [NCT04710199]Phase 244 participants (Actual)Interventional2021-02-23Completed
A Prospective, Randomised Study to Assess Safety, Changes in Platelet Reactivity, Plasma Cardiac Biomarkers, Immunological and Metabolic Parameters in HIV-1 Infected Subjects Undergoing a Switch in Antiretroviral Therapy [NCT00981773]Phase 418 participants (Actual)Interventional2009-09-30Terminated(stopped due to Recruitment problem)
A Multicenter, Randomized, Double-Blind, Comparative Trial Of Maraviroc + Darunavir/Ritonavir Versus Emtricitabine/Tenofovir + Darunavir/Ritonavir For The Treatment Of Antiretroviral-Naive Hiv-Infected Patients With Ccr5-Tropic Hiv-1 [NCT01345630]Phase 3813 participants (Actual)Interventional2011-09-30Terminated(stopped due to See termination reason in detailed description.)
Effect of Maraviroc (MCV) on the Immunological Recovery of HIV-1 Discordant Patients With CD4 Lymphocyte Counts Below 200 Cells/mm3 [NCT01235013]Phase 460 participants (Anticipated)InterventionalNot yet recruiting
Incidence And Risk Factors For Acquired Immune Deficiency Syndrome (AIDS) Defining And Non-AIDS Defining Malignancies, And Other AIDS Defining Illnesses In The EuroSIDA Study [NCT01329783]1,181 participants (Actual)Observational2007-04-30Completed
The Effects of Maraviroc Versus Efavirenz in Combination With Zidovudine/Abacavir on the CD4/CD8 Ratio in Treatment-naïve HIV-infected Individuals [NCT03178084]Phase 3721 participants (Actual)Interventional2014-10-15Completed
A Phase I/II Trial of Allogeneic Reduced-Intensity, HLA-Haploidentical Bone Marrow Transplantation Followed by GVHD Prophylaxis With Cyclophosphamide, Bortezomib and Maraviroc for Hematologic Malignancies in People Living With HIV (PLWH) [NCT05470491]Phase 1/Phase 2265 participants (Anticipated)Interventional2023-01-26Suspended(stopped due to On administrative hold for interim data analysis.)
Phase 1 Safety and Pharmacokinetics of Dapivirine/Maraviroc Vaginal Ring [NCT01363037]Phase 148 participants (Actual)Interventional2011-11-30Completed
A Multicenter, Randomized, Double-Blind Placebo-Controlled Trial of a Novel CCR5 Antagonist, UK-427,857, in Combination With Optimized Background Therapy Versus Optimized Background Therapy Alone for the Treatment of Antiretroviral-Experienced, Non CCR5-T [NCT00098748]Phase 2/Phase 3190 participants (Actual)Interventional2004-11-30Completed
A Multicenter, Randomized, Double-Blind, Placebo-Controlled Trial of a Novel CCR5 Antagonist, UK-427,857, in Combination With Optimized Background Therapy Versus Optimized Background Therapy Alone for the Treatment of Antiretroviral-Experienced HIV-1 Infe [NCT00098306]Phase 2/Phase 3601 participants (Actual)Interventional2004-11-30Completed
Effect of Cytoreductive Chemotherapy and a CCR5 Coreceptor Antagonist on HIV-1 Eradication [NCT02486510]Early Phase 17 participants (Actual)Interventional2012-07-31Terminated(stopped due to Futility criteria)
A Phase 1, Open-label, Drug-drug Interaction Study Between BMS-663068 and Maraviroc in Healthy Subjects [NCT02480894]Phase 1112 participants (Actual)Interventional2015-07-07Completed
Safety and Efficacy of Maraviroc, a CCR5-inhibitor in Prophylaxis of Graft-Versus-Host Disease in Patients Undergoing Non-Myeloablative Allogeneic Stem-Cell Transplantation [NCT00948753]Phase 1/Phase 238 participants (Actual)Interventional2009-06-30Completed
Safety of Maraviroc for Post-stroke Depression (Proof-of-concept) [NCT05932550]Phase 210 participants (Actual)Interventional2020-08-04Completed
Pilot Study Evaluating Maraviroc (Celsentri®)Intensification Benefit in HIV Infected Patients Presenting Insufficient Immune Restoration Despite Controlled Viral Load With Antiretroviral Treatment. ANRS 145 MARIMUNO [NCT00944541]Phase 260 participants (Actual)Interventional2009-09-30Completed
Open-Label Study of Maraviroc in Hospitalized Individuals Diagnosed With SARS-CoV-2 [NCT04435522]Phase 19 participants (Actual)Interventional2020-10-01Completed
DRUG USE INVESTIGATION FOR HIV INFECTION PATIENTS OF MARAVIROC (REGULATORY POST MARKETING COMMITMENT PLAN). [NCT00864474]68 participants (Actual)Observational2010-03-31Completed
Open-Label, Fixed-Sequence, Crossover Study To Estimate The Pharmacokinetic Interaction Between Multiple Dose Maraviroc And Fosamprenavir/Ritonavir In Healthy Subjects [NCT01140412]Phase 114 participants (Actual)Interventional2010-07-31Terminated(stopped due to The trial prematurely terminated on November 1, 2010, due to healthy volunteer participants experiencing non-serious fosamprenavir-related skin rash.)
Non-Interventional Study Of Celsentri® In Treatment Experienced Patients Infected With CCR5 Tropic HIV-1 [NCT00850395]79 participants (Actual)Observational2009-07-31Completed
Pharmacokinetic Properties of Antiretroviral and Related Drugs During Pregnancy and Postpartum [NCT00042289]1,578 participants (Actual)Observational2003-06-09Completed
A Phase IV, Open Label Study in Healthy Male Subjects to Investigate the Extent of Maraviroc Exposure in Blood, Saliva, Seminal Fluid, and Rectal Mucosal Tissue Following Single and Multiple Dosing of Maraviroc [NCT00775294]Phase 114 participants (Actual)Interventional2008-05-31Completed
Maraviroc Switch Vascular Endothelium (VE) Substudy: a Substudy of MARCH [NCT01585753]34 participants (Actual)Observational2012-06-30Completed
Switch to Darunavir/r + Maraviroc QD in Patients With R5 Tropism by Viral DNA Genotyping With Suppressed Viremia (GUSTA): a Multicenter, Open-label, Randomized Controlled Trial [NCT01367210]Phase 4165 participants (Actual)Interventional2011-06-30Terminated(stopped due to Because a higher rate of virological failures in study versus control arm.)
Open-Label, Fixed-Sequence Study To Estimate The Pharmacokinetic Interaction Between Multiple Dose Maraviroc And Fosamprenavir/Ritonavir In Healthy Subjects [NCT01290211]Phase 128 participants (Actual)Interventional2011-04-30Completed
Phase II Pilot Study of Simplification to Maraviroc - Raltegravir Dual Therapy After 6 Months of Maraviroc - Raltegravir - Tenofovir - Emtricitabine Quadruple Therapy in ARV Treatment-naive, HIV-1-infected Patients With CCR5- Virus [NCT01291459]Phase 240 participants (Anticipated)Interventional2011-09-30Active, not recruiting
A Pilot Evaluation of the Pharmacokinetics, Efficacy and Safety of Switching From Efavirenz to Maraviroc Administered at 600mg Then 300mg Twice-daily in Patients Suppressed on an Efavirenz-containing Regimen as Initial Therapy [NCT01190293]Phase 412 participants (Actual)Interventional2010-01-31Completed
Pharmacokinetics of Maraviroc and Boosted Atazanavir Dual Regimen in Stable HIV-infected Patients [NCT03708861]Phase 30 participants (Actual)Interventional2016-01-31Withdrawn
Phase I Safety and Pharmacokinetic Study of Maraviroc in HIV-1-Exposed Infants at Risk of Acquiring HIV-1 Infection [NCT02778204]Phase 147 participants (Actual)Interventional2017-06-05Completed
Maraviroc to Augment Rehabilitation Outcomes After Stroke [NCT03172026]Phase 2/Phase 32 participants (Actual)Interventional2019-08-01Terminated(stopped due to Poor recruitment for each site, partly related to Covid epidemic.)
Optimized Phase III Trial of Immuno-stimulation With Maraviroc, a CCR5 (Chemokine Receptor 5) Antagonist, Combined With Anti Retroviral Therapy in Advanced, Late Diagnosed HIV-1 Infected Patients With an AIDS-defining Event and/or CD4 (Cluster of Differen [NCT01348308]Phase 3407 participants (Actual)Interventional2011-09-30Completed
A Pilot Trial of Maraviroc for Treatment of Subjects on Antiretroviral Therapy With Suboptimal CD4 T-cell Count Recovery Despite Sustained Virologic Suppression [NCT00709111]34 participants (Actual)Interventional2009-01-31Completed
A Placebo Controlled Study of the Impact on Insulin Sensitivity and Lipid Profile of Maraviroc 300 mg Twice Daily in HIV Negative Male Volunteers [NCT00771823]Phase 116 participants (Actual)Interventional2008-05-31Completed
Safety and Efficacy of Maraviroc-Based Graft-Versus-Host-Disease Prophylaxis in HLA-Unrelated and HLA-Mismatched Related Donor Transplantation [NCT02799888]Phase 240 participants (Anticipated)Interventional2014-04-30Recruiting
Maraviroc and NeuroAIDS Pathogenesis [NCT02159027]Phase 2/Phase 348 participants (Actual)Interventional2015-06-30Completed
A Randomized Study to Compare the Efficacy of Vorinostat/Hydroxychloroquine/Maraviroc (VHM) in Controlling HIV After Treatment Interruption in Subjects Who Initiated ART During Acute HIV Infection [NCT02475915]Phase 1/Phase 215 participants (Actual)Interventional2015-01-31Completed
An Interventional Study of the Effect of CCR5 Inhibition With Maraviroc on Immune Cells in the the Lung and in Peripheral Blood of Patients With Sarcoidosis [NCT02134717]3 participants (Actual)Interventional2014-01-31Terminated(stopped due to Poor recruitment)
The Role of Home Packs of HIV Post-Exposure Prophylaxis for Sexual Exposure (PEPSE) to Improve the Speed and Appropriate Uptake of PEPSE in High Risk Individuals [NCT04965662]Phase 4139 participants (Actual)Interventional2018-01-01Completed
Pilot Assessment of Lopinavir/Ritonavir and Maraviroc in Experienced Patients [NCT00981318]Phase 43 participants (Actual)Interventional2009-12-31Terminated(stopped due to unable to enroll expected number of subjects)
A Multicenter, Randomized, Double-Blind, Comparative Trial Of A Novel CCR5 Antagonist, UK-427,857, In Combination With Zidovudine/Lamivudine Versus Efavirenz In Combination With Zidovudine/Lamivudine For The Treatment Of Antiretroviral-Naive HIV-1 Infecte [NCT00098293]Phase 3916 participants (Actual)Interventional2004-11-30Completed
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 Local, Multicentre, Open Label Access Program To Provide Maraviroc To Eligible Adult Patients Completing A4001050 Study Until Commercial Availability Of Maraviroc (Celsentri) In India [NCT00992654]0 participants Expanded AccessNo longer available
AN INTERNATIONAL, MULTICENTER, PROSPECTIVE OBSERVATIONAL STUDY OF THE SAFETY OF MARAVIROC USED WITH OPTIMIZED BACKGROUND THERAPY IN TREATMENT-EXPERIENCED HIV-1 INFECTED PATIENTS [NCT00665561]2,500 participants (Actual)Observational2008-03-31Completed
A Compassionate Access Protocol For Those Patients Who Have Completed A4001029 [NCT00801515]0 participants Expanded Access2009-08-31No longer available
SPRING: Safety, Efficacy, Pharmacokinetics of tipRanavi/r IN Race/Gender HIV+ Patients Randomized to Therapeutic Drug Monitoring or Standard of Care [NCT00440271]Phase 333 participants (Actual)Interventional2007-02-28Terminated
Open-Label, Fixed Sequence, Crossover Study To Estimate The Effect Of Multiple Dose Maraviroc On Single Dose Digoxin Pharmacokinetics In Healthy Subjects [NCT01056874]Phase 112 participants (Actual)Interventional2010-03-31Completed
Estudio Comparativo de Dos métodos Para Predecir el Uso de Co-receptores Por el Virus de la Inmunodeficiencia 1 (HIV-1): el Ensayo fenotípico (Trofile ESTA®) y la Respuesta virológica a Corto Plazo a un Antagonista de CCR5 [NCT01060618]Phase 2/Phase 358 participants (Actual)Interventional2009-05-31Completed
Surrogate Marker For Tropism-A Multi-Center, Open Label, Pilot Study [NCT00496782]Phase 116 participants (Actual)Interventional2007-07-31Terminated(stopped due to See Detailed Description)
Pilot Study to Assess the Safety and Efficacy of Switching the Nnrti or pi to Maraviroc in Hiv-1-infected Subjects With Persistent Viremia Suppression [NCT00966329]Phase 430 participants (Actual)Interventional2009-10-31Completed
A Randomised, Double Blind, Placebo-Controlled, Multicentre Study Of UK-427,857 25mg O.D. , 50mg B.I.D., 100mg B.I.D And 300mg B.I.D. In Asymptomatic HIV Infected Patients To Investigate Pharmacodynamics, Pharmacokinetics, Safety And Toleration. [NCT00643643]Phase 241 participants (Actual)Interventional2002-10-31Completed
Pilot Study Of Novel Combination Of Maraviroc + Atazanavir/Ritonavir vs. Atazanavir/Ritonavir + Emtricitabine/Tenofovir For The Treatment Of Naïve HIV-Infected Patients With R5 HIV-1 [NCT00827112]Phase 2129 participants (Actual)Interventional2009-03-31Completed
A Double-Blind (3rd Party Open), Placebo-Controlled, Crossover, Dose Escalating Study To Evaluate The Safety, Tolerability And Pharmacokinetics Of Single Oral Doses Of PF-03716539, To Assess The Potential Of PF-03716539 To Inhibit CYP3A4 (In Vivo) And To [NCT00783484]Phase 137 participants (Actual)Interventional2008-10-31Completed
A Two-Arm, Single Site, Proof of Concept Study of the Antiviral and Immunological Effects of Intensification of Suppressive Antiretroviral Therapy With Maraviroc, a CCR5 Antagonist (ADARC 2007-02) [NCT00703586]Phase 18 participants (Actual)Interventional2007-09-30Completed
AN OPEN-LABEL, MULTICENTER, MULTIPLE-DOSE PHARMACOKINETIC, SAFETY AND EFFICACY TRIAL OF MARAVIROC IN COMBINATION WITH OPTIMIZED BACKGROUND THERAPY FOR THE TREATMENT OF ANTIRETROVIRAL-EXPERIENCED CCR5-TROPIC HIV-1 INFECTED CHILDREN 2 - <18 YEARS OF AGE [NCT00791700]Phase 2103 participants (Actual)Interventional2009-04-22Active, not recruiting
An Open Label Phase 4 Study To Evaluate An Interaction Between Maraviroc And Raltegravir In Healthy Subjects [NCT00666705]Phase 418 participants (Actual)Interventional2008-02-29Completed
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
Steady State Pharmacokinetics (PK) of Fosamprenavir (FPV) Alone or in Combination With Low Dose Ritonavir (/r) (1400mg BID, 1400mg/100mg QD or 700/100mg BID) and the Chemokine Receptor 5 (CCR5) Entry Inhibitor Maraviroc (MVC) 300mg BID in Healthy Voluntee [NCT00764465]Phase 245 participants (Actual)Interventional2008-10-31Completed
CID 0708 - Sex, Aging and Antiretroviral Pharmacokinetics [NCT00666055]11 participants (Actual)Observational2008-03-31Completed
Maraviroc Immune Recovery Study, A Multicenter, Randomized, Placebo-controlled, Exploratory Mechanistic Study Into the Role of Maraviroc on Immune Recovery [NCT00875368]Phase 485 participants (Actual)Interventional2009-02-28Completed
An Open Label Single Dose Study To Investigate Safety And Pharmacokinetics Of Maraviroc Following A Single Oral Dose Of 300 Mg Maraviroc (Two 150 Mg Maraviroc Commercial Tablets) Under Fasting Conditions In Healthy Male Japanese Volunteers [NCT00821535]Phase 112 participants (Actual)Interventional2009-02-28Completed
Maraviroc Abacavir STudy - Effect on Endothelial Recovery [NCT01389063]Phase 424 participants (Anticipated)Interventional2012-01-31Recruiting
CCR5 Antagonism to Decrease the Incidence of the Immune Reconstitution Inflammatory Syndrome in HIV-Infected Patients [NCT00988780]276 participants (Anticipated)Interventional2009-12-31Active, not recruiting
Pilot Study Of The Effect Of A CCR5 Coreceptor Antagonist On The Latency And Reservoir Of HIV-1 In Patients Taking Highly Active Antiretroviral Therapy [NCT00795444]Phase 210 participants (Actual)Interventional2008-03-31Completed
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
An Investigation Into The Effects Of Food And Dose Regimen On Viral Load Response In HIV Infected Patients On Short-Term Monotherapy With UK-427,857 (Maraviroc) [NCT00634959]Phase 237 participants (Actual)Interventional2003-07-31Completed
A Phase I Trial of Combined PD-1 Inhibition (Pembrolizumab) and CCR5 Inhibition (Maraviroc) for the Treatment of Refractory Microsatellite Stable (MSS) Metastatic Colorectal Cancer (mCRC) [NCT03274804]Phase 120 participants (Actual)Interventional2018-04-01Completed
An Open-Label, Parallel Group, Single And Multiple Dose Study To Evaluate The Pharmacokinetics, Safety And Toleration Of Maraviroc Administered To Subjects With Various Degrees Of Renal Impaired And Normal Renal Function [NCT00717067]Phase 430 participants (Actual)Interventional2008-07-31Completed
Adding Maraviroc to the HAART Regimen of HIV-infected Patients Who Are Well Controlled, and the Effect on CD4 Lymphocyte Counts [NCT02934022]25 participants (Actual)Interventional2012-06-30Completed
A Multicenter, Open Label, Expanded Access Trial Of Maraviroc [NCT00426660]Phase 31,047 participants (Actual)Interventional2007-02-28Completed
Optimisation of Primary HIV1 Infection Treatment (ANRS 147 OPTIPRIM) [NCT01033760]Phase 390 participants (Actual)Interventional2010-04-30Completed
A Phase 2, Randomized, Double-Blind, Placebo-Controlled, Parallel-Group Study to Assess the Safety and Efficacy of Maraviroc in the Treatment of Rheumatoid Arthritis in Subjects Receiving Methotrexate [NCT00427934]Phase 2128 participants (Actual)Interventional2007-02-28Terminated(stopped due to See Detailed Description.)
Randomised, Placebo Controlled, Phase IV, Safety and Exploratory Immunogenicity Study on Maraviroc, an Oral ART CCR5 Inhibitor, on the Intensification of Immune Function in HIV-1 Infected Subjects Receiving Immunisation With Novel Antigens [NCT01049204]Phase 448 participants (Actual)Interventional2009-07-31Terminated(stopped due to The data from the first 48 patients recruited has now been analysed, and it has been determined that it is sufficient to meet the study objectives.)
A Multicenter, Randomized, Double-Blind, Placebo-Controlled Trial of a Novel CCR5 Antagonist, UK-427,857, in Combination With Optimized Background Therapy Versus Optimized Background Therapy Alone for the Treatment of Antiretroviral-Experienced HIV-1 Infe [NCT00098722]Phase 2/Phase 3474 participants (Actual)Interventional2004-12-31Completed
A Phase 4, Single Arm, Open Label, Pilot Study of Maraviroc (Celsentri) in Combination With Raltegravir and Darunavir/Ritonavir for the Treatment of Triple Class Failure in Adult HIV-1 Infected Patients. [NCT01013987]Phase 460 participants (Anticipated)Interventional2010-02-28Recruiting
LUMINESCENCE-001 Ipilimumab, Maraviroc and Nivolumab in Advanced Metastatic Colorectal and Pancreatic Cancer [NCT04721301]Phase 150 participants (Actual)Interventional2017-01-15Completed
Polymorphic Effects of Cytochrome P450 3A5 on Pharmacokinetics of Maraviroc and Its Metabolites [NCT01980329]Phase 124 participants (Actual)Interventional2013-01-31Completed
Randomized, Double-blinded, Controlled Trial of Intensive HAART Including Raltegravir, and Maraviroc, on HIV-1 Pro-viral DNA and Reservoir Decay in HIV-1-infected Individuals During the Acute/Early Infection [NCT01154673]Phase 2/Phase 332 participants (Actual)Interventional2011-11-30Completed
A Study in HIV+ Patients With CCR5-tropic Virus and Undetectable Viral Load on a First, Non-Selzentry®-Containing Regimen, Switching Them to Once-daily Selzentry® (600mg qd) Plus the Same 2 NRTIs Previously Administered [NCT01866267]Phase 431 participants (Actual)Interventional2013-01-31Completed
A Pilot Study to Evaluate Anti-Hepatitis C Virus Effect of Maraviroc in Patients Co-infected With Human Immunodeficiency Virus (HIV) and Hepatitis C [NCT02881762]Phase 410 participants (Actual)Interventional2017-06-01Completed
[NCT01680536]Phase 419 participants (Actual)Interventional2012-11-30Completed
A Phase 1 Pharmacokinetic Study to Assess the Steady State Pharmacokinetic Profile and Short Term Safety of Maraviroc Dosed With Darunavir/Ritonavir All Once Daily, With and Without Nucleoside Analogues, in HIV-1 Infected Subjects [NCT01348763]Phase 113 participants (Actual)Interventional2011-10-31Completed
A Multicenter, Randomized, Blinded, Placebo-controlled Study To Evaluate The Safety Of Maraviroc In Combination With Other Antiretroviral Agents In Hiv-1-infected Subjects Co-infected With Hepatitis C And/or Hepatitis B Virus [NCT01327547]Phase 4138 participants (Actual)Interventional2011-05-18Completed
Maraviroc as an Immunomodulatory Drug for Antiretroviral-treated HIV Infected Patients Exhibiting Immunologic Failure [NCT00735072]Phase 445 participants (Actual)Interventional2008-09-30Completed
A Multicenter, Open Label, Non-Comparative Safety Study Of Maraviroc [NCT00478231]Phase 3209 participants (Actual)Interventional2007-07-31Completed
Maraviroc Compassionate Use [NCT00719823]Phase 30 participants (Actual)Interventional2008-08-31Withdrawn
Efficacy of Treatment Intensification With Maraviroc on HIV-1 Viral Latency in Recently Infected Hiv-1 naïve Patients Starting Raltegravir Plus Tenofovir/Emtricitabine. [NCT00808002]Phase 330 participants (Actual)Interventional2009-02-28Completed
[NCT00844519]Phase 352 participants (Actual)Interventional2010-01-31Completed
Maraviroc Plus Darunavir/Ritonavir Study for Treatment-Naïve Patients Infected With R5-tropic HIV-1 Based on Enhanced Sensitivity Trofile [NCT00993148]Phase 225 participants (Actual)Interventional2010-05-31Completed
Randomised, Openlabel Study Evaluating Efficacy and Safety of Maraviroc as a Switch for Either NRTI or PI/r in HIV-1 Infected Individuals With Stable, Well-Controlled Plasma HIV-RNA While Taking Their First N(t)RTI + PI/r Regimen of cART [NCT01384682]Phase 4399 participants (Actual)Interventional2011-08-31Completed
CHARM-03: A Randomized, Open Label, Crossover Phase 1 Safety and Pharmacokinetic Study of Oral Maraviroc and Maraviroc 1% Gel Administered Rectally and Vaginally to HIV-1 Seronegative Adults [NCT02346084]Phase 119 participants (Actual)Interventional2015-01-31Completed
Phase II Pilot Study Evaluating the Efficacy of Dual Therapy With Raltegravir Plus Maraviroc in Patients Receiving Suppressive Antiretroviral Therapy and Presenting With Lipohypertrophy (ANRS 157 ROCnRAL). [NCT01420523]Phase 248 participants (Actual)Interventional2011-12-31Terminated
Safety Study of Maraviroc's Effect on Human Osteoclasts [NCT01428986]16 participants (Actual)Observational2009-11-30Completed
Use of Maraviroc (MVC) in Immunological Non-responder HIV-1-infected Patients. [NCT00884858]Phase 4100 participants (Actual)Interventional2009-04-30Completed
A Multicenter, Open Label Study Of Maraviroc, Zidovudine And Lamivudine Twice Daily For The Treatment Of Antiretroviral Naïve HIV-Infected Patients With R5 HIV-1 In Russia [NCT01275625]Phase 498 participants (Actual)Interventional2011-06-30Completed
A Pilot Project of Virologic, Pharmacologic and Immunologic Correlates of Gastrointestinal-Associated Lymphoid Tissue Immune Reconstitution Following Maraviroc Therapy [NCT00870363]Phase 444 participants (Actual)Interventional2009-04-30Completed
Kaletra and Maraviroc in Antiretroviral Therapy-Naïve Patients - KALMAR Study -Version 1.0 Amendment 2 [NCT01068873]Phase 41 participants (Actual)Interventional2010-04-30Terminated(stopped due to Poor enrollment)
ENLIGHTEN: Establishing Novel Antiretroviral (ARV) Imaging for Hair to Elucidate Non-Adherence [NCT03218592]Phase 436 participants (Actual)Interventional2017-06-28Completed
A Double-Blind, Randomised, Placebo-Controlled Phase I Trial to Compare the Pharmacokinetics of Maraviroc and Dapivirine Following Application of Maraviroc Vaginal Vaginal Gel, 0.1% 2.5g, Dapivirine Vaginal Gel, 0.05%, 2.5g and Maraviroc 0.1% + Dapivirine [NCT01242579]Phase 10 participants (Actual)Interventional2011-01-31Withdrawn(stopped due to In order to focus efforts on the combination ring formulation, IPM decided not to move forward with this trial.)
Effects of Maraviroc (MVC) on HIV-related Kaposi's Sarcoma (KS) [NCT01276236]Phase 213 participants (Actual)Interventional2011-03-09Completed
Multi Interventional Study Exploring HIV-1 Residual Replication: a Step Towards HIV-1 Eradication and Sterilizing Cure [NCT02961829]30 participants (Actual)Interventional2015-07-31Completed
Impact of CCR5 Blockade in HIV+ Kidney Transplant Recipients [NCT02741323]Phase 297 participants (Actual)Interventional2017-01-01Completed
Multicenter, Randomized, Non-comparative, Controlled Study of Therapeutic Intensification Plus Immunomodulation in HIV-infected Patients With Long-term Viral Suppression [NCT00976404]Phase 228 participants (Actual)Interventional2009-11-30Completed
Pilot Study of the Effect of Maraviroc Intensification on Peripheral Blood Monocyte HIV DNA Levels When Given to HIV-Infected Subjects Stable on Highly Active Antiretroviral Therapy With Undetectable Plasma HIV RNA [NCT00987948]Phase 215 participants (Actual)Interventional2010-01-31Completed
Open Randomized Study Comparing Two Alternatives of Antiretroviral Therapy as Post-exposure Prophylaxis to HIV-1: TENOFOVIR+EMTRICITABINA + LOPINAVIR/RITONAVIR VS TENOFOVIR+EMTRICITABINA + MARAVIROC [NCT01533272]Phase 4240 participants (Actual)Interventional2012-02-29Completed
A Phase II, Randomized Trial of Open-Label Truvada With Darunavir/Ritonavir Versus Multiclass Therapy With Truvada, Darunavir/Ritonavir, Maraviroc and Raltegravir in Acutely HIV-1 Infected Antiretroviral-Naïve Subjects [NCT00525733]40 participants (Actual)Interventional2007-10-31Completed
Switch to Maraviroc and Integrase Strand Transfer Inhibitor Combination Therapy (a Triple Class-Sparing Regimen) for the Treatment of HIV-1-Infected Patients on Suppressive Antiretroviral Regimens [NCT01896921]Phase 37 participants (Actual)Interventional2013-09-30Completed
A Phase II Study to Assess the Efficacy of Maraviroc in Prophylaxis of GVHD in Patients With Hematologic Malignancies Undergoing Reduced-Intensity Allogeneic SCT From Unrelated Donors [NCT01785810]Phase 237 participants (Actual)Interventional2013-02-28Completed
The Effects of the Direct Acting Antiviral Agent Boceprevir on the Pharmacokinetics of Maraviroc in Healthy Volunteers [NCT01627717]Phase 111 participants (Actual)Interventional2012-06-30Completed
Maraviroc Switch Central Nervous System (CNS) Substudy: a Substudy of MARCH, a Randomised, Open-label Study to Evaluate the Efficacy and Safety of Maraviroc (MVC) as a Switch for Either Nucleoside or Nucleotide Analogue Reverse Transcriptase Inhibitors (N [NCT01637233]28 participants (Actual)Observational2012-06-30Completed
Maraviroc Switch Collaborative Study Renal Substudy [NCT01637259]Phase 476 participants (Actual)Interventional2012-06-30Completed
PILOT STUDY OF PROTECTION AGAINST ex Vivo HIV INFECTION IN RECTAL MUCOSA IN HEALTHY VOLUNTEERS AFTER ADMINISTRATION OF MARAVIROC [NCT01719627]Phase 110 participants (Actual)Interventional2012-10-31Completed
Treatment of Advanced Colorectal Cancer Patients With Hepatic Liver Metastases Using the CCR5-Antagonist Maraviroc (Phase I Maracon Trial) [NCT01736813]12 participants (Actual)Observational2012-11-30Completed
Exploring HIV Entry Blockade as a Pre-exposure Prophylaxis Strategy in Women [NCT01749566]Phase 131 participants (Actual)Interventional2012-12-31Completed
A Randomized, Single-Blinded, Placebo-Controlled Two-Way Crossover Study To Investigate The Hemodynamic Effects Of Single Dose Vardenafil In Subjects Receiving Maraviroc [NCT00853840]Phase 418 participants (Actual)Interventional2008-04-30Completed
The Penetration of Maraviroc Into the Central Nervous System in HIV-1 Infected Subjects on Stable Antiretroviral Therapy; a Phase I Pharmacokinetic Study [NCT00982878]Phase 113 participants (Actual)Interventional2009-09-30Completed
A Phase 1, Open-label, Parallel-group Study To Assess The Effect Of Cyp3a5 Genotype On The Pharmacokinetics Of Maraviroc And Cyp3a5-derived Metabolites With And Without Darunavir/Cobicistat In African-american And Caucasian Healthy Volunteers [NCT02625207]Phase 147 participants (Actual)Interventional2015-11-06Completed
Characterization and Modulation of Mucosal Immunity for HIV Prevention in Women [NCT02333045]4 participants (Actual)Interventional2015-01-31Terminated(stopped due to Minimal efficacy of maraviroc alone was found in preliminary data analysis of another study.)
A Phase 2b, Double-Blind, Placebo-Controlled, Exploratory Randomized Trial to Determine the Bone, Immunologic, Virologic, and Neurocognitive Effects of a Novel Maraviroc-Containing Antiretroviral Regimen in Treatment-Naïve Patients Infected With R5-Tropic [NCT01400412]Phase 2262 participants (Actual)Interventional2012-01-17Completed
Safety and Efficacy of Maraviroc in Post-stroke Cognitive Impairment [NCT04966429]Phase 2150 participants (Anticipated)Interventional2021-05-01Recruiting
A Randomized, Open-Label, Crossover Study to Evaluate the Bioequivalence of a Combined Formulated Tablet Compared With Maraviroc and Combivir™ Administered Concurrently in Healthy Adult Subjects [NCT01597648]Phase 142 participants (Actual)Interventional2011-11-30Completed
Open-Label, Randomized, Fixed-Sequence, Crossover Study to Estimate the Effect of Telaprevir and Boceprevir on Maraviroc Pharmacokinetics in Healthy Subjects [NCT01597895]Phase 114 participants (Actual)Interventional2012-07-31Completed
The CAMAROS Trial: The Canadian Maraviroc RCT To Augment Rehabilitation Outcomes After Stroke [NCT04789616]Phase 2120 participants (Anticipated)Interventional2022-09-15Recruiting
Efficacy of Maraviroc in Modulating Atherosclerosis in HIV Patients. [NCT03402815]Phase 422 participants (Actual)Interventional2015-01-01Completed
A Randomized, Double-Blinded, Placebo-Controlled Trial Comparing Antiretroviral Intensification With Maraviroc and Dolutegravir With No Intensification or Intensification With Dolutegravir Alone for the Treatment of Cognitive Impairment in HIV [NCT02519777]Phase 4191 participants (Actual)Interventional2016-04-21Completed
MARaviroc-based Treatment Switch in HIV-positive Patients With HAND: Consequences of Reducing Antiretroviral-associated Neurotoxicity [NCT03163277]Phase 438 participants (Actual)Interventional2017-05-15Terminated(stopped due to Sloww accrual and COVID-19 related problems (impossible to perform LPs))
A Multicentre, 48 Week Randomised Controlled Factorial Trial of Adding Maraviroc and/or Metformin for Hepatic Steatosis in HIV-1-infected Adults on Combination Antiretroviral Therapy. [NCT03129113]Phase 2/Phase 390 participants (Actual)Interventional2017-03-01Completed
A Feasibility Study to Assess Protection of Vaginal and Cervical Tissues From Ex-Vivo HIV-1 Challenge Following Oral Administration of Maraviroc and Tenofovir [NCT02039323]Phase 16 participants (Actual)Interventional2014-02-28Completed
HIV Neurocognitive Disorders: A Randomized Clinical Trial of CNS-Targeted HAART [NCT00624195]Phase 2/Phase 359 participants (Actual)Interventional2007-03-31Completed
Raltegravir and Maraviroc in Combination for the Treatment of Antiretroviral Naïve HIV-1 Infected Patients [NCT01204905]7 participants (Actual)Interventional2010-09-30Completed
The Impact of CCR5 Inhibitor Treatment Intensification on CD4+ T-cell Recovery and Gene Expression Profiles in HIV-Infected Patients With Viral Suppression [NCT00925756]Phase 432 participants (Actual)Interventional2008-10-20Completed
A Drug Interaction Study Investigating the Effect of Rifabutin on the Pharmacokinetics of Maraviroc [NCT01894776]Phase 115 participants (Actual)Interventional2013-06-30Completed
A Multi-center Phase II Trial Randomizing Novel Approaches for Graft-versus-Host Disease Prevention Compared to Contemporary Controls (BMT CTN #1203; Progress I) [NCT02208037]Phase 2279 participants (Actual)Interventional2014-08-31Completed
The Effect of Sirolimus Plus Maraviroc on the Expression of Chemokine Receptor 5 (CCR5) and the HIV-1 Viral Reservoir in HIV-Infected Renal Transplant Recipients [NCT02990312]Phase 40 participants (Actual)Interventional2017-05-01Withdrawn(stopped due to Lack of enrollment)
A Randomised Controlled Clinical Trial of the Efficacy of HAART Intensification With Maraviroc in HIV Virally Suppressed Patients With Cognitive Impairment [NCT01449006]Phase 419 participants (Actual)Interventional2011-10-31Completed
Bicentric, Phase 2, Randomized, Open-label Study to Evaluate the Efficacy and Safety of Maraviroc Associated With Standard Treatment in Hospitalized Patients With Pulmonary SARS-CoV-2 Infection (COVID-19). [NCT04441385]Phase 260 participants (Actual)Interventional2020-06-26Terminated(stopped due to lack of viability)
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
"Phase2, Randomized, Controlled Study to Evaluate the Efficacy and Safety of Maraviroc and/or Favipiravir Plus Standard Therapy in Adult Patients With Severe Non-critical COVID-19" [NCT04475991]Phase 219 participants (Actual)Interventional2021-07-13Terminated(stopped due to No more patients complying the selection criteria were available for recruitment)
A Phase II Randomized, Double-Blind, Study of the Safety and Tolerability of Maraviroc (MVC), Maraviroc + Emtricitabine (MVC+FTC), Maraviroc + Tenofovir Disoproxil Fumarate (MVC+TDF), or Tenofovir Disoproxil Fumarate + Emtricitabine (TDF+FTC) For Pre-Expo [NCT01505114]Phase 2594 participants (Actual)Interventional2012-06-30Completed
Maraviroc as Graft Versus Host Disease Prophylaxis in Pediatric and Adult Stem Cell Transplant Recipients [NCT02167451]Phase 1/Phase 231 participants (Actual)Interventional2014-07-31Terminated(stopped due to slow recruitment and no further drug supply)
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00042289 (26) [back to overview]PK Parameter: Trough Concentration (C12) With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Trough Concentration (C24) With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Area Under the Curve From 0 to 12 Hours (AUC12) With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Area Under the Curve From 0 to 24 Hours (AUC24) With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Maximum Concentration (Cmax) in mg/L With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Trough Concentration (C24) With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Trough Concentration (C24) With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]Area Under the Curve From 0 to 12 Hours (AUC12) of ARVs for Contraceptive Arms
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 Geometric Mean (95% CI) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Maximum Concentration (Cmax) in ng/mL With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Maximum Concentration (Cmax) in ng/mL With Median (95% CI) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Maximum Concentration (Cmax) in mg/L With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Maximum Concentration (Cmax) in mg/L With Median (IQR) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]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 24 Hours (AUC24) With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Area Under the Curve From 0 to 24 Hours (AUC24) With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Area Under the Curve From 0 to 12 Hours (AUC12) With Geometric Mean (95% CI) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]Pharmacokinetic (PK) Parameter: Infant Plasma Washout Concentration of ARVs and TB Drugs
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]Area Under the Curve From 0 to 24 Hours (AUC24) of ARVs for Contraceptive Arms
NCT00042289 (26) [back to overview]Plasma Concentration for Contraceptives
NCT00042289 (26) [back to overview]PK Parameter: Cord/Maternal Blood Concentration Ratio With Median (Range) for ARVs and TB Drugs
NCT00042289 (26) [back to overview]Pharmacokinetic (PK) Parameter: Infant Plasma Washout Half-life (T1/2) of ARVs and TB Drugs
NCT00042289 (26) [back to overview]PK Parameter: Cord/Maternal Blood Concentration Ratio With Median (IQR) for ARVs and TB Drugs
NCT00098293 (17) [back to overview]Change From Baseline in Lymphocyte Cluster of Differentiation 8 (CD8) Count at Week 48 and 96
NCT00098293 (17) [back to overview]Number of Participants Per Tropism Status at Baseline and at the Time of Treatment Failure Through Week 48
NCT00098293 (17) [back to overview]Number of Participants Per Tropism Status at Baseline and at the Time of Treatment Failure Through Week 96
NCT00098293 (17) [back to overview]Number of Participants With NRTI Associated Mutations at Time of Treatment Failure Through Week 48 and 96
NCT00098293 (17) [back to overview]Percentage of Participants With Viral Load of Less Than 400 Copies/Milliliter [Copies/mL] and Less Than 50 Copies/mL of Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA) at Week 48 for Full Analysis Set (FAS) Population
NCT00098293 (17) [back to overview]Time-Averaged Difference (TAD) in log10-transformed HIV-1 RNA Levels
NCT00098293 (17) [back to overview]Time to Virologic Failure
NCT00098293 (17) [back to overview]Percentage of Participants With Viral Load of Less Than 400 Copies/mL and Less Than 50 Copies/mL of HIV-1 RNA at Week 96
NCT00098293 (17) [back to overview]Percentage of Participants With Viral Load of Less Than 400 Copies/mL and Less Than 50 Copies/mL of HIV-1 RNA at Week 48 for Per Protocol (PP) Population
NCT00098293 (17) [back to overview]Percentage of Participants With HIV-1 RNA Levels of Less Than 400 Copies/mL and Less Than 50 Copies/mL at Week 96 for Enhanced Sensitivity Trofile Assay (ESTA) R5 Participants
NCT00098293 (17) [back to overview]Number of Participants With Efavirenz Associated Mutations at Time of Treatment Failure Through Week 48 and 96
NCT00098293 (17) [back to overview]Percentage of Participants With HIV-1 RNA Levels of Less Than 400 Copies/mL and Less Than 50 Copies/mL at Week 96 Analyzed Using Logistic Regression
NCT00098293 (17) [back to overview]Percentage of Participants With HIV-1 RNA Levels of Less Than 400 Copies/mL and Less Than 50 Copies/mL at Week 48 for Enhanced Sensitivity Trofile Assay (ESTA) R5 Participants
NCT00098293 (17) [back to overview]Change From Baseline in Log 10-transformed Plasma Viral Load (HIV-1 RNA) Levels at Week 48 and 96
NCT00098293 (17) [back to overview]Number of Participants With Phenotypic Resistance at Time of Treatment Failure Through Week 48 and 96
NCT00098293 (17) [back to overview]Percentage of Participants With HIV-1 RNA Levels of Less Than 400 Copies/mL and Less Than 50 Copies/mL at Week 48 Analyzed Using Logistic Regression
NCT00098293 (17) [back to overview]Change From Baseline in Lymphocyte Cluster of Differentiation 4 (CD4) Count at Week 48 and 96
NCT00098306 (18) [back to overview]Time to Virological Failure
NCT00098306 (18) [back to overview]Change From Baseline in CD4 Cell Count at Week 24 and 48
NCT00098306 (18) [back to overview]Change From Baseline in CD8 Cell Count at Week 24 and 48
NCT00098306 (18) [back to overview]Number of Participants With Change in GSS and PSS From Screening at the Time of Treatment Failure Through Week 24
NCT00098306 (18) [back to overview]Cluster of Differentiation 4 (CD4) and Cluster of Differentiation 8 (CD8) Cell Count at Baseline
NCT00098306 (18) [back to overview]Number of Participants Per Tropism Status at Baseline and at the Time of Treatment Failure Through Week 24
NCT00098306 (18) [back to overview]Number of Participants Per Tropism Status at Baseline and at the Time of Treatment Failure Through Week 48
NCT00098306 (18) [back to overview]Number of Participants With Change in GSS and PSS From Screening at the Time of Treatment Failure at Week 48
NCT00098306 (18) [back to overview]Change From Baseline in Log 10-transformed HIV-1 RNA Levels at Week 24
NCT00098306 (18) [back to overview]Number of Participants With Genotypic Susceptibility Scores (GSS) and Phenotypic Susceptibility Score (PSS) at Screening
NCT00098306 (18) [back to overview]Change From Baseline in Viral Load at Week 24 and Week 48 by Overall Susceptibility Score (OSS) at Screening
NCT00098306 (18) [back to overview]Percentage of Participants With HIV-1 RNA Levels Less Than 400 Copies/mL or With at Least 0.5 log10 Decrease From Baseline
NCT00098306 (18) [back to overview]Percentage of Participants With HIV-1 RNA Levels Less Than 400 Copies/mL or With at Least 1.0 log10 Decrease From Baseline
NCT00098306 (18) [back to overview]Percentage of Participants With HIV-1 RNA Levels Less Than 50 Copies/mL
NCT00098306 (18) [back to overview]Time-Averaged Difference (TAD) From Baseline in log10 Transformed HIV-1 RNA Levels
NCT00098306 (18) [back to overview]Change From Baseline in Log 10-transformed HIV-1 RNA Levels at Week 48
NCT00098306 (18) [back to overview]Log 10-transformed Human Immunodeficiency Virus Ribonucleic Acid (HIV-1 RNA) Levels at Baseline
NCT00098306 (18) [back to overview]Percentage of Participants With HIV-1 RNA Levels Less Than 400 Copies/mL
NCT00098722 (18) [back to overview]Change From Baseline in CD8 Cell Count at Week 24 and 48
NCT00098722 (18) [back to overview]Percentage of Participants With HIV-1 RNA Levels Less Than 50 Copies/mL
NCT00098722 (18) [back to overview]Time-Averaged Difference (TAD) From Baseline in log10 Transformed HIV-1 RNA Levels
NCT00098722 (18) [back to overview]Percentage of Participants With HIV-1 RNA Levels Less Than 400 Copies/ml or With at Least 0.5 log10 Decrease From Baseline
NCT00098722 (18) [back to overview]Percentage of Participants With HIV-1 RNA Levels Less Than 400 Copies/mL
NCT00098722 (18) [back to overview]Number of Participants With Genotypic Susceptibility Score (GSS) and Phenotypic Susceptibility Score (PSS) at Screening
NCT00098722 (18) [back to overview]Number of Participants With Change in GSS and PSS From Screening at the Time of Treatment Failure Through Week 48
NCT00098722 (18) [back to overview]Number of Participants With Change in GSS and PSS From Screening at the Time of Treatment Failure Through Week 24
NCT00098722 (18) [back to overview]Number of Participants Per Tropism Status at Baseline and at the Time of Treatment Failure Through Week 48
NCT00098722 (18) [back to overview]Number of Participants Per Tropism Status at Baseline and at the Time of Treatment Failure Through Week 24
NCT00098722 (18) [back to overview]Cluster of Differentiation 4 (CD4) and Cluster of Differentiation 8 (CD8) Cell Count at Baseline
NCT00098722 (18) [back to overview]Change From Baseline in Viral Load at Week 24 and Week 48 by Overall Susceptibility Score (OSS) at Screening
NCT00098722 (18) [back to overview]Change From Baseline in CD4 Cell Count at Week 24 and 48
NCT00098722 (18) [back to overview]Time to Virological Failure
NCT00098722 (18) [back to overview]Log 10-transformed Human Immunodeficiency Virus Ribonucleic Acid (HIV-1 RNA) Levels at Baseline
NCT00098722 (18) [back to overview]Change From Baseline in Log 10-transformed HIV-1 RNA Levels at Week 48
NCT00098722 (18) [back to overview]Change From Baseline in Log 10-transformed HIV-1 RNA Levels at Week 24
NCT00098722 (18) [back to overview]Percentage of Participants With HIV-1 RNA Levels Less Than 400 Copies/ml or With at Least 1.0 log10 Decrease From Baseline
NCT00098748 (15) [back to overview]Number of Subjects With HIV-1 RNA Levels < 400 Copies/mL or at Least 0.5 Log 10-transformed Decrease From Baseline in HIV-1 RNA Levels
NCT00098748 (15) [back to overview]Number of Subjects With HIV-1 RNA Levels < 50 Copies/mL
NCT00098748 (15) [back to overview]Number of Subjects With HIV-1 RNA Levels < 400 Copies/mL or at Least 1.0 Log 10-transformed Decrease From Baseline in HIV-1 RNA Levels
NCT00098748 (15) [back to overview]Change From Baseline in CD8 Cell Count
NCT00098748 (15) [back to overview]Number of Subjects With Treatment Failure at Week 24 by Overall Susceptibility Score (OSS) at Screening
NCT00098748 (15) [back to overview]Time (50% Quartile Point Estimate) to Virologic Failure
NCT00098748 (15) [back to overview]Number of Subjects With Treatment Failure at Week 48 by Overall Susceptibility Score (OSS) at Screening
NCT00098748 (15) [back to overview]Number of Subjects With HIV-1 RNA Levels < 400 Copies/mL
NCT00098748 (15) [back to overview]Number of Subjects With Acquired Immunodeficiency Syndrome (AIDS)-Defining Opportunistic Illnesses (Analysis at Week 48)
NCT00098748 (15) [back to overview]Number of Subjects With Acquired Immunodeficiency Syndrome (AIDS)-Defining Opportunistic Illnesses (Analysis at Week 24)
NCT00098748 (15) [back to overview]Number of Subjects Per Tropism Status at Screening and Time of Treatment Failure (Analysis at Week 48)
NCT00098748 (15) [back to overview]Number of Subjects Per Tropism Status at Screening and at the Time of Treatment Failure (Analysis at Week 24)
NCT00098748 (15) [back to overview]Change From Baseline in Time Averaged Difference (TAD) in log10 HIV-1 RNA
NCT00098748 (15) [back to overview]Change From Baseline in Human Immunodeficiency Virus (HIV-1) Viral Load (Ribonucleic Acid [RNA])
NCT00098748 (15) [back to overview]Change From Baseline in CD4 Cell Count
NCT00426660 (21) [back to overview]Percentage of Participants With All Causality Treatment-emergent Adverse (AEs) Events by Gender
NCT00426660 (21) [back to overview]Percentage of Participants With ≥0.5 log10 Reduction From Baseline in Human Immunodeficiency Virus 1 Ribonucleic Acid (HIV 1 RNA)
NCT00426660 (21) [back to overview]Change From Baseline in CD8 Cell Count
NCT00426660 (21) [back to overview]Percentage of Participants With HIV-1 RNA Levels Below the Limit of Quantification: <50 Copies/mL
NCT00426660 (21) [back to overview]Percentage of Participants With HIV-1 RNA Levels Below the Limit of Quantification: <400 Copies/mL
NCT00426660 (21) [back to overview]Percentage of Participants With Grade 4 Laboratory Abnormalities Without Regards to Baseline Abnormalities
NCT00426660 (21) [back to overview]Percentage of Participants With Change in Chemokine Co-receptor Tropism From Screening to Time of Virologic Failure
NCT00426660 (21) [back to overview]Change From Baseline in CD8 Cell Count Percent
NCT00426660 (21) [back to overview]Percentage of Participants With Treatment-emergent Averse Events (AEs) by Baseline Hepatitis B and Hepatitis C Virus Serology Status
NCT00426660 (21) [back to overview]Percentage of Participants With Treatment-emergent Adverse Events (AEs) by Race
NCT00426660 (21) [back to overview]Percentage of Participants With Treatment-emergent Adverse Events (AEs) by Age
NCT00426660 (21) [back to overview]Percentage of Participants With ≥1.0 log10 Reduction From Baseline in HIV 1 RNA
NCT00426660 (21) [back to overview]Percentage of Participants With Acquired Immunodeficiency Syndrome (AIDS)-Defining Illnesses
NCT00426660 (21) [back to overview]Median Time to Virologic Failure
NCT00426660 (21) [back to overview]Number of Participants With Emergence of Resistance to Maraviroc as Defined by Genotypic Changes in the V3 Loop of Glycoprotein 120 (gp 120)
NCT00426660 (21) [back to overview]Number of Participants With Reduced Maraviroc Susceptibility as Defined by Change From Baseline to Time of Virologic Failure in Inhibitory Concentration of 50% (IC 50) and Presence of Plateau
NCT00426660 (21) [back to overview]Change From Baseline in CD4 Cell Count
NCT00426660 (21) [back to overview]Change From Baseline in CD4 Cell Count Percent
NCT00426660 (21) [back to overview]Percentage of Participants With Changes in HIV-1 RNA Level in Participants Meeting the Definition of Virologic Failure
NCT00426660 (21) [back to overview]Percentage of Participants With Grade 3 and Grade 4 Adverse Events (AE)
NCT00426660 (21) [back to overview]Percentage of Participants With Grade 3 Laboratory Abnormalities Without Regards to Baseline Abnormalities
NCT00427934 (25) [back to overview]Number of Subjects With Categorical Absolute Vital Signs and Vital Sign Changes Compared to Baseline
NCT00427934 (25) [back to overview]Number of Subjects With Withdrawal From Study Due to Lack of Efficacy
NCT00427934 (25) [back to overview]Survival Analysis of Time to Withdrawal: Proportion of Subjects Who Did Not Withdraw From the Study Due to Lack of Efficacy.
NCT00427934 (25) [back to overview]Change From Baseline in 12-Lead Electrocardiogram (ECG) Parameters (RR Interval, PR Interval, QRS Complex, QT Interval, Corrected QT [QTc] Interval, QTcB Interval [Bazett's Correction], QTcF Interval [Fridericia's Correction]).
NCT00427934 (25) [back to overview]Change From Baseline in 12-Lead Electrocardiogram (ECG) Parameters (Heart Rate).
NCT00427934 (25) [back to overview]American College of Rheumatology (ACR) 20% Responders at Week 12
NCT00427934 (25) [back to overview]ACR 70% Responders at Weeks 1, 2, 4, 8, and 12
NCT00427934 (25) [back to overview]Change From Baseline in CRP at Weeks 1, 2, 4, 8, and 12
NCT00427934 (25) [back to overview]Change From Baseline in Disease Activity Score Using CRP (DAS28-4[CRP]) at Weeks 1, 2, 4, 8, and 12
NCT00427934 (25) [back to overview]Change From Baseline in HAQ-DI at Weeks 1, 2, 4, 8, and 12
NCT00427934 (25) [back to overview]Change From Baseline in Mean Heart Rate
NCT00427934 (25) [back to overview]Change From Baseline in Mean Orthostatic Blood Pressure (BP)
NCT00427934 (25) [back to overview]Change From Baseline in Patient's Assessment of Arthritis Pain at Weeks 1, 2, 4, 8, and 12
NCT00427934 (25) [back to overview]Change From Baseline in Patient's Global Assessment of Arthritis Pain at Weeks 1, 2, 4, 8, and 12
NCT00427934 (25) [back to overview]Change From Baseline in Physician's Global Assessment of Arthritis Pain at Weeks 1, 2, 4, 8, and 12
NCT00427934 (25) [back to overview]Area Under the Plasma Concentration-Time Profile From Time Zero to Four Hours Postdose (AUC 0-4) for MTX at Screening and Week 1 and Maraviroc at Week 1
NCT00427934 (25) [back to overview]Change From Baseline in SF-36 Mental Component Summary at Weeks 4 and 12
NCT00427934 (25) [back to overview]Change From Baseline in Short Form-36 (SF-36) Physical Component Summary at Weeks 4 and 12
NCT00427934 (25) [back to overview]Change From Baseline in Swollen Joint Count at Weeks 1, 2, 4, 8, and 12
NCT00427934 (25) [back to overview]Change From Baseline in Tender/Painful Joint Count at Weeks 1, 2, 4, 8, and 12
NCT00427934 (25) [back to overview]Maximum Observed Concentration (Cmax) During the Dosing Interval for MTX at Screening and Week 1 and Maraviroc at Week 1
NCT00427934 (25) [back to overview]Number of Subjects With Categorical Absolute ECG Parameters and ECG Changes Compared to Baseline
NCT00427934 (25) [back to overview]Time for Cmax (Tmax) for MTX at Screening and Week 1 and Maraviroc at Week 1
NCT00427934 (25) [back to overview]ACR 50% Responders at Weeks 1, 2, 4, 8, and 12
NCT00427934 (25) [back to overview]ACR 20% Responders at Weeks 1, 2, 4, and 8
NCT00478231 (16) [back to overview]Time to Virologic Failure (VF)
NCT00478231 (16) [back to overview]Change From Baseline in CD4 Percent at Week 4, 8, 12, 24, 36, 48, 60, 72, 84 and Week 96 or EOT
NCT00478231 (16) [back to overview]Change From Baseline in CD8 Percent at Week 4, 8, 12, 24, 36, 48, 60, 72, 84 and Week 96 or EOT
NCT00478231 (16) [back to overview]Change From Baseline in Human Immunodeficiency Virus (HIV) -1 Viral Load (Ribonucleic Acid [RNA]) at Week 4, 8, 12, 24, 36, 48, 60, 72, 84 and Week 96 or EOT
NCT00478231 (16) [back to overview]Change From Baseline in Lymphocyte Cluster of Differentiation 4 (CD4) Count at Week 4, 8, 12, 24, 36, 48, 60, 72, 84 and Week 96 or EOT
NCT00478231 (16) [back to overview]Change From Baseline in Lymphocyte Cluster of Differentiation 8 (CD8) Count at Week 4, 8, 12, 24, 36, 48, 60, 72, 84 and Week 96 or EOT
NCT00478231 (16) [back to overview]Number of Participants With C-X-C Chemokine Receptor Type 4 {CXCR4} [X4] Tropism Status
NCT00478231 (16) [back to overview]Number of Participants With Category C Acquired Immunodeficiency Syndrome (AIDS) Related Infections
NCT00478231 (16) [back to overview]Number of Participants With Division of Acquired Immunodeficiency Syndrome (DAIDS) Grade 3 and Grade 4 Laboratory Abnormalities
NCT00478231 (16) [back to overview]Number of Participants With Genotype Resistance
NCT00478231 (16) [back to overview]Number of Participants With Grade 3 and Grade 4 Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT00478231 (16) [back to overview]Number of Participants With Laboratory Test Abnormalities
NCT00478231 (16) [back to overview]Number of Participants With Treatment Emergent Malignancies
NCT00478231 (16) [back to overview]Percentage of Participants Achieving HIV-1 RNA Below Limit of Quantification
NCT00478231 (16) [back to overview]Percentage of Participants With at Least 0.5 Log 10 Reduction in Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA)
NCT00478231 (16) [back to overview]Percentage of Participants With at Least 1.0 Log 10 Reduction in HIV-1 RNA
NCT00525733 (1) [back to overview]The Primary Outcome of This Study is the Proportion of Patients Having Detectable HIV-1 RNA Using the Single Copy Assay After 48 Weeks of Treatment and the Study Hypothesis is That New Treatment is Better Than the Control Group.
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 CD4 Count From Baseline
NCT00537394 (15) [back to overview]Change in Plasma HIV-1 Viral Load From Baseline to Week 1
NCT00537394 (15) [back to overview]Number of Participants With Change in Virus Co-receptor Tropism Among Those With R5-only Tropic Virus at Study Entry
NCT00537394 (15) [back to overview]Participants With Newly Acquired HIV Drug Resistance Between Study Entry and Confirmed Virologic Failure
NCT00537394 (15) [back to overview]Percent of Participants With Regimen Failure, Defined as a Confirmed Virologic Failure or Discontinuation of Randomized NRTI Component of Study Treatment
NCT00537394 (15) [back to overview]Change in Cardiovascular Risk Score From Baseline
NCT00537394 (15) [back to overview]Change in 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
NCT00624195 (1) [back to overview]Neuropsychological Performance Change
NCT00665561 (15) [back to overview]Density Rate Per 1000 Participant-Years for Incidence of Myocardial Infarction or Ischemia: Events Adjudicated as 'Definite' or 'Possible'
NCT00665561 (15) [back to overview]Density Rate Per 1000 Participant-Years for Incidence of Centers for Disease Control and Prevention Category C AIDS -Defining Opportunistic Infections
NCT00665561 (15) [back to overview]Adjusted Density Rate Per 1000 Participant-Years for Incidence of Myocardial Infarction or Ischemia: Events Adjudicated as 'Definite' or 'Possible' or 'Insufficient Data'
NCT00665561 (15) [back to overview]Adjusted Density Rate Per 1000 Participant-Years for Incidence of Myocardial Infarction or Ischemia: Events Adjudicated as 'Definite' or 'Possible'
NCT00665561 (15) [back to overview]Adjusted Density Rate Per 1000 Participant-Years for Incidence of Death Due to Any Cause
NCT00665561 (15) [back to overview]Density Rate Per 1000 Participant-Years for Incidence of Death Due to Any Cause
NCT00665561 (15) [back to overview]Density Rate Per 1000 Participant-Years for Incidence of Death From Liver-Related Cause
NCT00665561 (15) [back to overview]Adjusted Density Rate Per 1000 Participant-Years for Incidence of Centers for Disease Control and Prevention Category C Aids-Defining Opportunistic Infections
NCT00665561 (15) [back to overview]Density Rate Per 1000 Participant-Years for Incidence of Liver Failure
NCT00665561 (15) [back to overview]Density Rate Per 1000 Participant-Years for Incidence of Myocardial Infarction or Ischemia: Events Adjudicated as 'Definite' or 'Possible' or 'Insufficient Data'
NCT00665561 (15) [back to overview]Density Rate Per 1000 Participant-Years for Incidence of Rhabdomyolysis
NCT00665561 (15) [back to overview]Density Rate Per 1000 Participant-Years for Incidence of Viral Encephalitis
NCT00665561 (15) [back to overview]Percentage of Participants With All-Cause Mortality
NCT00665561 (15) [back to overview]Adjusted Density Rate Per 1000 Participant-Years for Incidence of All Malignancies (AIDS Defining Malignancies and Non-AIDS Defining Malignancies)
NCT00665561 (15) [back to overview]Density Rate Per 1000 Participant-Years for Incidence of All Malignancies (AIDS Defining Malignancies and Non-AIDS Defining Malignancies)
NCT00666705 (6) [back to overview]Maraviroc Pharmacokinetic (PK) Parameter: 12-Hour Trough Concentration (C12)
NCT00666705 (6) [back to overview]Maraviroc Pharmacokinetic (PK) Parameter: Maximum Concentration (Cmax)
NCT00666705 (6) [back to overview]Raltegravir Pharmacokinetics (PK) Parameter: 12-Hour Trough Concentration (C12)
NCT00666705 (6) [back to overview]Raltegravir Pharmacokinetics (PK) Parameter: Area Under the Plasma Concentration-time Profile Over the Dosing Interval (AUCτ)
NCT00666705 (6) [back to overview]Maraviroc Pharmacokinetic (PK) Parameter: Area Under the Plasma Concentration-time Profile Over the Dosing Interval (AUCτ)
NCT00666705 (6) [back to overview]Raltegravir Pharmacokinetics (PK) Parameter: Maximum Concentration (Cmax)
NCT00709111 (35) [back to overview]Change in IL-6, MCP-1, MCP-2, and Plasma CD40L
NCT00709111 (35) [back to overview]Change in IL-6, MCP-1, MCP-2, and Plasma CD40L
NCT00709111 (35) [back to overview]Change in Intercellular Cell Adhesion Molecule (ICAM)-1, Plasma P-selectin, Soluble TNFRII (sTNFRII), and Matrix Metalloproteinase (MMP)-9
NCT00709111 (35) [back to overview]Change in Interleukin (IL)-6, Monocyte Chemoattractant Protein (MCP)-1, MCP-2, and Plasma CD40 Ligand (CD40L)
NCT00709111 (35) [back to overview]Change in Percentage of CD4+ T-cells That Are: naïve (%CD45RA+CCR7+), Central Memory (%CD45RA-CCR7+), Effector Memory (%CD45RA-CCR7-), Effector (%CD45RA+CCR7-), %HLA-DR+CD38+, %CD38+, %Ki67+, %caspase3+, %Bcl-2-, and %CD57+
NCT00709111 (35) [back to overview]Change in Percentage of CD4+ T-cells That Are: naïve (%CD45RA+CCR7+), Central Memory (%CD45RA-CCR7+), Effector Memory (%CD45RA-CCR7-), Effector (%CD45RA+CCR7-), %HLA-DR+CD38+, %CD38+, %Ki67+, %caspase3+, %Bcl-2-, and %CD57+
NCT00709111 (35) [back to overview]Change in Percentage of CD4+ T-cells That Are: naïve (%CD45RA+CCR7+), Central Memory (%CD45RA-CCR7+), Effector Memory (%CD45RA-CCR7-), Effector (%CD45RA+CCR7-), %HLA-DR+CD38+, %CD38+, %Ki67+, %caspase3+, %Bcl-2-, and %CD57+
NCT00709111 (35) [back to overview]Change in Percentage of CD8+ T-cells That Are: naïve (%CD45RA+CCR7+), Central Memory (%CD45RA-CCR7+), Effector Memory (%CD45RA-CCR7-), Effector (%CD45RA+CCR7-), %HLA-DR+CD38+, %CD38+, %Ki67+, %caspase3+, %Bcl-2-, and %CD57+
NCT00709111 (35) [back to overview]Change in Percentage of CD8+ T-cells That Are: naïve (%CD45RA+CCR7+), Central Memory (%CD45RA-CCR7+), Effector Memory (%CD45RA-CCR7-), Effector (%CD45RA+CCR7-), %HLA-DR+CD38+, %CD38+, %Ki67+, %caspase3+, %Bcl-2-, and %CD57+
NCT00709111 (35) [back to overview]Change in Percentage of CD8+ T-cells That Are: naïve (%CD45RA+CCR7+), Central Memory (%CD45RA-CCR7+), Effector Memory (%CD45RA-CCR7-), Effector (%CD45RA+CCR7-), %HLA-DR+CD38+, %CD38+, %Ki67+, %caspase3+, %Bcl-2-, and %CD57+
NCT00709111 (35) [back to overview]Drug Adherence Assessed as Number of Missed Doses Over a 4-day Recall
NCT00709111 (35) [back to overview]Number of Subjects Who Experience a Grade 2, 3 or 4 Signs and Symptoms, Grade 3 or 4 Laboratory Abnormalities, or Death.
NCT00709111 (35) [back to overview]Proportion of Participants With Detectable HIV-1 Viremia as Measured by Single Copy Assay (SCA)
NCT00709111 (35) [back to overview]Change in ICAM-1, Plasma P-selectin, sTNFRII, and MMP-9
NCT00709111 (35) [back to overview]Change in D-dimer
NCT00709111 (35) [back to overview]Change From Within-subject Pre-treatment CD4+ T-cell Count Slopes to Corresponding Within-subject CD4+ T-cell Count Slopes From Baseline Through Week 24
NCT00709111 (35) [back to overview]Change in CD4 Percentage
NCT00709111 (35) [back to overview]Change in CD4 Percentage
NCT00709111 (35) [back to overview]Change in CD4 Percentage
NCT00709111 (35) [back to overview]Change in ICAM-1, Plasma P-selectin, sTNFRII, and MMP-9
NCT00709111 (35) [back to overview]Change in CD4+ T-cell Count
NCT00709111 (35) [back to overview]Change in CD4+ T-cell Count
NCT00709111 (35) [back to overview]Change in CD4+ T-cell Count
NCT00709111 (35) [back to overview]Change in D-dimer
NCT00709111 (35) [back to overview]Change in D-dimer
NCT00709111 (35) [back to overview]Change in High Sensitivity C-reactive Protein (Hs-CRP)
NCT00709111 (35) [back to overview]Change in Hs-CRP
NCT00709111 (35) [back to overview]Change From Within-subject Pre-treatment CD4 Percentage Slopes to Corresponding Within-subject CD4 Percentage Slopes From Baseline Through Week 24
NCT00709111 (35) [back to overview]Change in Hs-CRP
NCT00709111 (35) [back to overview]Change in Soluble CD14
NCT00709111 (35) [back to overview]Change in Soluble CD14
NCT00709111 (35) [back to overview]Change in Soluble CD14
NCT00709111 (35) [back to overview]Proportion of Participants Achieving a 50-cell Increase in CD4+ T-cell Count
NCT00709111 (35) [back to overview]Within-subject CD4 Percentage Slopes
NCT00709111 (35) [back to overview]Within-subject CD4+ T-cell Count Slopes
NCT00717067 (13) [back to overview]Renal Clearance (CLR) in Subjects With Normal, Mild, Moderate and Severe Renal Function
NCT00717067 (13) [back to overview]Maximum Observed Plasma Concentration (Cmax)
NCT00717067 (13) [back to overview]Hemodialysis Clearance of Maraviroc (MVC) in Subjects With End Stage Renal Disease (ESRD) Undergoing Hemodialysis: CLdD
NCT00717067 (13) [back to overview]Half-life (t1/2)
NCT00717067 (13) [back to overview]Derivation of Renal Clearance in Subjects With Normal, Mild, Moderate and Severe Renal Function: Ae
NCT00717067 (13) [back to overview]AUCtau
NCT00717067 (13) [back to overview]Safety and Tolerability of Maraviroc in the Absence and Presence of a Potent CYP3A4 Inhibitor in Subjects With Various Degrees of Renal Impairment or Undergoing Hemodialysis: Number of Subjects With Maximum EGC QTC, QTCB and QTCF Intervals
NCT00717067 (13) [back to overview]Safety and Tolerability of Maraviroc in the Absence and Presence of a Potent CYP3A4 Inhibitor in Subjects With Various Degrees of Renal Impairment or Undergoing Hemodialysis: Number of Subjects With Maximum Increase and Decrease in Supine Blood Pressure
NCT00717067 (13) [back to overview]Area Under the Plasma Concentration Time-curve From Zero to the Last Measured Concentration (AUClast)
NCT00717067 (13) [back to overview]Safety and Tolerability of Maraviroc in the Absence and Presence of a Potent CYP3A4 Inhibitor in Subjects With Various Degrees of Renal Impairment or Undergoing Hemodialysis: Number of Subjects With Pulse Rate < 40 and > 120 Beats Per Minute
NCT00717067 (13) [back to overview]Plasma Protein Binding
NCT00717067 (13) [back to overview]Time of First Occurrence (Tmax)
NCT00717067 (13) [back to overview]Area Under the Time Curve From 0 to Infinity (AUCinf)
NCT00735072 (5) [back to overview]Change in CD4+ T Cell Count
NCT00735072 (5) [back to overview]Week 24 Change in Percentage of CD8+ T Cells That Co-express CD38 and HLA DR (Week 24 %CD38+HLA-DR+ CD8+ T Cells Minus Baseline %CD38+HLA-DR+ CD8+ T Cells)
NCT00735072 (5) [back to overview]Change in Ultra-sensitive Plasma HIV RNA Level (Single Copy/ml Assay)
NCT00735072 (5) [back to overview]Change in Gut-associated Lymphoid Tissue HIV RNA Level (UCSF Site Only)
NCT00735072 (5) [back to overview]Change in Brachial Artery Flow-mediated Dilatation (UCSF Site Only)
NCT00764465 (5) [back to overview]AUC: Steady-state Plasma MVC PK Following Administration of RTV
NCT00764465 (5) [back to overview]AUC: Steady-state Plasma Amprenavir (APV) Pharmacokinetics ( PK) Following Admin of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent (Maraviroc) MVC 300mg BID.
NCT00764465 (5) [back to overview]Number of Participants Who Experienced an Adverse Event
NCT00764465 (5) [back to overview]Cmin/Cmax: Steady-state Plasma Amprenavir (APV) Pharmacokinetics ( PK) Following Admin of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent (Maraviroc) MVC 300mg BID.
NCT00764465 (5) [back to overview]Cmin/Cmax: Steady-state Plasma MVC PK Following Administration of RTV
NCT00791700 (19) [back to overview]Percentage of Participants With HIV-1 RNA Levels < 48 Copies/mL at Weeks 24 and 48 Using MD=F Approach
NCT00791700 (19) [back to overview]Percentage of Participants With HIV-1 RNA Levels <400 Copies/mL at Weeks 24 and 48 Using Missing, Discontinuation = Failure (MD=F)Approach
NCT00791700 (19) [back to overview]Percentage of Participants With Optimized Background Treatment Susceptibility Scores
NCT00791700 (19) [back to overview]Pharmacokinetic(PK): Maraviroc PK Parameter, Average Plasma Concentration (Cavg), Trough Concentration(Cmin), Maximum Plasma Concentration (Cmax)
NCT00791700 (19) [back to overview]Time to Reach Maximum Plasma Concentration (Tmax)
NCT00791700 (19) [back to overview]Percentage of Participants With HIV-1 RNA <400 Copies/mL and <48 Copies/mL Using the Time to Loss of Virologic Response Algorithm (TLOVR) at Week 48
NCT00791700 (19) [back to overview]Percentage of Participants With HIV-1 RNA <400 Copies/mL Through Week 48 Using Missing, Discontinuation = Failure (MD=F) Approach
NCT00791700 (19) [back to overview]Treatment Discontinuation: Secondary Reasons- Serious Adverse Event (SAE) Related to Study Drug
NCT00791700 (19) [back to overview]Area Under the Curve at Steady State (AUCtau)
NCT00791700 (19) [back to overview]Change From Baseline in Cluster of Differentiation 4 (CD4+) Cell Count at Weeks 24 and 48
NCT00791700 (19) [back to overview]Change From Baseline in HIV-1 RNA (Log10 Copies/mL)
NCT00791700 (19) [back to overview]Change From Baseline in HIV-1 RNA (Original)
NCT00791700 (19) [back to overview]Change From Baseline in Percentage (%) of CD4+ Cells at Weeks 24 and 48
NCT00791700 (19) [back to overview]Incidence and Severity of Grade 3 and Grade 4 Treatment-Emergent Adverse Events(AEs) (All Causality)
NCT00791700 (19) [back to overview]Number of Participants With Emergence of Reverse Transcriptase Inhibitor (RTI) and Protease Inhibitor (PI) Resistance Associated Mutations (RAMs) Between Screening and On-Treatment Confirmed PDVF
NCT00791700 (19) [back to overview]Number of Participants With Protocol Defined Virologic Failure
NCT00791700 (19) [back to overview]Number of Participants With Viral Tropism Between Screening and Confirmed Protocol Defined Virologic Failure (PDVF) Prior to Week 48
NCT00791700 (19) [back to overview]Percentage of Participants With >= 1.0 log10 Reduction in HIV-1RNA Concentration From Baseline to Week 24 and Week 48
NCT00791700 (19) [back to overview]Percentage of Participants With HIV-1 RNA <48 Copies/mL Through Week 48 Using Missing, Discontinuation = Failure (MD=F)Approach
NCT00827112 (16) [back to overview]HIV-1 RNA Levels at Baseline
NCT00827112 (16) [back to overview]Maximum Observed Plasma Concentration (Cmax) of Maraviroc
NCT00827112 (16) [back to overview]Number of Participants With Phenotypic Resistance
NCT00827112 (16) [back to overview]Change From Baseline in Cluster of Differentiation 8+T Lymphocyte (CD8) Cell Count at Weeks 16, 24, 48 and 96
NCT00827112 (16) [back to overview]Percentage of Participants With Less Than 400 Copies/mL of HIV-1 RNA
NCT00827112 (16) [back to overview]Change From Baseline in Cluster of Differentiation 4+T Lymphocyte (CD4) Cell Counts at Weeks 16, 24, 48 and 96
NCT00827112 (16) [back to overview]Time to Loss of Virological Response (TLOVR)
NCT00827112 (16) [back to overview]Percentage of Participants With Plasma Human Immuno Deficiency Virus-1 Ribonucleic Acid (HIV-1 RNA) Levels Less Than 50 Copies/Milliliter (mL)
NCT00827112 (16) [back to overview]Time-Averaged Difference (TAD) in log10 Viral Load
NCT00827112 (16) [back to overview]Average Observed Plasma Concentration (Cavg) of Maraviroc
NCT00827112 (16) [back to overview]Number of Participants With Genotypic Resistance
NCT00827112 (16) [back to overview]Change From Baseline in Plasma log10 Viral Load at Weeks 16, 24, 48 and 96
NCT00827112 (16) [back to overview]Number of Participants With HIV-1 RNA Tropism Status Using Trofile Assay
NCT00827112 (16) [back to overview]Percentage of Participants With Less Than 50 Copies/mL of HIV-1 RNA
NCT00827112 (16) [back to overview]Minimum Observed Plasma Concentration (Cmin) of Maraviroc
NCT00827112 (16) [back to overview]Change From Baseline in HIV-1 RNA Levels of First 15 Participants at Days 4, 7, 10 and 14
NCT00844519 (1) [back to overview]Percent Change in FMD
NCT00850395 (13) [back to overview]Change From Baseline in Cluster of Differentiation 4 (CD4+) Cell Counts at Month 6
NCT00850395 (13) [back to overview]Change From Baseline in Log 10 Transformed Human Immunodeficiency Virus-1 Ribonucleic Acid (HIV-1 RNA) at Month 3
NCT00850395 (13) [back to overview]Change From Baseline in Log 10 Transformed Human Immunodeficiency Virus-1 Ribonucleic Acid (HIV-1 RNA) at Month 6
NCT00850395 (13) [back to overview]Number of Participants With Human Immunodeficiency Virus (HIV) Response
NCT00850395 (13) [back to overview]Change From Baseline in Acquired Immune Deficiency Syndrome (AIDS) Clinical Trials Group (ACTG) Symptom Distress Module (SDM) Overall Score at Months 6 and 12
NCT00850395 (13) [back to overview]Number of Participants Taking Concomitant Therapy
NCT00850395 (13) [back to overview]Number of Participants With Centers for Disease Control and Prevention (CDC) Classification at Month 12
NCT00850395 (13) [back to overview]Number of Participants With Centers for Disease Control and Prevention (CDC) Classification at Month 3
NCT00850395 (13) [back to overview]Number of Participants With Centers for Disease Control and Prevention (CDC) Classification at Month 6
NCT00850395 (13) [back to overview]Physician's Assessment of Efficacy
NCT00850395 (13) [back to overview]Change From Baseline in Log 10 Transformed Human Immunodeficiency Virus-1 Ribonucleic Acid (HIV-1 RNA) at Month 12
NCT00850395 (13) [back to overview]Change From Baseline in Cluster of Differentiation 4 (CD4+) Cell Counts at Month 12
NCT00850395 (13) [back to overview]Change From Baseline in Cluster of Differentiation 4 (CD4+) Cell Counts at Month 3
NCT00853840 (5) [back to overview]Postural Changes in Pulse Rate
NCT00853840 (5) [back to overview]Number of Subjects With Postural Hypotension
NCT00853840 (5) [back to overview]Standing and Supine Systolic and Diastolic Blood Pressure (BP)
NCT00853840 (5) [back to overview]Standing and Supine Pulse Rate
NCT00853840 (5) [back to overview]Postural Changes in Systolic and Diastolic Blood Pressure
NCT00864474 (35) [back to overview]Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Mean Daily Dose of Celsentri
NCT00864474 (35) [back to overview]Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Inpatient or Outpatient Status
NCT00864474 (35) [back to overview]Percentage of Participants With Adverse Drug Reactions (ADRs): Factor HIV Infection Duration
NCT00864474 (35) [back to overview]Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Gender
NCT00864474 (35) [back to overview]Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Ethnicity
NCT00864474 (35) [back to overview]Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Centers for Disease Control and Prevention (CDC) Classification
NCT00864474 (35) [back to overview]Percentage of Participants With Adverse Drug Reactions (ADRs): Factor as Per Presence or Absence of Concomitant Therapies
NCT00864474 (35) [back to overview]Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Age
NCT00864474 (35) [back to overview]Mean Number of Plasma Human Immuno-Deficiency Virus-Ribosomal Ribonucleic Acid (HIV-RNA) Copies: Factor The Presence or Absence of Comorbidities
NCT00864474 (35) [back to overview]Mean Number of Plasma Human Immuno-Deficiency Virus-Ribosomal Ribonucleic Acid (HIV-RNA) Copies: Factor Gender
NCT00864474 (35) [back to overview]Mean Number of Plasma HIV-RNA Copies: Factor The Presence or Absence of Use of Cytochrome P450 3A4 (CYP3A4) Enzyme Inducer Taken Along With Celsentri
NCT00864474 (35) [back to overview]Mean Number of Plasma HIV-RNA Copies: Factor The Presence or Absence of History of Therapies for HIV Infection
NCT00864474 (35) [back to overview]Mean Number of Plasma HIV-RNA Copies: Factor The Centers for Disease Control and Prevention (CDC) Classification
NCT00864474 (35) [back to overview]Mean Number of Plasma HIV-RNA Copies for Participants Who Took Concomitant Therapies Along With Celsentri
NCT00864474 (35) [back to overview]Mean Number of CD4+ Lymphocyte: Factor The Presence or Absence of Use of Cytochrome P450 3A4 (CYP3A4) Enzyme Inducer Taken Along With Celsentri
NCT00864474 (35) [back to overview]Mean Number of CD4+ Lymphocyte Counts: Fcator The Presence or Absence of Comorbidities
NCT00864474 (35) [back to overview]Mean Number of CD4+ Lymphocyte Counts: Factor The Centers for Disease Control and Prevention (CDC) Classification
NCT00864474 (35) [back to overview]Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Presence or Absence of Hemophilia
NCT00864474 (35) [back to overview]Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Presence or Absence of Comorbidities
NCT00864474 (35) [back to overview]Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Presence or Absence of Allergies
NCT00864474 (35) [back to overview]Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Number of Concomitant Anti-HIV Drugs Use
NCT00864474 (35) [back to overview]Mean Number of CD4+ Lymphocyte Counts: Factor Presence or Absence of History of Therapies for HIV Infection
NCT00864474 (35) [back to overview]Percentage of Participants With Adverse Drug Reactions (ADRs)
NCT00864474 (35) [back to overview]Number of Participants With Unknown Adverse Drug Reactions (ADRs)
NCT00864474 (35) [back to overview]Number of Participants With Tropism Switch From CCR5- to CXCR4-Tropic Variants
NCT00864474 (35) [back to overview]Number of Adverse Drug Reactions (ADRs) Considered to Have Occurred Due to Effect of Celsentri on Immune Function
NCT00864474 (35) [back to overview]Number of Adverse Drug Reactions (ADRs) Considered to Have Occurred Due to Effect of Celsentri on Hepatic Function
NCT00864474 (35) [back to overview]Number of Adverse Drug Reactions (ADRs) Considered to Have Occurred Due to Effect of Celsentri on Cardiovascular Effects
NCT00864474 (35) [back to overview]Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Mean Total Dose of Celsentri
NCT00864474 (35) [back to overview]Mean Number of Cluster of Differentiation of More Than 4 (CD4+) Lymphocyte Count: Factor Gender
NCT00864474 (35) [back to overview]Primary: Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Presence or Absence of History of Therapies for Human Immuno-Deficiency Virus (HIV) Infection
NCT00864474 (35) [back to overview]Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Total Number of Days of Administration of Celsentri
NCT00864474 (35) [back to overview]Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Presence or Absence of Use of Cytochrome P450 3A4 (CYP3A4) Enzyme Inducer Taken Along With Celsentri
NCT00864474 (35) [back to overview]Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Presence or Absence of Renal Impairment
NCT00864474 (35) [back to overview]Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Presence or Absence of Hepatic Impairment
NCT00870363 (4) [back to overview]Trough Plasma and Tissue Drug Levels in Volunteers at the Time of the Upper Endoscopy
NCT00870363 (4) [back to overview]Change in HIV DNA Per 10^6 Cells in Duodenal Tissue Versus PBMC by Drug Regimen Received
NCT00870363 (4) [back to overview]Changes in CD4+ T-cell Numbers by Treatment Regimen
NCT00870363 (4) [back to overview]Change in the Density of CD3+/CD4+ Cells Per Cubic Millimeter at the Effector Sites in the Duodenal Tissues Following Antiretroviral Therapy Regimen
NCT00925756 (3) [back to overview]Change in CD4+/CD8+ T-cell Immune Activation, Maturation, Regulatory and Apoptosis Markers at Baseline and Weeks 4 and 24.
NCT00925756 (3) [back to overview]Differences in Gene Expression Profiles Obtained at Baseline and Week 4 and Week 24.
NCT00925756 (3) [back to overview]CD4+ T-cell Absolute Count and Percentage at Baseline, Weeks 4 and 24.
NCT00948753 (6) [back to overview]Rate of Early Mortality After Transplant
NCT00948753 (6) [back to overview]Number of Patients Treated With Maraviroc During SCT That Develop Chronic GVHD
NCT00948753 (6) [back to overview]Number of Participants Who Relapsed During Study Period
NCT00948753 (6) [back to overview]Efficacy of Maraviroc
NCT00948753 (6) [back to overview]Pharmacokinetic Profile of Maraviroc in Patients Undergoing Nonmyeloablative Allogeneic SCT
NCT00948753 (6) [back to overview]Safety of Maraviroc
NCT00976404 (5) [back to overview]Change From Baseline in HIV DNA in Rectal Tissue at Week 56
NCT00976404 (5) [back to overview]Serious Adverse Events Attributed to Study Treatments
NCT00976404 (5) [back to overview]HIV Specific T-cell Response to Env
NCT00976404 (5) [back to overview]Change From Baseline in HIV DNA in PBMCs at Week 56
NCT00976404 (5) [back to overview]Change From Baseline in CD4+ T Cell Count at Week 56
NCT00982878 (2) [back to overview]Number of Participants With Adverse Events
NCT00982878 (2) [back to overview]CSF (Cerebrospinal Fluid) : Plasma Ratio of Maraviroc 1H Magnetic Resonance Spectroscopy (1H-MRS)
NCT00987948 (2) [back to overview]Change From Baseline to 24 Weeks in Neuropsychological Performance As Measured by Age- and Education-Adjusted Z-Scores
NCT00987948 (2) [back to overview]Change From Baseline to 24 Weeks in HIV DNA (Log-10 Copies/10^6 Cells) as Measured by HIV DNA Within CD14+ Peripheral Blood Mononuclear Cells
NCT00993148 (9) [back to overview]Drug Adherence, Number of Participants With Missed Doses
NCT00993148 (9) [back to overview]Drug Resistance Mutations and Co-receptor Tropism Assessed by Trofile ES
NCT00993148 (9) [back to overview]Median CD4 Count Change From Baseline
NCT00993148 (9) [back to overview]Percentage of Participants With Plasma HIV-1 RNA >50
NCT00993148 (9) [back to overview]Percentage of Participants With Plasma HIV-1 RNA >50 Copies/mL
NCT00993148 (9) [back to overview]Percentage of Participants With Virologic Failure or Off Study Treatment Regimen
NCT00993148 (9) [back to overview]Proportion of Participants With Plasma HIV-1 RNA >50 Copies/mL
NCT00993148 (9) [back to overview]Signs/Symptoms or Laboratory Toxicities of Grade 3 or Higher
NCT00993148 (9) [back to overview]Trough Concentrations (Ctrough) of Maraviroc
NCT01068873 (1) [back to overview]Number of Participants With HIV RNA < 50 and <400 Copies/ml.
NCT01154673 (1) [back to overview]Change in Proviral HIV-1 DNA in Total CD4+ T-cells From Baseline to Week 48 in Participants Randomized to the Intensified Arm Versus the Control Arm Who Received Placebo in Addition to Standard HAART.
NCT01204905 (2) [back to overview]Viral Load
NCT01204905 (2) [back to overview]Viral Suppression
NCT01275625 (11) [back to overview]Percentage of Participants With Plasma Human Immuno Deficiency Virus-1 Ribonucleic Acid (HIV-1 RNA) Load <50 Copies/Milliliter (mL) at 48 Weeks.
NCT01275625 (11) [back to overview]Number of Participants With Genotypic Resistance.
NCT01275625 (11) [back to overview]Number of Participants With HIV-1 RNA Tropism Status Using Genotyping Assay at Screening and at the Time of Virologic Failure.
NCT01275625 (11) [back to overview]Virologic Response: Percentage of Participants With Plasma HIV-1 RNA Load < 400 Copies/mL at Post-baseline Visits.
NCT01275625 (11) [back to overview]Virologic Response: Percentage of Participants With Plasma HIV-1 RNA Load <50 Copies/mL at Post-baseline Visits.
NCT01275625 (11) [back to overview]Virologic Response: Rate of Virologic Failure at Week 48.
NCT01275625 (11) [back to overview]Immunological Response at Week 48: Absolute Change From Baseline in Absolute Cluster of Differentiation 4 (CD4)
NCT01275625 (11) [back to overview]Immunological Response at Week 48: Absolute Change From Baseline in Absolute Cluster of Differentiation 8 (CD8)
NCT01275625 (11) [back to overview]Immunological Response at Week 48: Change From Baseline in Absolute Cluster of Differentiation 4 (CD4)/ Cluster of Differentiation 8 (CD8) Ratio.
NCT01275625 (11) [back to overview]Immunological Response at Week 48: Percentage Change From Baseline in Absolute Cluster of Differentiation 4 (CD4)
NCT01275625 (11) [back to overview]Immunological Response at Week 48: Percentage Change From Baseline in Absolute Cluster of Differentiation 8 (CD8)
NCT01276236 (7) [back to overview]Change in Edema Grade
NCT01276236 (7) [back to overview]Number of Participants With a Decrease in Kaposi's Sarcoma (KS) Total Surface Area
NCT01276236 (7) [back to overview]Percent Change in CCR5 Levels on CD4+ T-Cells
NCT01276236 (7) [back to overview]Percent Change in CCR5 Levels on CD8+ T-cells
NCT01276236 (7) [back to overview]Percent Change in CD69 Expression in a Subset of Double Negative DR-CD38 Negative (DR-CD38-) T-cells
NCT01276236 (7) [back to overview]Percent Change in CD69 Expression in a Subset of Double Negative DR-CD38 Positive (DR-CD38+) T-cells
NCT01276236 (7) [back to overview]Percent Change in KS Total Surface Area
NCT01327547 (21) [back to overview]Mean Change From Baseline in the Hepatic Elastography (FibroscanTM) at Week 48, 96 and 144
NCT01327547 (21) [back to overview]Mean Change From Baseline in Plasma Hepatitis B Virus (HBV) DNA at Week 48, 96 and 144
NCT01327547 (21) [back to overview]Mean Change From Baseline in Markers of Immune Activation: Transforming Growth Factor-beta (TGF Beta) - Week 48, 96 and 144
NCT01327547 (21) [back to overview]Mean Change From Baseline in Markers of Immune Activation: D Dimer - Week 48, 96 and 144
NCT01327547 (21) [back to overview]Mean Change From Baseline in Markers of Immune Activation: C-reactive Protein (CRP) - Week 48, 96 and 144.
NCT01327547 (21) [back to overview]Mean Change From Baseline in Log10 Plasma Hepatitis C Virus (HCV) RNA at Week 48, 96 and 144
NCT01327547 (21) [back to overview]Mean Change From Baseline in Enhanced Liver Fibrosis (ELF) Test at Week 48, 96 and 144
NCT01327547 (21) [back to overview]Mean Change From Baseline in CD38 Expression on CD4 and CD8 Cells at Weeks 48, 96 and 144
NCT01327547 (21) [back to overview]Change From Baseline in Fibrosis Score (Ishak) in Liver Biopsy Samples at Week 144
NCT01327547 (21) [back to overview]Absolute Fibrosis Score (Ishak) in Liver Biopsy Samples at Baseline and at Week 144
NCT01327547 (21) [back to overview]Time to Development of Grade 3 and Grade 4 ALT Abnormalities at Week 144 Associated With a Change From Baseline ALT >100 IU/L
NCT01327547 (21) [back to overview]Mean Change From Baseline in CD4+ and CD8+ Cell Counts at Week 48, 96 and 144
NCT01327547 (21) [back to overview]Time to Development of Grade 3 and Grade 4 ALT Abnormalities
NCT01327547 (21) [back to overview]Percentage of Participants With Grade 3 and Grade 4 Alanine Aminotransferase (ALT) Abnormalities at Week 48
NCT01327547 (21) [back to overview]Number of Participants With Hy's Law Abnormalities Through Week 144
NCT01327547 (21) [back to overview]Exposure-response Relationship Between Change From Baseline in Liver Fibrosis Biomarkers Versus MVC Cavg at Week 48
NCT01327547 (21) [back to overview]Summary of Estimated Maraviroc PK Parameters
NCT01327547 (21) [back to overview]Percentage of Participants With Plasma Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA) Concentration <40 Copies/mL at Week 48, 96 and 144
NCT01327547 (21) [back to overview]Percentage of Participants With Grade 3 and Grade 4 ALT Abnormalities Through Week 144
NCT01327547 (21) [back to overview]Percentage of Participants With Grade 3 and Grade 4 ALT Abnormalities Associated With a Change From Baseline ALT >100 IU/L
NCT01327547 (21) [back to overview]Percentage of Participants Who Were Hospitalized Due to Hepatic Disease Through Week 144
NCT01345630 (25) [back to overview]Number of Participants With Viral Resistance to Maraviroc (Maraviroc Treated Participants Only) in Participants Meeting PDTF Criteria.
NCT01345630 (25) [back to overview]Percent Change From Baseline in Immune Cell Function at Week 48: Lymphocyte Activation Marker CD4 (%)
NCT01345630 (25) [back to overview]Percent Change From Baseline in Immune Cell Function at Week 48: Lymphocyte Activation Marker CD8 (%)
NCT01345630 (25) [back to overview]Severity of Abnormal Laboratory Values
NCT01345630 (25) [back to overview]Tropism Change Between Screening or Baseline and PDTF
NCT01345630 (25) [back to overview]Virologic Outcomes at Week 48 Using Protocol-Defined Treatment Failure (PDTF).
NCT01345630 (25) [back to overview]Change in Bone Turnover Markers From Baseline and at Week 48 - Blood Osteocalcin
NCT01345630 (25) [back to overview]Change in Bone Turnover Markers From Baseline and at Week 48 - Type 1 Collagen Peptide (CTX-1)
NCT01345630 (25) [back to overview]Changes in Bone Mineral Density (Using DEXA Scan and Serum Markers) From Baseline and at Week 48 - AP Lumbar Spine (L1 - L4) BMD
NCT01345630 (25) [back to overview]Changes in Bone Mineral Density (Using DEXA Scan and Serum Markers) From Baseline and at Week 48 - Femoral Neck BMD
NCT01345630 (25) [back to overview]Changes in Bone Mineral Density (Using DEXA Scan and Serum Markers) From Baseline and at Week 48 - Total Hip BMD
NCT01345630 (25) [back to overview]Changes in Peripheral Fat Distribution Using Dual Energy X-ray Absorptiometry [DEXA] Scan From Baseline and at Week 48.
NCT01345630 (25) [back to overview]Changes in Trunk to Limb Fat Distribution Using DEXA Scan From Baseline and at Week 48
NCT01345630 (25) [back to overview]Frequency of Adverse Events (AE).
NCT01345630 (25) [back to overview]Number of Participants Who Discontinued Due to AEs
NCT01345630 (25) [back to overview]Number of Participants With Grade 3 or 4 AEs
NCT01345630 (25) [back to overview]Number of Participants With Treatment-emergent Serious Adverse Events
NCT01345630 (25) [back to overview]Number of Treatment-related AEs
NCT01345630 (25) [back to overview]Percentage of Participants With Plasma HIV-1 RNA <50 Copies/mL.
NCT01345630 (25) [back to overview]The Relationship Between the Proportion of Participants With Plasma HIV-1 RNA <50 Copies/mL at the Week 48 and the Screening Tropism Test (Genotype Test or ESTA).
NCT01345630 (25) [back to overview]Absolute Change From Baseline in Immune Cell Function at Week 48: Lymphocyte Marker Cluster of Differentiation 4 (CD4, Cell/mm^3)
NCT01345630 (25) [back to overview]Absolute Change From Baseline in Immune Cell Function at Week 48: Lymphocyte Marker Cluster of Differentiation 8 (CD8, Cell/mm^3)
NCT01345630 (25) [back to overview]Absolute Change in CD4+/CD8+ Ratio From Baseline to Week 48
NCT01345630 (25) [back to overview]Number of Participants With Abnormal Laboratory Values
NCT01345630 (25) [back to overview]Number of Participants With Resistance to Nucleoside/Nucleotide Reverse Transcriptase Inhibitors (NRTI), Non-nucleoside Reverse Transcriptase Inhibitors (NNRTI), and Protease Inhibitors (PI) in Participants Meeting PDTF Criteria
NCT01348763 (2) [back to overview]The Ratio of the Maximum Plasma Concentration of Maraviroc Between 20 and 10 Day
NCT01348763 (2) [back to overview]Number of Participants With Changes in Haematology and Biochemistry Laboratory Tests
NCT01400412 (20) [back to overview]Change in Level of IP-10 From Baseline to Week 48
NCT01400412 (20) [back to overview]Percent Change in Expression of CD57+ on CD8+ T Cells From Baseline to Week 48
NCT01400412 (20) [back to overview]Percent Change in Expression of CD28+/CD57+ on CD8+ T Cells From Baseline to Week 48
NCT01400412 (20) [back to overview]Percent Change in Expression of CD28+ on CD8+ T Cells From Baseline to Week 48
NCT01400412 (20) [back to overview]Change in CD4 Count From Baseline to Week 24
NCT01400412 (20) [back to overview]Change in CD4 Count From Baseline to Week 48
NCT01400412 (20) [back to overview]Percent Change in Lumbar Spine Bone Mineral Density (BMD)
NCT01400412 (20) [back to overview]CD8+ T-cell Change From Baseline to Week 48
NCT01400412 (20) [back to overview]Percent Change in Expression of RANKL+ on CD8+ T Cells From Baseline to Week 48
NCT01400412 (20) [back to overview]Change in Levels of D-dimer From Baseline
NCT01400412 (20) [back to overview]Percentage Change in Expression of CD38+/HLA-DR+ on CD4+ T Cells From Baseline to Week 48
NCT01400412 (20) [back to overview]Change in Levels of IL-6 From Baseline to Week 48
NCT01400412 (20) [back to overview]Change in Levels of sCD14 From Baseline
NCT01400412 (20) [back to overview]Change in Levels of sCD163 From Baseline to Week 48
NCT01400412 (20) [back to overview]Number of Participants Who Developed Grade 3 or 4 Primary Adverse Events
NCT01400412 (20) [back to overview]Number of Participants Who Died During the Study
NCT01400412 (20) [back to overview]Cumulative Probability of Virologic Failure by Week 48
NCT01400412 (20) [back to overview]Number of Participants Who Experienced Bone Fractures
NCT01400412 (20) [back to overview]Percent Change From Baseline in Total Hip Bone Mineral Density (BMD)
NCT01400412 (20) [back to overview]Percentage Change in Expression of CD38+/HLA-DR+ on CD8+ T Cells From Baseline to Week 48
NCT01449006 (4) [back to overview]Change in MRS Cerebral Metabolite Ratios in Frontal White Matter
NCT01449006 (4) [back to overview]Change in MRS Cerebral Metabolite Ratios in Basal Ganglia
NCT01449006 (4) [back to overview]Change in CSF Neopterin Concentration
NCT01449006 (4) [back to overview]Change in Neurocognitive Functioning
NCT01505114 (1) [back to overview]Occurrence of Grade 3 or Higher Adverse Events (AEs)
NCT01785810 (1) [back to overview]Day +180 Rate of Grade II-IV Acute GVHD
NCT01894776 (3) [back to overview]Safety/Tolerability of the Treatments
NCT01894776 (3) [back to overview]Pharmacokinetics of Maraviroc and Rifabutin AUC 0-12/24
NCT01894776 (3) [back to overview]Maraviroc and Rifabutin C12/C24/Cmax PK Concentrations in Plasma.
NCT01896921 (3) [back to overview]Number of Participants With Adverse Events
NCT01896921 (3) [back to overview]Number of Patients Virologically Suppressed (HIV RNA <50 Copies/ml) at 48 Weeks.
NCT01896921 (3) [back to overview]Number of Patients Who Are Virologically Suppressed (HIV RNA < 50 Copies/ml)
NCT02167451 (12) [back to overview]Toxicities
NCT02167451 (12) [back to overview]Area Under The Concentration-Time Curve (AUC) of Maraviroc
NCT02167451 (12) [back to overview]Infectious Complications
NCT02167451 (12) [back to overview]Area Under The Concentration-Time Curve (AUC) of Maraviroc
NCT02167451 (12) [back to overview]Feasibility of Maraviroc
NCT02167451 (12) [back to overview]Graft Failure
NCT02167451 (12) [back to overview]GVHD Incidence
NCT02167451 (12) [back to overview]Incidence of Visceral GVHD
NCT02167451 (12) [back to overview]Overall Survival
NCT02167451 (12) [back to overview]Primary Disease Relapse
NCT02167451 (12) [back to overview]Time to Neutrophil
NCT02167451 (12) [back to overview]Time to Platelet Engraftment
NCT02208037 (13) [back to overview]Percentage of Participants With Neutrophil Recovery
NCT02208037 (13) [back to overview]Percentage of Participants With Chronic GVHD
NCT02208037 (13) [back to overview]Percentage of Participants With Chronic GVHD Requiring Immunosupressive Therapy
NCT02208037 (13) [back to overview]Percentage of Participants With Disease Relapse or Progression
NCT02208037 (13) [back to overview]Percentage of Participants With Grade III-IV Acute GVHD
NCT02208037 (13) [back to overview]Percentage of Participants With GVHD-free Survival
NCT02208037 (13) [back to overview]Percentage of Participants With Grade II-IV Acute GVHD
NCT02208037 (13) [back to overview]Percentage of Participants With GVHD/Relapse or Progression-free Survival (GRFS)
NCT02208037 (13) [back to overview]Percentage of Participants With Overall Survival
NCT02208037 (13) [back to overview]Percentage of Participants With Transplant-Related Mortality (TRM)
NCT02208037 (13) [back to overview]Percentage of Participants With Platelet Recovery
NCT02208037 (13) [back to overview]Donor Cell Engraftment
NCT02208037 (13) [back to overview]Percentage of Participants With Disease-free Survival
NCT02333045 (1) [back to overview]Count of Total Cells Obtained From Cervicovaginal Lavage Samples
NCT02519777 (17) [back to overview]Number of Participants With Treatment Related Adverse Events (AEs)
NCT02519777 (17) [back to overview]CD8+ T-cell Counts
NCT02519777 (17) [back to overview]Change in CD4+ T-cell Count
NCT02519777 (17) [back to overview]Change in CD8+ T-cell Count
NCT02519777 (17) [back to overview]Change in Functional Status Scores
NCT02519777 (17) [back to overview]Change in Normalized Composite Neurocognitive Test Score at Weeks 24, 72, and 96 From Baseline
NCT02519777 (17) [back to overview]Number of Participants With Plasma HIV-1 RNA Greater Than or Equal to 50 Copies/mL
NCT02519777 (17) [back to overview]CD4+ T-cell Counts
NCT02519777 (17) [back to overview]Change in Log10 IP-10 in CSF at Week 48 From Baseline
NCT02519777 (17) [back to overview]Change in Log10 MIP-1 Beta in Cerebrospinal Fluid (CSF) at Week 48 From Baseline
NCT02519777 (17) [back to overview]Change in Log10 MIP-1 Beta in Plasma at Week 48 From Baseline
NCT02519777 (17) [back to overview]Change in Log10 Neopterin in CSF at Week 48 From Baseline
NCT02519777 (17) [back to overview]Change in Log10 NFL in CSF at Week 48 From Baseline
NCT02519777 (17) [back to overview]Change in Log10 sCD14 in Plasma at Week 48 From Baseline
NCT02519777 (17) [back to overview]Change in Log10 sTNFr-II in Plasma at Week 48 From Baseline
NCT02519777 (17) [back to overview]Change in Log10 VCAM in Plasma at Week 48 From Baseline
NCT02519777 (17) [back to overview]Change in Normalized Composite Neurocognitive Test Score at Week 48 From Baseline
NCT02625207 (24) [back to overview]Part 2: Plasma Concentration of Maraviroc at 24 Hours Post-dose
NCT02625207 (24) [back to overview]Part 2: Number of Participants With Laboratory Abnormalities
NCT02625207 (24) [back to overview]Part 2: Number of Participants With Clinically Significant Vital Sign Abnormalities
NCT02625207 (24) [back to overview]Part 2: Number of Participants With 12-Lead Electrocardiogram (ECG) Abnormalities
NCT02625207 (24) [back to overview]Part 2: Maximum Observed Plasma Concentration (Cmax) of Maraviroc
NCT02625207 (24) [back to overview]Part 2: Average Plasma Concentration (Cavg) of Maraviroc
NCT02625207 (24) [back to overview]Part 2: Area Under The Plasma Concentration-Time Curve From Time 0 to 24 Hours (AUC [0-24]) of Maraviroc
NCT02625207 (24) [back to overview]Part 1: Time to Reach Maximum Observed Plasma Concentration (Tmax) of Maraviroc
NCT02625207 (24) [back to overview]Part 1: Plasma Concentration of Maraviroc at 12 Hours Post-dose
NCT02625207 (24) [back to overview]Part 1: Number of Participants With 12-Lead Electrocardiogram (ECG) Abnormalities
NCT02625207 (24) [back to overview]Part 1: Maximum Observed Plasma Concentration (Cmax) of Maraviroc
NCT02625207 (24) [back to overview]Part 1: Number of Participants With Clinically Significant Vital Sign Abnormalities
NCT02625207 (24) [back to overview]Part 1: Average Plasma Concentration (Cavg) of Maraviroc
NCT02625207 (24) [back to overview]Part 1: Area Under The Plasma Concentration-Time Curve From Time 0 to 12 Hours (AUC [0-12]) of Maraviroc
NCT02625207 (24) [back to overview]Part 2: Number of Participants With Treatment Emergent Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT02625207 (24) [back to overview]Part 1: Number of Participants With Laboratory Abnormalities
NCT02625207 (24) [back to overview]Part 1: Time to Reach Maximum Observed Plasma Concentration (Tmax) of Metabolites of Maraviroc
NCT02625207 (24) [back to overview]Part 1: Plasma Concentration of Metabolites of Maraviroc at 12 Hour Post-dose
NCT02625207 (24) [back to overview]Part 1: Number of Participants With Treatment Emergent Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT02625207 (24) [back to overview]Part 1: Metabolite to Parent Ratio for Area Under the Concentration-Time Curve From Time 0 to 12 Hours for Maraviroc and Its Metabolites (MRAUC12)
NCT02625207 (24) [back to overview]Part 1: Maximum Observed Plasma Concentration (Cmax) of Metabolites of Maraviroc
NCT02625207 (24) [back to overview]Part 1: Average Plasma Concentration (Cav) of Metabolites of Maraviroc
NCT02625207 (24) [back to overview]Part 1: Area Under The Plasma Concentration-Time Curve From Time 0 to 12 Hours (AUC [0-12]) of Metabolites of Maraviroc
NCT02625207 (24) [back to overview]Part 2: Time to Reach Maximum Observed Plasma Concentration (Tmax) of Maraviroc
NCT02741323 (29) [back to overview]Tissue Common Rejection Module (tCRM) Score Using the 11-gene tCRM Module on FFPE Biopsy Shaves
NCT02741323 (29) [back to overview]Trough Levels of CCR5 Blockade (Maraviroc)
NCT02741323 (29) [back to overview]Urine Common Rejection Module (uCRM) Score Using the 11-gene uCRM Module on Urine Cell Pellets
NCT02741323 (29) [back to overview]Urine Common Rejection Module (uCRM) Score Using the 11-gene uCRM Module on Urine Cell Pellets
NCT02741323 (29) [back to overview]AUC of CCR5 Blockade (Maraviroc)
NCT02741323 (29) [back to overview]Incidence/Proportion of Participants With HIV Infection in the Renal Allograft
NCT02741323 (29) [back to overview]Calcineurin Inhibitor (Tacrolimus) AUC for Participants on Maraviroc Versus Placebo
NCT02741323 (29) [back to overview]Calcineurin Inhibitor (Tacrolimus) Trough Levels for Participants on Maraviroc Versus Placebo
NCT02741323 (29) [back to overview]Cumulative Incidence of Graft Loss, Toxicities ≥ Grade 3 Per the DAIDS Toxicity Table and/or Permanent Treatment Discontinuation
NCT02741323 (29) [back to overview]Cumulative Incidence/Proportion of Acute Cellular Rejection Grade Equal to or Greater Than 1A During the Entire Study Follow-up
NCT02741323 (29) [back to overview]Cumulative Incidence/Proportion of Biopsy Proven Acute Rejection Within 1 Year Post Transplant
NCT02741323 (29) [back to overview]HIV DNA in Peripheral Blood CD4+ T Cells at Week 52
NCT02741323 (29) [back to overview]HIV RNA in Peripheral Blood CD4+ T Cells at Week 52.
NCT02741323 (29) [back to overview]Incidence of All Adverse Events (AEs) Greater Than or Equal to Grade 3 at Year 1
NCT02741323 (29) [back to overview]Incidence of Death at Year 1
NCT02741323 (29) [back to overview]Incidence of Graft Loss in Year 1
NCT02741323 (29) [back to overview]Incidence of Non-opportunistic Infections Requiring Hospitalization Within Year 1
NCT02741323 (29) [back to overview]Incidence of Opportunistic Infections or Neoplasms Within Year 1
NCT02741323 (29) [back to overview]Incidence of Serious Adverse Events (SAEs) Greater Than or Equal to Grade 3 Within Year 1
NCT02741323 (29) [back to overview]Incidence/Proportion of Antibody Mediated Rejection
NCT02741323 (29) [back to overview]Mean CD45 Gene Expression Count (PTPRC)
NCT02741323 (29) [back to overview]Mean CD45 Quantitative Immunohistochemistry (IHC)
NCT02741323 (29) [back to overview]Mean eGFR at Week 52 Based on CKD-EPI Creatinine Equation
NCT02741323 (29) [back to overview]Mean Glomerular Filtration Rate by Iohexol Clearance at Week 52
NCT02741323 (29) [back to overview]Plasma HIV RNA Levels (Single Copy Assay) at Week 52
NCT02741323 (29) [back to overview]Proportion of Participants With de Novo Anti-donor Human Leukocyte Antigen (HLA) Antibodies
NCT02741323 (29) [back to overview]Proportion of Participants With Defined CKD Stage 4 or 5 at Year 1
NCT02741323 (29) [back to overview]Proportion of Participants With Estimated Glomerular Filtration Rate (eGFR) Less Than 60 mL/Min/1.73 m² at Week 52
NCT02741323 (29) [back to overview]The Slope of eGFR Over Time in Year 1
NCT02778204 (13) [back to overview]Number of Participants Failing to Meet PK Target
NCT02778204 (13) [back to overview]Pharmacokinetic (PK) Parameter: Area-under-the-curve (AUC)
NCT02778204 (13) [back to overview]Percentage of Participants Failing to Meet Long-Term Safety Endpoint Related to Maraviroc for Dose-Finding
NCT02778204 (13) [back to overview]Pharmacokinetic (PK) Parameter: Area-under-the-curve (AUC)
NCT02778204 (13) [back to overview]Pharmacokinetic (PK) Parameter: Average Concentration (Cavg)
NCT02778204 (13) [back to overview]Pharmacokinetic (PK) Parameter: Average Concentration (Cavg)
NCT02778204 (13) [back to overview]Number of Participants Failing to Meet PK Target
NCT02778204 (13) [back to overview]Percentage of Participants Failing to Meet Safety Endpoint Related to Maraviroc for Dose-Finding
NCT02778204 (13) [back to overview]Percentage of Participants Failing to Meet Safety Endpoint Related to Maraviroc for Analysis
NCT02778204 (13) [back to overview]Pharmacokinetic (PK) Parameter: Maximum Concentration (Cmax)
NCT02778204 (13) [back to overview]Pharmacokinetic (PK) Parameter: Time of Maximum Concentration (Tmax)
NCT02778204 (13) [back to overview]Pharmacokinetic (PK) Parameter: Trough Concentration (Ctau)
NCT02778204 (13) [back to overview]Percentage of Participants Failing to Meet Long-Term Safety Endpoint Related to Maraviroc for Analysis
NCT03218592 (4) [back to overview]Hair Antiretroviral Imaging
NCT03218592 (4) [back to overview]Plasma Antiretroviral Concentrations
NCT03218592 (4) [back to overview]Whole Blood Antiretroviral Concentrations
NCT03218592 (4) [back to overview]Peripheral Blood Mononuclear Cells (PBMC) Antiretroviral Concentrations
NCT03274804 (6) [back to overview]Efficacy Endpoint: Disease Control Rate
NCT03274804 (6) [back to overview]Efficacy Endpoint: Objective Response Rate
NCT03274804 (6) [back to overview]Efficacy Endpoint: Progression-free Survival
NCT03274804 (6) [back to overview]Feasibility Rate of a Combined Therapy
NCT03274804 (6) [back to overview]Overall Survival
NCT03274804 (6) [back to overview]Safety and Toxicity of a Combined Therapy Based on Subjects Who Experienced Toxicities

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

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

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

[back to top]

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

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

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

[back to top]

PK Parameter: Maximum Concentration (Cmax) in mg/L With Median (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: Trough Concentration (C24) With Median (Range) for ARVs and TB Drugs

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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 24 Hours (AUC24) With Median (Range) for ARVs and TB Drugs

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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]

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

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

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

[back to top]

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

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

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

[back to top]

Plasma Concentration for Contraceptives

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

PK Parameter: 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]

Change From Baseline in Lymphocyte Cluster of Differentiation 8 (CD8) Count at Week 48 and 96

Baseline value calculated as the average of pre-dose measurements collected at screening and immediately pre-dose. Change from baseline in lymphocyte CD8 count at Week 48 and 96 was not analyzed for participants originally randomized to maraviroc once daily arm since after termination, focus was shifted from efficacy and safety to only safety as reflected in the abbreviated set of efficacy measures noted in the amended planned analysis. (NCT00098293)
Timeframe: Baseline, Week 48, Week 96

,
Interventioncells/µL (Mean)
BaselineChange at Week 48Change at Week 96
Efavirenz Once Daily + CBV (DB)935.78-126.83-150.27
Maraviroc Twice Daily + CBV (DB)938.8038.3420.74

[back to top]

Number of Participants Per Tropism Status at Baseline and at the Time of Treatment Failure Through Week 48

Number of participants per tropism status (C-X-C chemokine receptor 5 {CCR5} [R5], C-X-C chemokine receptor type 4 {CXCR4} [X4], Dual/mixed [DM], or Non-reportable/Non-phenotypable [NR/NP]) at baseline and time of treatment failure analyzed through week 48 visit. Treatment failure: discontinuation due to insufficient clinical response. Tropism result was censored for participants with viral load <500 copies/mL at time of treatment failure categorized as below lower limit of quantification (BLQ). The assessment for time of treatment failure was defined as last on treatment assessment. (NCT00098293)
Timeframe: Baseline, time of failure through Week 48

,,
Interventionparticipants (Number)
Baseline: R5; Treatment failure: R5Baseline: R5; Treatment failure: X4Baseline: R5; Treatment failure: DMBaseline: R5; Treatment failure: NR/NPBaseline: DM; Treatment failure: R5Baseline: DM; Treatment failure: X4Baseline: DM; Treatment failure: DMBaseline: DM; Treatment failure: NR/NP
Efavirenz Once Daily + CBV (DB)60040000
Maraviroc Once Daily + CBV (DB), Then Twice Daily + CBV (OL)50350050
Maraviroc Twice Daily + CBV (DB)1101051240

[back to top]

Number of Participants Per Tropism Status at Baseline and at the Time of Treatment Failure Through Week 96

Number of participants per tropism status (R5, X4, DM, or NR/NP) at baseline and time of treatment failure analyzed through week 96 visit. Treatment failure defined as insufficient clinical response. Tropism result was censored for participants with viral load <500 copies/mL at time of treatment failure categorized as BLQ. The assessment for time of treatment failure was defined as last on treatment assessment. (NCT00098293)
Timeframe: Baseline, time of failure through Week 96

,,
Interventionparticipants (Number)
Baseline: R5; Treatment failure: R5Baseline: R5; Treatment failure: X4Baseline: R5; Treatment failure: DMBaseline: R5; Treatment failure: NR/NPBaseline: DM; Treatment failure: R5Baseline: DM; Treatment failure: X4Baseline: DM; Treatment failure: DMBaseline: DM; Treatment failure: NR/NPBaseline: NR/NP; Treatment failure: R5Baseline: NR/NP; Treatment failure: X4Baseline: NR/NP; Treatment failure: DMBaseline: NR/NP; Treatment failure: NR/NP
Efavirenz Once Daily + CBV (DB)1000510000000
Maraviroc Once Daily + CBV (DB), Then Twice Daily + CBV (OL)904600500000
Maraviroc Twice Daily + CBV (DB)14111712401001

[back to top]

Number of Participants With NRTI Associated Mutations at Time of Treatment Failure Through Week 48 and 96

Genotypic resistance to NRTIs was assessed by identification of relevant mutations at screening using MBPSGT assay and repeated for all participants with HIV-1 viral load more than 500 copies/mL at treatment failure through week 48 and week 96. Following mutations associated with NRTIs were summarized at time of failure: Any zidovudine/lamivudine (Zid/Lam), Any thymidine analogue-associated mutation (TAM), methionine (M) to valine/isoleucine (V/I) substitution at residue (r) 184 (M184V/I), lysine (K) to arginine (R) substitution at residue 65 (K65R) and any other NRTI mutations. (NCT00098293)
Timeframe: Screening, time of failure through Week 48, Week 96

,,
Interventionparticipants (Number)
Any Zid/Lam Mutation: Week 48 (n= 20, 43, 15)Any TAM Mutation: Week 48 (n= 20, 43, 15)K65R Mutation: Week 48 (n= 20, 43, 15)M184V/I Mutation: Week 48 (n= 20, 43, 15)Other NRTI Mutation: Week 48 (n= 20, 43, 15)Any Zid/Lam Mutation: Week 96 (n= 27, 55, 23)Any TAM Mutation: Week 96 (n= 27, 55, 23)K65R Mutation: Week 96 (n= 27, 55, 23)M184V/I Mutation: Week 96 (n= 27, 55, 23)Other NRTI Mutation: Week 96 (n= 27, 55, 23)
Efavirenz Once Daily + CBV (DB)3003082080
Maraviroc Once Daily + CBV (DB), Then Twice Daily + CBV (OL)14101402030200
Maraviroc Twice Daily + CBV (DB)27612713361331

[back to top]

Percentage of Participants With Viral Load of Less Than 400 Copies/Milliliter [Copies/mL] and Less Than 50 Copies/mL of Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA) at Week 48 for Full Analysis Set (FAS) Population

(NCT00098293)
Timeframe: Week 48

,,
InterventionPercentage of participants (Number)
Less than 400 copies/mLLess than 50 copies/mL
Efavirenz Once Daily + CBV (DB)73.169.3
Maraviroc Once Daily + CBV (DB), Then Twice Daily + CBV (OL)61.555.8
Maraviroc Twice Daily + CBV (DB)70.665.3

[back to top]

Time-Averaged Difference (TAD) in log10-transformed HIV-1 RNA Levels

TAD from baseline was calculated as area under the curve (AUC) of HIV-1 RNA load (log10 copies/mL) divided by time period minus baseline HIV-1 RNA load (log10 copies/mL). Baseline value calculated as average of pre-dose measurements collected at screening, randomization, and immediately pre-dose. Data not analyzed for participants originally randomized to maraviroc once daily arm since after termination, focus was shifted from efficacy and safety to only safety as reflected in the abbreviated set of efficacy measures noted in the amended planned analysis. (NCT00098293)
Timeframe: Baseline up to Week 48 and Week 96

,
Interventionlog10 copies/mL (Least Squares Mean)
Week 48Week 96
Efavirenz Once Daily + CBV (DB)-2.262-2.034
Maraviroc Twice Daily + CBV (DB)-2.152-1.945

[back to top]

Time to Virologic Failure

Time to virologic failure based on observed HIV-1 RNA levels and failure events (death;permanent discontinuation of drug;lost to follow-up [LTFU];new anti-retroviral drug added [except background drug change to drug of same class];or on open label for early non-response or rebound). Failure:at Time 0 if level not <400 copies/mL(2 consecutive visits) before events or last available visit;at time of earliest event if level <400 copies/mL(2 consecutive visits);failure if level >=400 copies/mL(2 consecutive visits) or 1 visit >=400 copies/mL followed by permanent discontinuation of drug or LTFU. (NCT00098293)
Timeframe: Week 48, Week 96

,
Interventiondays (Median)
Week 48Week 96
Efavirenz Once Daily + CBV (DB)NANA
Maraviroc Twice Daily + CBV (DB)NANA

[back to top]

Percentage of Participants With Viral Load of Less Than 400 Copies/mL and Less Than 50 Copies/mL of HIV-1 RNA at Week 96

(NCT00098293)
Timeframe: Week 96

,,
InterventionPercentage of participants (Number)
Less than 400 copies/mLLess than 50 copies/mL
Efavirenz Once Daily + CBV (DB)64.562.6
Maraviroc Once Daily + CBV (DB), Then Twice Daily + CBV (OL)52.948.3
Maraviroc Twice Daily + CBV (DB)61.456.9

[back to top]

Percentage of Participants With Viral Load of Less Than 400 Copies/mL and Less Than 50 Copies/mL of HIV-1 RNA at Week 48 for Per Protocol (PP) Population

Percentage of participants with viral load of less than 400 copies/mL and less than 50 copies/mL of HIV-1 RNA were not analyzed for participants originally randomized to maraviroc once daily arm since after termination, focus was shifted from efficacy and safety to only safety as reflected in the abbreviated set of efficacy measures noted in the amended planned analysis. (NCT00098293)
Timeframe: Week 48

,
InterventionPercentage of participants (Number)
Less than 400 copies/mLLess than 50 copies/mL
Efavirenz Once Daily + CBV (DB)78.2774.44
Maraviroc Twice Daily + CBV (DB)75.0070.00

[back to top]

Percentage of Participants With HIV-1 RNA Levels of Less Than 400 Copies/mL and Less Than 50 Copies/mL at Week 96 for Enhanced Sensitivity Trofile Assay (ESTA) R5 Participants

Percentage of participants with HIV-1 RNA levels of less than 400 copies/mL and less than 50 copies/mL were not analyzed for maraviroc once daily, then twice daily arm in order to avoid misinterpretation due to possible bias due to the fact that only a non-random sample of participants in the terminated arm were re-assayed with ESTA. (NCT00098293)
Timeframe: Week 96

,
InterventionPercentage of participants (Number)
Less than 400 copies/mLLess than 50 copies/mL
Efavirenz Once Daily + CBV (DB)64.462.7
Maraviroc Twice Daily + CBV (DB)64.058.8

[back to top]

Number of Participants With Efavirenz Associated Mutations at Time of Treatment Failure Through Week 48 and 96

Genotypic resistance: mutations at screening by MBPSGT assay, repeated if viral load >500 copies/mL at treatment failure through week 48, 96. Efavirenz mutation:lysine to aspargine at r103(K103N);tyrosine to cysteine/isoleucine at r181(Y181C/I);tyrosine to cysteine/leucine/histidine at r188(Y188C/L/H);glycine to alanine/serine at r190(G190A/S);valine to alanine to r106(V106A);leucine to isoleucine at r100(L100I);alanine to glycine at r98(A98G);lysine to glutamic acid at r101(K101E);valine to isoleucine at r108(V108I);proline to histidine at r225(P225H);methionine to leucine at r230(M230L). (NCT00098293)
Timeframe: Screening, time of failure through Week 48, Week 96

,
Interventionparticipants (Number)
L100I Mutation: Week 48 (n= 43, 15)K103N Mutation: Week 48 (n= 43, 15)V106M Mutation: Week 48 (n= 43, 15)V108I Mutation: Week 48 (n= 43, 15)Y181C/I Mutation: Week 48 (n= 43, 15)Y188L Mutation: Week 48 (n= 43, 15)G190S/A Mutation: Week 48 (n= 43, 15)P225H Mutation: Week 48 (n= 43, 15)L100I Mutation: Week 96 (n= 55, 23)K103N Mutation: Week 96 (n= 55, 23)V106M Mutation: Week 96 (n= 55, 23)V108I Mutation: Week 96 (n= 55, 23)Y181C/I Mutation: Week 96 (n= 55, 23)Y188L Mutation: Week 96 (n= 55, 23)G190S/A Mutation: Week 96 (n= 55, 23)P225H Mutation: Week 96 (n= 55, 23)
Efavirenz Once Daily + CBV (DB)06000010012110021
Maraviroc Twice Daily + CBV (DB)0001000000010000

[back to top]

Percentage of Participants With HIV-1 RNA Levels of Less Than 400 Copies/mL and Less Than 50 Copies/mL at Week 96 Analyzed Using Logistic Regression

(NCT00098293)
Timeframe: Week 96

,,
InterventionPercentage of participants (Number)
Less than 400 copies/mLLess than 50 copies/mL
Efavirenz Once Daily + CBV (DB)64.562.6
Maraviroc Once Daily + CBV (DB), Then Twice Daily + CBV (OL)52.948.3
Maraviroc Twice Daily + CBV (DB)61.456.9

[back to top]

Percentage of Participants With HIV-1 RNA Levels of Less Than 400 Copies/mL and Less Than 50 Copies/mL at Week 48 for Enhanced Sensitivity Trofile Assay (ESTA) R5 Participants

Percentage of participants with HIV-1 RNA levels of less than 400 copies/mL and less than 50 copies/mL were not analyzed for maraviroc once daily, then twice daily arm in order to avoid misinterpretation due to possible bias due to the fact that only a non-random sample of participants in the terminated arm were re-assayed with ESTA. (NCT00098293)
Timeframe: Week 48

,
InterventionPercentage of participants (Number)
Less than 400 copies/mLLess than 50 copies/mL
Efavirenz Once Daily + CBV (DB)72.368.3
Maraviroc Twice Daily + CBV (DB)73.368.5

[back to top]

Change From Baseline in Log 10-transformed Plasma Viral Load (HIV-1 RNA) Levels at Week 48 and 96

Change from baseline in log 10-transformed plasma viral load (HIV-1 RNA) levels (log10 copies/mL). Baseline value calculated as average of pre-dose measurements collected at screening, randomization, and immediately pre-dose. (NCT00098293)
Timeframe: Baseline, Week 48, Week 96

,,
Interventionlog10 copies/mL (Mean)
BaselineChange at Week 48Change at Week 96
Efavirenz Once Daily + CBV (DB)4.857-2.347-2.053
Maraviroc Once Daily + CBV (DB), Then Twice Daily + CBV (OL)4.899-2.665-2.565
Maraviroc Twice Daily + CBV (DB)4.851-2.240-1.961

[back to top]

Number of Participants With Phenotypic Resistance at Time of Treatment Failure Through Week 48 and 96

Phenotypic resistance to nucleoside reverse transcriptase inhibitors (NRTIs) and non-nucleoside reverse transcriptase inhibitors (NNRTIs) assessed at screening by Monogram Bioscience PhenoSense genotype (MBPSGT) assay, repeated if viral load >500 copies/mL at treatment failure through week 48, 96. Phenotypic resistance to maraviroc was assumed in maraviroc treatment failures with X4-using virus and in R5 maraviroc treatment failures using Monogram Bioscience PhenoSense Entry Assay. Phenotypic resistance to zidovudine, lamivudine, efavirenz and maraviroc at time of failure was summarized. (NCT00098293)
Timeframe: Screening, time of failure through Week 48, Week 96

,,
Interventionparticipants (Number)
Resistance to Zidovudine: Week 48 (n= 20, 43, 15)Resistance to Lamivudine: Week 48 (n= 20, 43, 15)Resistance to Efavirenz: Week 48 (n= 20, 43, 15)Resistance to Maraviroc: Week 48 (n= 20, 43, 15)Resistance to Zidovudine: Week 96 (n= 27, 55, 23)Resistance to Lamivudine: Week 96 (n= 27, 55, 23)Resistance to Efavirenz: Week 96 (n= 27, 55, 23)Resistance to Maraviroc: Week 96 (n= 27, 55, 23)
Efavirenz Once Daily + CBV (DB)037NA0813NA
Maraviroc Once Daily + CBV (DB), Then Twice Daily + CBV (OL)0140NA0200NA
Maraviroc Twice Daily + CBV (DB)0270120330NA

[back to top]

Percentage of Participants With HIV-1 RNA Levels of Less Than 400 Copies/mL and Less Than 50 Copies/mL at Week 48 Analyzed Using Logistic Regression

(NCT00098293)
Timeframe: Week 48

,,
InterventionPercentage of participants (Number)
Less than 400 copies/mLLess than 50 copies/mL
Efavirenz Once Daily + CBV (DB)73.169.3
Maraviroc Once Daily + CBV (DB), Then Twice Daily + CBV (OL)61.555.8
Maraviroc Twice Daily + CBV (DB)70.665.3

[back to top]

Change From Baseline in Lymphocyte Cluster of Differentiation 4 (CD4) Count at Week 48 and 96

Baseline value calculated as the average of pre-dose measurements collected at screening and immediately pre-dose. (NCT00098293)
Timeframe: Baseline, Week 48, Week 96

,,
Interventioncells per microliter (cells/µL) (Mean)
BaselineChange at Week 48Change at Week 96
Efavirenz Once Daily + CBV (DB)271.87143.52171.50
Maraviroc Once Daily + CBV (DB), Then Twice Daily + CBV (OL)274.1172.50183.75
Maraviroc Twice Daily + CBV (DB)264.70169.53206.31

[back to top]

Time to Virological Failure

Time to virologic failure based on observed HIV-1 RNA levels and failure events (death;permanent discontinuation of drug;lost to follow-up [LTFU];new anti-retroviral drug added [except background drug change to drug of same class];or on open label for early non-response or rebound). Failure:at Time 0 if level not <400 copies/mL(2 consecutive visits) before events or last available visit;at time of earliest event if level <400 copies/mL(2 consecutive visits);failure if level >=400 copies/mL(2 consecutive visits) or 1 visit >=400 copies/mL followed by permanent discontinuation of drug or LTFU. (NCT00098306)
Timeframe: Week 48

Interventiondays (Median)
Maraviroc QD344.00
Maraviroc BIDNA
Placebo0.00

[back to top]

Change From Baseline in CD4 Cell Count at Week 24 and 48

Change from baseline in CD4 cell count measured as cells/µL. Baseline value calculated as the average of pre-dose measurements collected at screening and immediately pre-dose. (NCT00098306)
Timeframe: Baseline, Week 24 and 48

,,
Interventioncells/µL (Least Squares Mean)
Week 24Week 48
Maraviroc BID111.08122.44
Maraviroc QD106.63112.53
Placebo52.1453.97

[back to top]

Change From Baseline in CD8 Cell Count at Week 24 and 48

Change from baseline in CD8 cell count measured as cells/µL. Baseline value calculated as the average of pre-dose measurements collected at screening and immediately pre-dose. (NCT00098306)
Timeframe: Baseline, Week 24 and 48

,,
Interventioncells/µL (Least Squares Mean)
Week 24Week 48
Maraviroc BID302.33220.40
Maraviroc QD283.48198.00
Placebo-0.74-15.34

[back to top]

Number of Participants With Change in GSS and PSS From Screening at the Time of Treatment Failure Through Week 24

Number of participants with GSS and PSS were used as surrogates for assessing genotype and phenotype. Genotypic and phenotypic resistance to PIs, NRTIs and NNRTIs were evaluated at screening and time of treatment failure analyzed through week 24 visit, by Monogram Biosciences PhenoSense GT assay. Score was determined for each drug in OBT, giving 1: drug 'sensitive'/'susceptible' and 0: 'resistant'. GSS and PSS score range:0 to >3. Genotypic enfuvirtide value was used for PSS, no phenotypic enfuvirtide was recorded. (NCT00098306)
Timeframe: Screening and time of failure through week 24

,,
Interventionparticipants (Number)
Change in GSS by less than -3 (n= 35 ,40, 47)Change in GSS by -3 (n= 35 ,40, 47)Change in GSS by -2 (n= 35 ,40, 47)Change in GSS by -1 (n= 35 ,40, 47)Change in GSS by 0 (n= 35 ,40, 47)Change in GSS by 1 (n= 35 ,40, 47)Change in GSS by 2 (n= 35 ,40, 47)Change in GSS by 3 (n= 35 ,40, 47)Change in GSS by greater than 3 (n= 35 ,40, 47)Change in PSS by less than -3 (n= 35 ,40, 45)Change in PSS by -3 (n= 35 ,40, 45)Change in PSS by -2 (n= 35 ,40, 45)Change in PSS by -1 (n= 35 ,40, 45)Change in PSS by 0 (n= 35 ,40, 45)Change in PSS by 1 (n= 35 ,40, 45)Change in PSS by 2 (n= 35 ,40, 45)Change in PSS by 3 (n= 35 ,40, 45)Change in PSS by greater than 3 (n= 35 ,40, 45)
Maraviroc BID0001326100001118173000
Maraviroc QD003923000004410152000
Placebo1011132101011412252000

[back to top]

Cluster of Differentiation 4 (CD4) and Cluster of Differentiation 8 (CD8) Cell Count at Baseline

Baseline value calculated as average of pre-dose measurements collected at screening and immediately pre-dose. (NCT00098306)
Timeframe: Baseline

,,
Interventioncells per microliter (cells/µL) (Mean)
CD4CD8
Maraviroc BID176.5880.1
Maraviroc QD187.5941.1
Placebo178.5880.4

[back to top]

Number of Participants Per Tropism Status at Baseline and at the Time of Treatment Failure Through Week 24

Number of participants per tropism status (C-X-C chemokine receptor 5 {CCR5} [R5], C-X-C chemokine receptor type 4 {CXCR4} [X4], Dual/mixed [DM], or Non-reportable/Non-phenotypable [NR/NP]) at baseline and time of treatment failure analyzed through week 24 visit. Treatment failure defined as discontinuation due to insufficient clinical response. HIV-1 RNA viral load <500 copies/mL categorized as below lower limit of quantification (BLQ). The assessment for time of treatment failure was defined as the last on treatment assessment. (NCT00098306)
Timeframe: Baseline and time of failure through week 24

,,
Interventionparticipants (Number)
Baseline: R5; Treatment failure: R5Baseline: R5; Treatment failure: X4Baseline: R5; Treatment failure: DMBaseline: R5; Treatment failure: NR/NPBaseline: DM; Treatment failure: R5Baseline: DM; Treatment failure: X4Baseline: DM; Treatment failure: DMBaseline: DM; Treatment failure: NR/NP
Maraviroc BID1051930441
Maraviroc QD1061810131
Placebo440131110

[back to top]

Number of Participants Per Tropism Status at Baseline and at the Time of Treatment Failure Through Week 48

Number of participants per tropism status (R5, X4, DM, or NR/NP) at baseline and time of treatment failure analyzed through week 48 visit. Treatment failure defined as insufficient clinical response. HIV-1 RNA viral load <500 copies/mL categorized as BLQ. The assessment for time of treatment failure was defined as the last on treatment assessment. (NCT00098306)
Timeframe: Baseline and time of failure through week 48

,,
Interventionparticipants (Number)
Baseline: R5, Treatment failure: R5Baseline: R5, Treatment failure: X4Baseline: R5, Treatment failure: DMBaseline: R5, Treatment failure: NR/NPBaseline: DM, Treatment failure: R5Baseline: DM, Treatment failure: X4Baseline: DM, Treatment failure: DMBaseline: DM, Treatment failure: NR/NP
Maraviroc BID1572230451
Maraviroc QD1661910242
Placebo490332110

[back to top]

Number of Participants With Change in GSS and PSS From Screening at the Time of Treatment Failure at Week 48

Number of participants with GSS and PSS were used as surrogates for assessing genotype and phenotype. Genotypic and phenotypic resistance to PIs, NRTIs and NNRTIs were evaluated at screening and time of treatment failure analyzed through week 48 visit, by Monogram Biosciences PhenoSense GT assay. Score was determined for each drug in OBT, giving 1: drug 'sensitive'/'susceptible' and 0: 'resistant'. GSS and PSS score range:0 to >3. Genotypic enfuvirtide value was used for PSS, no phenotypic enfuvirtide was recorded. (NCT00098306)
Timeframe: Screening and time of failure through week 48

,,
Interventionparticipants (Number)
Change in GSS by less than -3 (n= 44 ,52, 55)Change in GSS by -3 (n= 44 ,52, 55)Change in GSS by -2 (n= 44 ,52, 55)Change in GSS by -1 (n= 44 ,52, 55)Change in GSS by 0 (n= 44 ,52, 55)Change in GSS by 1 (n= 44 ,52, 55)Change in GSS by 2 (n= 44 ,52, 55)Change in GSS by 3 (n= 44 ,52, 55)Change in GSS by greater than 3 (n= 44 ,52, 55)Change in PSS by less than -3 (n= 44 , 52, 53)Change in PSS by -3 (n= 44 , 52, 53)Change in PSS by -2 (n= 44 , 52, 53)Change in PSS by -1 (n= 44 , 52, 53)Change in PSS by 0 (n= 44 , 52, 53)Change in PSS by 1 (n= 44 , 52, 53)Change in PSS by 2 (n= 44 , 52, 53)Change in PSS by 3 (n= 44 , 52, 53)Change in PSS by greater than 3 (n= 44 , 52, 53)
Maraviroc BID0011731210011321233000
Maraviroc QD0051226100004613173010
Placebo1121533111013612264100

[back to top]

Change From Baseline in Log 10-transformed HIV-1 RNA Levels at Week 24

Change from baseline in log 10-transformed plasma viral load (HIV-1 RNA) levels (log10 copies/mL). Baseline value calculated as average of pre-dose measurements collected at screening, randomization, and immediately pre-dose. (NCT00098306)
Timeframe: Baseline and Week 24

Interventionlog10 copies/mL (Least Squares Mean)
Maraviroc QD-1.818
Maraviroc BID-1.952
Placebo-1.030

[back to top]

Number of Participants With Genotypic Susceptibility Scores (GSS) and Phenotypic Susceptibility Score (PSS) at Screening

Number of participants with GSS and PSS were used as surrogates for assessing genotype and phenotype. Genotypic and phenotypic resistance to protease inhibitors(PIs), nucleoside reverse transcriptase inhibitors(NRTIs), non-nucleoside reverse transcriptase inhibitors(NNRTIs) were evaluated at screening (not at baseline), by Monogram Biosciences PhenoSense genotyping (GT) assay. Score was determined for each drug in OBT, giving 1:drug 'sensitive'/'susceptible' and 0:'resistant'. GSS and PSS score range:0 to >3. Genotypic enfuvirtide value was used for PSS, no phenotypic enfuvirtide was recorded. (NCT00098306)
Timeframe: Screening

,,
Interventionparticipants (Number)
GSS: 0GSS: 1GSS: 2GSS: greater than or equal to 3GSS: missingPSS: 0PSS: 1PSS: 2PSS: greater than or equal to 3PSS: missing
Maraviroc BID598048471247369663
Maraviroc QD528238573257051833
Placebo312921343171835453

[back to top]

Change From Baseline in Viral Load at Week 24 and Week 48 by Overall Susceptibility Score (OSS) at Screening

Association between baseline resistance and virological response was assessed as change in viral load by OSS at screening. OSS categorized as 0, 1, 2, >3 (maximum value of 6) and calculated as the sum of the net assessment of in-vitro phenotypic and genotypic susceptibility using a binary scoring system (0= resistant, 1= sensitive or susceptible) for each antiretroviral agent in OBT. Higher scores indicate greater susceptibility. Baseline value is the average of the values from screening, randomization and immediately pre-dose. (NCT00098306)
Timeframe: Baseline, Week 24 and Week 48

,,
Interventionlog10copies/mL (Mean)
Week 24; Screening OSS 0 (n= 29, 27, 19)Week 24; Screening OSS 1 (n= 76, 86, 21)Week 24; Screening OSS 2 (n= 51, 65, 38)Week 24; Screening OSS >=3 (n= 68, 53, 35)Week 24; Screening OSS= Missing (n= 4, 3, 3)Week 48; Screening OSS 0 (n= 29, 27, 19)Week 48; Screening OSS 1 (n= 77, 86, 20)Week 48; Screening OSS 2 (n= 51, 64, 38)Week 48; Screening OSS >=3 (n= 67, 54, 36)Week 48; Screening OSS= Missing (n= 4, 3, 3)
Maraviroc BID-1.524-1.895-2.220-2.672-3.075-1.419-1.868-2.182-2.634-2.698
Maraviroc QD-0.945-1.911-2.093-2.536-2.070-0.876-1.843-2.146-2.427-2.106
Placebo-0.083-0.350-1.251-2.471-0.530-0.088-0.262-1.147-2.205-0.370

[back to top]

Percentage of Participants With HIV-1 RNA Levels Less Than 400 Copies/mL or With at Least 0.5 log10 Decrease From Baseline

(NCT00098306)
Timeframe: Week 24 and 48

,,
Interventionpercentage of participants (Number)
Week 24Week 48
Maraviroc BID69.464.3
Maraviroc QD67.759.9
Placebo45.835.6

[back to top]

Percentage of Participants With HIV-1 RNA Levels Less Than 400 Copies/mL or With at Least 1.0 log10 Decrease From Baseline

(NCT00098306)
Timeframe: Week 24 and 48

,,
Interventionpercentage of participants (Number)
Week 24Week 48
Maraviroc BID67.763.0
Maraviroc QD64.757.8
Placebo38.131.4

[back to top]

Percentage of Participants With HIV-1 RNA Levels Less Than 50 Copies/mL

(NCT00098306)
Timeframe: Week 24 and 48

,,
Interventionpercentage of participants (Number)
Week 24Week 48
Maraviroc BID48.546.8
Maraviroc QD42.241.8
Placebo24.616.1

[back to top]

Time-Averaged Difference (TAD) From Baseline in log10 Transformed HIV-1 RNA Levels

TAD from baseline was calculated as area under the curve of HIV-1 RNA load (log10 copies/mL) divided by time period minus baseline HIV-1 RNA load (log10 copies/mL). Baseline value calculated as the average of pre-dose measurements collected at screening, randomization, and immediately pre-dose. (NCT00098306)
Timeframe: Baseline to Week 24 and Week 48

,,
Interventionlog10 copies/mL (Least Squares Mean)
Week 24Week 48
Maraviroc BID-1.741-1.720
Maraviroc QD-1.636-1.556
Placebo-0.939-0.788

[back to top]

Change From Baseline in Log 10-transformed HIV-1 RNA Levels at Week 48

Change from baseline in log 10-transformed plasma viral load (HIV-1 RNA) levels (log10 copies/mL). Baseline value calculated as average of pre-dose measurements collected at screening, randomization and immediately pre-dose. (NCT00098306)
Timeframe: Baseline and Week 48

Interventionlog10 copies/mL (Least Squares Mean)
Maraviroc QD-1.656
Maraviroc BID-1.824
Placebo-0.803

[back to top]

Log 10-transformed Human Immunodeficiency Virus Ribonucleic Acid (HIV-1 RNA) Levels at Baseline

Baseline value calculated as average of pre-dose measurements collected at screening, randomization, and immediately pre-dose. (NCT00098306)
Timeframe: Baseline

Interventionlog10 copies/milliliter(log10 copies/mL) (Mean)
Maraviroc QD4.854
Maraviroc BID4.861
Placebo4.840

[back to top]

Percentage of Participants With HIV-1 RNA Levels Less Than 400 Copies/mL

(NCT00098306)
Timeframe: Week 24 and 48

,,
Interventionpercentage of participants (Number)
Week 24Week 48
Maraviroc BID60.457.5
Maraviroc QD54.750.9
Placebo31.422.0

[back to top]

Change From Baseline in CD8 Cell Count at Week 24 and 48

Change from baseline in CD8 cell count measured as cells/µL. Baseline value calculated as average of pre-dose measurements collected at screening and immediately pre-dose. (NCT00098722)
Timeframe: Week 24 and 48

,,
Interventioncells/μL (Least Squares Mean)
Week 24Week 48
Maraviroc BID255.40244.75
Maraviroc QD340.74241.43
Placebo122.18104.51

[back to top]

Percentage of Participants With HIV-1 RNA Levels Less Than 50 Copies/mL

(NCT00098722)
Timeframe: Week 24 and 48

,,
Interventionpercentage of participants (Number)
Week 24Week 48
Maraviroc BID40.8444.50
Maraviroc QD45.6045.05
Placebo20.8817.58

[back to top]

Time-Averaged Difference (TAD) From Baseline in log10 Transformed HIV-1 RNA Levels

TAD from baseline was calculated as area under the curve of HIV-1 RNA load (log10 copies/mL) divided by time period minus baseline HIV-1 RNA load (log10 copies/mL). Baseline value calculated as the average of pre-dose measurements collected at screening, randomization, and immediately pre-dose. (NCT00098722)
Timeframe: Baseline to Week 24 and Week 48

,,
Interventionlog10 copies/mL (Least Squares Mean)
Week 24Week 48
Maraviroc BID-1.755-1.781
Maraviroc QD-1.748-1.603
Placebo-0.872-0.748

[back to top]

Percentage of Participants With HIV-1 RNA Levels Less Than 400 Copies/ml or With at Least 0.5 log10 Decrease From Baseline

(NCT00098722)
Timeframe: Week 24 and 48

,,
Interventionpercentage of participants (Number)
Week 24Week 48
Maraviroc BID72.2565.97
Maraviroc QD69.7860.99
Placebo37.3631.87

[back to top]

Percentage of Participants With HIV-1 RNA Levels Less Than 400 Copies/mL

(NCT00098722)
Timeframe: Week 24 and 48

,,
Interventionpercentage of participants (Number)
Week 24Week 48
Maraviroc BID61.2654.97
Maraviroc QD55.4952.75
Placebo23.0823.08

[back to top]

Number of Participants With Genotypic Susceptibility Score (GSS) and Phenotypic Susceptibility Score (PSS) at Screening

Number of participants with GSS and PSS were used as surrogates for genotype and phenotype. Genotypic and phenotypic resistance to protease inhibitors(PIs), nucleoside reverse transcriptase inhibitors(NRTIs) and non-nucleoside reverse transcriptase inhibitors(NNRTIs) were evaluated at screening (not at baseline), by Monogram Biosciences PhenoSense genotyping (GT) assay. Score was determined for each drug in OBT, giving 1:drug 'sensitive'/'susceptible' and 0:'resistant'. GSS and PSS score range:0 to >3. Genotypic enfuvirtide value was used for PSS, no phenotypic enfuvirtide was recorded. (NCT00098722)
Timeframe: Screening

,,
Interventionparticipants (Number)
GSS: 0GSS: 1GSS: 2GSS: greater than or equal to 3GSS: missingPSS: 0PSS: 1PSS: 2PSS: greater than or equal to 3PSS: missing
Maraviroc BID435832571264238841
Maraviroc QD396425522204642713
Placebo202420252122023342

[back to top]

Number of Participants With Change in GSS and PSS From Screening at the Time of Treatment Failure Through Week 48

Number of participants with GSS and PSS were used as surrogates for genotype and phenotype. Genotypic and phenotypic resistance to PIs, NRTIs, NNRTIs were evaluated at screening and time of treatment failure analyzed through Week 48 visit, by Monogram Biosciences PhenoSense GT assay. Score was determined for each drug in OBT, giving 1:drug 'sensitive'/'susceptible' and 0:'resistant'. GSS and PSS score range:0 to >3. Genotypic enfuvirtide value was used for PSS, no phenotypic enfuvirtide was recorded. (NCT00098722)
Timeframe: Screening and time of failure through Week 48

,,
Interventionparticipants (Number)
Change in GSS by less than -3 (n= 35, 33, 46)Change in GSS by -3 (n= 35, 33, 46)Change in GSS by -2 (n= 35, 33, 46)Change in GSS by -1 (n= 35, 33, 46)Change in GSS by 0 (n= 35, 33, 46)Change in GSS by 1 (n= 35, 33, 46)Change in GSS by 2 (n= 35, 33, 46)Change in GSS by 3 (n= 35, 33, 46)Change in GSS by greater than 3 (n= 35, 33, 46)Change in PSS by less than -3 (n= 35, 32, 46)Change in PSS by -3 (n= 35, 32, 46)Change in PSS by -2 (n= 35, 32, 46)Change in PSS by -1 (n= 35, 32, 46)Change in PSS by 0 (n= 35, 32, 46)Change in PSS by 1 (n= 35, 32, 46)Change in PSS by 2 (n= 35, 32, 46)Change in PSS by 3 (n= 35, 32, 46)Change in PSS by greater than 3 (n= 35, 32, 46)
Maraviroc BID10062240000039190100
Maraviroc QD0001223000001313162000
Placebo0021724210003418192000

[back to top]

Number of Participants With Change in GSS and PSS From Screening at the Time of Treatment Failure Through Week 24

Number of participants with GSS and PSS were used as surrogates for genotype and phenotype. Genotypic and phenotypic resistance to PIs, NRTIs, NNRTIs were evaluated at screening and time of treatment failure analyzed through Week 24 visit, by Monogram Biosciences PhenoSense GT assay. Score was determined for each drug in OBT, giving 1:drug 'sensitive'/'susceptible' and 0:'resistant'. GSS and PSS score range:0 to >3. Genotypic enfuvirtide value was used for PSS, no phenotypic enfuvirtide was recorded. (NCT00098722)
Timeframe: Screening and time of failure through Week 24

,,
Interventionparticipants (Number)
Change in GSS by less than -3 (n= 22, 25, 40)Change in GSS by -3 (n= 22, 25, 40)Change in GSS by -2 (n= 22, 25, 40)Change in GSS by -1 (n= 22, 25, 40)Change in GSS by 0 (n= 22, 25, 40)Change in GSS by 1 (n= 22, 25, 40)Change in GSS by 2 (n= 22, 25, 40)Change in GSS by 3 (n= 22, 25, 40)Change in GSS by greater than 3 (n= 22, 25, 40)Change in PSS by less than -3 (n= 22, 24, 40)Change in PSS by -3 (n= 22, 24, 40)Change in PSS by -2 (n= 22, 24, 40)Change in PSS by -1 (n= 22, 24, 40)Change in PSS by 0 (n= 22, 24, 40)Change in PSS by 1 (n= 22, 24, 40)Change in PSS by 2 (n= 22, 24, 40)Change in PSS by 3 (n= 22, 24, 40)Change in PSS by greater than 3 (n= 22, 24, 40)
Maraviroc BID10061530000038130000
Maraviroc QD00091300000128101000
Placebo0011522110002316181000

[back to top]

Number of Participants Per Tropism Status at Baseline and at the Time of Treatment Failure Through Week 48

Number of participants per tropism status R5, X4, DM, or NR/NP at baseline and time of failure analyzed through week 48 visit. Treatment failure defined as discontinuation due to insufficient clinical response. HIV-1 RNA viral load <500 copies/ml categorized as BLQ. The assessment for time of treatment failure was defined as the last on treatment assessment. (NCT00098722)
Timeframe: Baseline and time of failure through Week 48

,,
Interventionparticipants (Number)
Baseline: R5; Treatment failure: R5Baseline: R5; Treatment failure: X4Baseline: R5; Treatment failure: DMBaseline: R5; Treatment failure: NR/NPBaseline: DM; Treatment failure: R5Baseline: DM; Treatment failure: X4Baseline: DM; Treatment failure: DMBaseline: DM; Treatment failure: NR/NP
Maraviroc BID921060271
Maraviroc QD172861030
Placebo400323000

[back to top]

Number of Participants Per Tropism Status at Baseline and at the Time of Treatment Failure Through Week 24

Number of participants per tropism status (C-X-C chemokine receptor 5 {CCR5} [R5], C-X-C chemokine receptor type 4 {CXCR4} [X4], Dual/Mixed [DM], or Non-reportable/Non-phenotypable [NR/NP]) at baseline and time of treatment failure analyzed through week 24 visit. Treatment failure defined as discontinuation due to insufficient clinical response. HIV-1 RNA viral load <500 copies/ml categorized as below lower limit of quantification (BLQ). The assessment for time of treatment failure was defined as the last on treatment assessment. (NCT00098722)
Timeframe: Baseline and time of failure through Week 24

,,
Interventionparticipants (Number)
Baseline: R5; Treatment failure: R5Baseline: R5; Treatment failure: X4Baseline: R5; Treatment failure: DMBaseline: R5; Treatment failure: NR/NPBaseline: DM; Treatment failure: R5Baseline: DM; Treatment failure: X4Baseline: DM; Treatment failure: DMBaseline: DM; Treatment failure: NR/NP
Maraviroc BID72650271
Maraviroc QD82561030
Placebo360322000

[back to top]

Cluster of Differentiation 4 (CD4) and Cluster of Differentiation 8 (CD8) Cell Count at Baseline

Baseline value calculated as average of pre-dose measurements collected at screening and immediately pre-dose. (NCT00098722)
Timeframe: Baseline

,,
Interventioncells per microliter (cells/μL) (Mean)
CD4CD8
Maraviroc BID204.9996.9
Maraviroc QD206.0994.6
Placebo198.5952.3

[back to top]

Change From Baseline in Viral Load at Week 24 and Week 48 by Overall Susceptibility Score (OSS) at Screening

Association between baseline resistance and virological response was assessed as change in viral load by OSS at screening. OSS categorized as 0, 1, 2, >3 (maximum value of 6) and calculated as the sum of the net assessment of in-vitro phenotypic and genotypic susceptibility using a binary scoring system (0= resistant, 1= sensitive or susceptible) for each antiretroviral agent in OBT. Higher scores indicate greater susceptibility. Baseline value is the average of the values from screening, randomization and immediately pre-dose. (NCT00098722)
Timeframe: Baseline, Week 24 and Week 48

,,
Interventionlog10copies/mL (Mean)
Week 24; Screening OSS 0 (n= 22, 29, 16)Week 24; Screening OSS 1 (n= 54, 48, 23)Week 24; Screening OSS 2 (n= 37, 39, 21)Week 24; Screening OSS >=3 (n= 64, 68, 29)Week 24; Screening OSS= Missing (n= 3, 1, 2)Week 48; Screening OSS 0 (n= 22, 29, 16)Week 48; Screening OSS 1 (n= 54, 48, 23)Week 48; Screening OSS 2 (n= 38, 39, 21)Week 48; Screening OSS >=3 (n= 63, 68, 29)Week 48; Screening OSS= Missing (n= 3, 1, 2)
Maraviroc BID-1.231-2.051-2.515-2.530-0.544-1.290-1.899-2.565-2.426-0.833
Maraviroc QD-1.721-1.822-2.145-2.655-2.642-1.745-1.678-2.207-2.448-2.795
Placebo-0.264-0.651-0.611-2.109-1.169-0.249-0.594-0.591-1.942-1.105

[back to top]

Change From Baseline in CD4 Cell Count at Week 24 and 48

Change from baseline in CD4 cell count measured as cells/µL. Baseline value calculated as average of pre-dose measurements collected at screening and immediately pre-dose. (NCT00098722)
Timeframe: Week 24 and 48

,,
Interventioncells/μL (Least Squares Mean)
Week 24Week 48
Maraviroc BID101.89127.80
Maraviroc QD111.72121.49
Placebo63.7869.34

[back to top]

Time to Virological Failure

Time to virologic failure based on observed HIV-1 RNA levels and failure events (death;permanent discontinuation of drug;lost to follow-up[LTFU];new anti-retroviral drug added [except background drug change to drug of same class];or on open label for early non-response or rebound). Failure:at Time 0 if level not <400 copies/mL (2 consecutive visits) before events or last available visit;at time of earliest event if level <400 copies/mL (2 consecutive visits);failure if level >=400 copies/mL (2 consecutive visits) or 1 visit >=400 copies/mL followed by permanent discontinuation of drug or LTFU. (NCT00098722)
Timeframe: Week 48

Interventiondays (Median)
Maraviroc QD345.00
Maraviroc BID361.00
Placebo0.00

[back to top]

Log 10-transformed Human Immunodeficiency Virus Ribonucleic Acid (HIV-1 RNA) Levels at Baseline

Baseline value calculated as average of pre-dose measurements collected at screening, randomization, and immediately pre-dose. (NCT00098722)
Timeframe: Baseline

Interventionlog10 copies/milliliter(log10 copies/mL) (Mean)
Maraviroc QD4.873
Maraviroc BID4.840
Placebo4.886

[back to top]

Change From Baseline in Log 10-transformed HIV-1 RNA Levels at Week 48

Change from baseline in log 10-transformed plasma viral load (HIV-1 RNA) levels (log10 copies/mL). Baseline value calculated as average of pre-dose measurements collected at screening, randomization, and immediately pre-dose. (NCT00098722)
Timeframe: Baseline and Week 48

Interventionlog10 copies/mL (Least Squares Mean)
Maraviroc QD-1.718
Maraviroc BID-1.865
Placebo-0.757

[back to top]

Change From Baseline in Log 10-transformed HIV-1 RNA Levels at Week 24

Change from baseline in log 10-transformed plasma viral load (HIV-1 RNA) levels (log10 copies/mL). Baseline value calculated as average of pre-dose measurements collected at screening, randomization, and immediately pre-dose. (NCT00098722)
Timeframe: Baseline and Week 24

Interventionlog10 copies/mL (Least Squares Mean)
Maraviroc QD-1.950
Maraviroc BID-1.971
Placebo-0.929

[back to top]

Percentage of Participants With HIV-1 RNA Levels Less Than 400 Copies/ml or With at Least 1.0 log10 Decrease From Baseline

(NCT00098722)
Timeframe: Week 24 and 48

,,
Interventionpercentage of participants (Number)
Week 24Week 48
Maraviroc BID69.6363.35
Maraviroc QD66.4858.79
Placebo30.7727.47

[back to top]

Number of Subjects With HIV-1 RNA Levels < 400 Copies/mL or at Least 0.5 Log 10-transformed Decrease From Baseline in HIV-1 RNA Levels

Number of subjects with HIV-1 RNA levels < 400 copies/mL or at least 0.5 log 10-transformed decrease from baseline in HIV-1 RNA levels. Baseline value calculated as average of pre-dose measurements collected at screening, randomization, and baseline visits. (NCT00098748)
Timeframe: Baseline, Week 24, Week 48

,,
Interventionparticipants (Number)
Week 24Week 48
Maraviroc BID2522
Maraviroc QD2414
Placebo2318

[back to top]

Number of Subjects With HIV-1 RNA Levels < 50 Copies/mL

(NCT00098748)
Timeframe: Baseline, Week 24, Week 48

,,
Interventionparticipants (Number)
Week 24Week 48
Maraviroc BID1414
Maraviroc QD1210
Placebo913

[back to top]

Number of Subjects With HIV-1 RNA Levels < 400 Copies/mL or at Least 1.0 Log 10-transformed Decrease From Baseline in HIV-1 RNA Levels

Number of subjects with HIV-1 RNA levels < 400 copies/mL or at least 1.0 log 10-transformed decrease from baseline in HIV-1 RNA levels. Baseline value calculated as average of pre-dose measurements collected at screening, randomization, and baseline visits. (NCT00098748)
Timeframe: Baseline, Week 24, Week 48

,,
Interventionparticipants (Number)
Week 24Week 48
Maraviroc BID2320
Maraviroc QD1813
Placebo2115

[back to top]

Change From Baseline in CD8 Cell Count

Change from baseline in CD8 cell count (measured as cells/µL). Baseline value calculated as the average of pre-dose measurements collected at screening, randomization, and baseline visits. (NCT00098748)
Timeframe: Baseline to Week 24 and Week 48

,,
Interventioncells/µL (Mean)
Week 24Week 48
Maraviroc BID322.683342.87
Maraviroc QD391.061351.23
Placebo154.293192.30

[back to top]

Number of Subjects With Treatment Failure at Week 24 by Overall Susceptibility Score (OSS) at Screening

Number of subjects for association between screening resistance and virologic response as determined by treatment failure and OSS at screening. OSS categorized as 0-1, 2-4, >4 (maximum value of 6) and calculated as the sum of the net assessment of in vitro phenotypic and genotypic susceptibility using a binary scoring system (0= reduced susceptibility, 1=susceptible) for each antiretroviral agent in OBT. Higher scores indicate greater susceptibility. (NCT00098748)
Timeframe: Screening, Week 24

,,
Interventionparticipants (Number)
Scr score: 0-1Scr score: 2-4Scr score: missing
Maraviroc BID12351
Maraviroc QD21351
Placebo17401

[back to top]

Time (50% Quartile Point Estimate) to Virologic Failure

Time to virologic failure based on observed HIV-1 RNA levels and failure events (death; permanent discontinuation of test drug [perm DC]; lost to follow-up [LTFU]; new anti-retroviral drug added (except background drug change to drug of same class); or on open label for early non-response or rebound). Failure: at Time 0 if level not <400 copies/mL (2 consecutive visits) before event(s) or last available visit; at time of earliest event if level <400 copies/mL (on 2 consecutive visits); failure if level ≥400 copies/mL (2 consecutive visits) or 1 visit ≥400 copies/mL followed by perm DC or LTFU. (NCT00098748)
Timeframe: Day 1 through Week 24 and through Week 48

,,
Interventiondays (Median)
Week 24 (n=35, 23, 29)Week 48 (n=45, 37, 44)
Maraviroc BID189.000.00
Maraviroc QD88.000.00
Placebo100.000.00

[back to top]

Number of Subjects With Treatment Failure at Week 48 by Overall Susceptibility Score (OSS) at Screening

Number of subjects for association between screening resistance and virologic response as determined by treatment failure and OSS at screening. OSS categorized as 0, 1, 2, or ≥3 (maximum value of 6) and calculated as the sum of the net assessment of in vitro phenotypic and genotypic susceptibility using a binary scoring system (0= reduced susceptibility, 1=susceptible) for each antiretroviral agent in OBT. Higher scores indicate greater susceptibility. (NCT00098748)
Timeframe: Screening, Week48

,,
Interventionparticpants (Number)
Scr score: 0Scr score: 1Scr score: 2Scr score: ≥3Scr score: missing
Maraviroc BID21114241
Maraviroc QD11921151
Placebo21513271

[back to top]

Number of Subjects With HIV-1 RNA Levels < 400 Copies/mL

(NCT00098748)
Timeframe: Week 24, Week 48

,,
Interventionparticipants (Number)
Week 24Week 48
Maraviroc BID1616
Maraviroc QD1413
Placebo1413

[back to top]

Number of Subjects With Acquired Immunodeficiency Syndrome (AIDS)-Defining Opportunistic Illnesses (Analysis at Week 48)

Number of subjects with AIDS-defining opportunistic illnesses based on investigator classification guided by a predefined list of clinical Category C Adverse Events per CDC HIV Classification System. Includes events occurring up to 7 days after last dose of study drug. (NCT00098748)
Timeframe: Baseline through Week 48

,,
Interventionparticipants (Number)
CandidiasisCytomegalovirus chorioretinitisHistoplasmosisOesophageal candidiasisPneumococcal SepsisPneumocystis jiroveci pneumoniaPneumoniaEncephalitis
Maraviroc BID10021010
Maraviroc QD01100300
Placebo10000001

[back to top]

Number of Subjects With Acquired Immunodeficiency Syndrome (AIDS)-Defining Opportunistic Illnesses (Analysis at Week 24)

Number of subjects with AIDS-defining opportunistic illnesses based on investigator classification guided by a predefined list of clinical Category C Adverse Events per Center for Disease Control (CDC) HIV Classification System. Includes events occurring up to 7 days after last dose of study drug. (NCT00098748)
Timeframe: Baseline through Week 24

,,
Interventionparticipants (Number)
CandidiasisCytomegalovirus chorioretinitisHerpes simplexHistoplasmosisMycobacterium avium complex infectionOesophageal candidiasisPneumocystis jiroveci pneumoniaPneumoniaEncephalitis
Maraviroc BID000002100
Maraviroc QD011110300
Placebo100000011

[back to top]

Number of Subjects Per Tropism Status at Screening and Time of Treatment Failure (Analysis at Week 48)

Number of subjects per Tropism status (CCR5 [R5], CXCR4 [X4], Dual Mixed [DM], or Non-reportable/Non-phenotypable [NR/NP]) at Screening (Scr) and at time of treatment failure (Tx fail). Treatment failure defined as insufficient clinical response. HIV-1 RNA viral load <500 copies/ml categorized as below lower limit of quantification (BLQ). Tropism may have been assessed at either the Screening or Baseline visit. The assessment for time of treatment failure is defined as the last on-treatment assessment. (NCT00098748)
Timeframe: Screening through Week 48

,,
Interventionparticipants (Number)
Scr: X4, Tx fail: X4Scr: X4, Tx fail: DMScr: DM, Tx fail: R5Scr: DM, Tx fail: X4Scr: DM, Tx fail: DMScr: DM, Tx fail: NR/NPScr: DM, Tx fail: BLQScr: NR/NP, Tx fail: NR/NP
Maraviroc BID1111210101
Maraviroc QD1111224010
Placebo105218100

[back to top]

Number of Subjects Per Tropism Status at Screening and at the Time of Treatment Failure (Analysis at Week 24)

Number of subjects per Tropism status (CCR5 [R5], CXCR4 [X4], Dual Mixed [DM], or Non-reportable/Non-phenotypable [NR/NP]) at Screening (Scr) and at time of treatment failure (Tx fail). Treatment failure defined as insufficient clinical response. HIV-1 RNA viral load <500 copies/ml categorized as below lower limit of quantification (BLQ). Tropism may have been assessed at either the Screening or Baseline visit. The assessment for time of treatment failure is defined as the last on-treatment assessment. (NCT00098748)
Timeframe: Screening through Week 24

,,
Interventionparticipants (Number)
Scr: X4, Tx fail: X4Scr: X4, Tx fail: DMScr: DM, Tx fail: R5Scr: DM, Tx fail: X4Scr: DM, Tx fail: DMScr DM, Tx fail: NR/NPScr DM, Tx fail: BLQScr NR/NP, Tx fail: NR/NP
Maraviroc BID110129001
Maraviroc QD1111219010
Placebo104216100

[back to top]

Change From Baseline in Time Averaged Difference (TAD) in log10 HIV-1 RNA

Change from baseline of TAD in log10 HIV-1 RNA viral load calculated as [AUC of HIV-1 RNA viral load (log10 copies/mL) / time period] - Baseline HIV-1 RNA viral load (log10 copies/mL). Baseline value calculated as the average of pre-dose measurements collected at screening, randomization, and baseline visits. (NCT00098748)
Timeframe: Baseline to Week 24 and Week 48

,,
Interventionlog10 copies/mL (Mean)
Week 24Week 48
Maraviroc BID-1.151-1.066
Maraviroc QD-0.850-0.561
Placebo-0.926-0.776

[back to top]

Change From Baseline in Human Immunodeficiency Virus (HIV-1) Viral Load (Ribonucleic Acid [RNA])

Change from baseline in log 10-transformed plasma viral load (HIV-1 RNA) levels (log 10 copies per milliliter [log10 copies/mL]). Baseline value calculated as average of pre-dose measurements collected at screening, randomization, and baseline visits. (NCT00098748)
Timeframe: Baseline to Week 24 and Week 48

,,
Interventionlog10 copies/mL (Mean)
Week 24Week 48
Maraviroc BID-1.194-1.105
Maraviroc QD-0.890-0.604
Placebo-0.9530.839

[back to top]

Change From Baseline in CD4 Cell Count

Change from baseline in CD4 cell count (measured as cells per microliter [cells/µL]). Baseline value calculated as the average of pre-dose measurements collected at screening, randomization, and baseline visits. (NCT00098748)
Timeframe: Baseline to Week 24 and Week 48

,,
Interventioncells/µL (Mean)
Week 24Week 48
Maraviroc BID62.65178.87
Maraviroc QD59.23765.86
Placebo36.36751.29

[back to top]

Percentage of Participants With All Causality Treatment-emergent Adverse (AEs) Events by Gender

Treatment-emergent AEs by gender that occurred up to 30 days after the last dose of study medication. (NCT00426660)
Timeframe: Baseline up to Week 144

Interventionpercentage of participants (Number)
Male (n=827)Female (n=205)
Maraviroc69.673.7

[back to top]

Percentage of Participants With ≥0.5 log10 Reduction From Baseline in Human Immunodeficiency Virus 1 Ribonucleic Acid (HIV 1 RNA)

"Defined as HIV-1 RNA levels < 400 Copies/mL or at least 0.5 Log 10-decrease from baseline in HIV-1 RNA levels.~Baseline value calculated as average of the screening and baseline values if both values were within 1 log10 difference." (NCT00426660)
Timeframe: Baseline up to Week 144

Interventionpercentage of participants (Number)
Day 2-7 (n=26)Week 2 (n=226)Week 4 (n=773)Week 8 (n=761)Week 12 (n=756)Week 16 (n=155)Week 20 (n=131)Week 24 (n=557)Week 32 (n=202)Week 40 (n=227)Week 48 (n=228)Week 60 (n=160)Week 72 (n=125)Week 84 (n=79)Week 96 (n=48)Week 108 (n=18)Week 120 (n=9)Week 132 (n=4)Week 144 (n=1)
Maraviroc46.293.89393.29487.793.191.993.191.289908884.891.7100100100100

[back to top]

Change From Baseline in CD8 Cell Count

Change from baseline in cluster of differentiation 8 suppressor T cells (CD8) cell count. If baseline value was not available, it was taken from immediate preceding non-missing value. (NCT00426660)
Timeframe: Baseline up to Week 144

Interventioncells/uL (Mean)
Day 2-7 (n=6)Week 2 (n=201)Week 4 (n=701)Week 8 (n=692)Week 12 (n=696)Week 16 (n=129)Week 20 (n=119)Week 24 (n=523)Week 32 (n=185)Week 40 (n=222)Week 48 (n=214)Week 60 (n=157)Week 72 (n=120)Week 84 (n=70)Week 96 (n=45)Week 108 (n=19)Week 120 (n=8)Week 132 (n=4)Week 144 (n=1)
Maraviroc268.578.2205.6345.6309.4259.6325.2251.3185.2160.9153.276.968.7-11.656.6147.5-135.1-363.3642.0

[back to top]

Percentage of Participants With HIV-1 RNA Levels Below the Limit of Quantification: <50 Copies/mL

Limit of quantification defined as <50 copies/mL. Baseline value calculated as average of the screening and baseline values if both values were within 1 log 10 difference. (NCT00426660)
Timeframe: Baseline up to Week 144

Interventionpercentage of participants (Number)
Baseline (n=979)Day 2-7 (n=26)Week 2 (n=226)Week 4 (n=773)Week 8 (n=761)Week 12 (n=756)Week 16 (n=155)Week 20 (n=131)Week 24 (n=557)Week 32 (n=202)Week 40 (n=227)Week 48 (n=228)Week 60 (n=160)Week 72 (n=125)Week 84 (n=79)Week 96 (n=48)Week 108 (n=18)Week 120 (n=9)Week 132 (n=4)Week 144 (n=1)
Maraviroc1.70.019.930.143.145.837.442.755.857.454.654.851.949.655.754.277.844.475.0100

[back to top]

Percentage of Participants With HIV-1 RNA Levels Below the Limit of Quantification: <400 Copies/mL

Limit of quantification defined as <400 copies/mL. Baseline value calculated as average of the screening and baseline values if both values were within 1 log 10 difference. (NCT00426660)
Timeframe: Baseline up to Week 144

Interventionpercentage of participants (Number)
Baseline (n=979)Day 2-7 (n=26)Week 2 (n=226)Week 4 (n=773)Week 8 (n=761)Week 12 (n=756)Week 16 (n=155)Week 20 (n=131)Week 24 (n=557)Week 32 (n=202)Week 40 (n=227)Week 48 (n=228)Week 60 (n=160)Week 72 (n=125)Week 84 (n=79)Week 96 (n=48)Week 108 (n=18)Week 120 (n=9)Week 132 (n=4)Week 144 (n=1)
Maraviroc6.73.858.872.778.882.575.583.284.483.781.180.778.172.874.775.088.977.8100100

[back to top]

Percentage of Participants With Grade 4 Laboratory Abnormalities Without Regards to Baseline Abnormalities

Laboratory abnormalities as defined by the Division of AIDS (DAIDS) toxicity grading scale: Grade 4, Very Severe = events which were unacceptable and intolerable or were irreversible or caused the participant to be in imminent danger of death. (NCT00426660)
Timeframe: Baseline up to Week 144

Interventionpercentage of participants (Number)
ALT (n=1013)AST (n=1013)Absolute Neutrophil Count (n=1004)Alkaline Phosphatase (n=1014)Creatinine (n=1014)GGT (n=1014)Hemoglobin (n=1005)Hyperbilirubinemia (n=1013)Hyperuricemia (n=1014)Hypophosphatemia (n=1014)Lipase (n=409)Platelets (n=997)Potassium (hyperkalemia) (n=1014)Serum Amylase (n=1014)Sodium (hypernatremia) (n=1014)Triglycerides (n=1014)
Maraviroc0.790.390.200.000.101.480.100.300.100.001.220.300.200.300.201.97

[back to top]

Percentage of Participants With Change in Chemokine Co-receptor Tropism From Screening to Time of Virologic Failure

Tropism status (CCR5 [R5], CXCR4 [X4], Dual Mixed [DM], or Non-reportable [NR]) at Screening (Scr) and time of virologic failure (V fail). Virologic failure defined as: failure to achieve a reduction from baseline (BL) in HIV 1 RNA ≥0.5 log10 copies/mL by second viral load determination (unless viral load was below lower limit level of quantification [LLOQ]); or a ≥ 0.5 log10 increase from nadir in HIV 1 RNA after achieving HIV 1 RNA reduction from BL >0.5 log10 copies/mL; or a HIV 1 RNA level of >1000 copies/mL after having achieved a HIV 1 RNA level below LLOQ. (NCT00426660)
Timeframe: Screening up to Week 144

Interventionpercentage of participants (Number)
Scr: Missing; V fail: R5Scr: Missing; V fail: X4Scr: Missing; V fail: DMScr: Missing; V fail: NRScr: Missing; V fail: MissingScr: NR; V fail: R5Scr: R5; V fail: R5Scr: R5; V fail: X4Scr: R5; V fail: DMScr: R5; V fail: NRScr: R5; V fail: Missing
Maraviroc5.730.521.047.291.560.5231.254.1718.7522.926.25

[back to top]

Change From Baseline in CD8 Cell Count Percent

Change from baseline in CD8 cell count percent . If baseline value was not available, it was taken from immediate preceding non-missing value. (NCT00426660)
Timeframe: Baseline up to Week 144

Interventionpercentage of lymphocytes (Mean)
Day 2-7 (n=6)Week 2 (n=197)Week 4 (n=689)Week 8 (n=683)Week 12 (n=675)Week 16 (n=125)Week 20 (n=115)Week 24 (n=510)Week 32 (n=184)Week 40 (n=215)Week 48 (n=202)Week 60 (n=146)Week 72 (n=113)Week 84 (n=64)Week 96 (n=40)Week 108 (n=19)Week 120 (n=6)Week 132 (n=4)Week 144 (n=1)
Maraviroc3.0-1.5-0.30.4-0.90.7-0.7-3.2-4.0-4.5-5.1-5.0-6.2-8.2-9.5-14.5-14.5-16.8-2.0

[back to top]

Percentage of Participants With Treatment-emergent Averse Events (AEs) by Baseline Hepatitis B and Hepatitis C Virus Serology Status

Treatment emergent AEs by hepatis B and hepatitis C serology status that occurred up to 30 days post last dose. (NCT00426660)
Timeframe: Baseline up to Week 144

Interventionpercentage of participants (Number)
HBV: negative (n=897)HBV: positive (n=60)HBV: missing: (n=75)HCV: negative (n=794)HCV: positive (n=158)HCV: missing (n=80)
Maraviroc71.365.064.072.762.763.8

[back to top]

Percentage of Participants With Treatment-emergent Adverse Events (AEs) by Race

Treatment-emergent AEs by race that occurred up to 30 days after the last dose of study medication. (NCT00426660)
Timeframe: Baseline up to Week 144

Interventionpercentage of participants (Number)
White (n=817)Black (n=106)Asian (n=54)Other Unspecified (n=55)
Maraviroc68.567.988.985.5

[back to top]

Percentage of Participants With Treatment-emergent Adverse Events (AEs) by Age

Treatment-emergent AEs by age that occurred up to 30 days after the last dose of study medication. (NCT00426660)
Timeframe: Baseline up to Week 144

Interventionpercentage of participants (Number)
<18 years (n=1)18 to 44 years (n=515)45 to 64 years (n=493)>=65 years (n=23)
Maraviroc100.071.768.682.6

[back to top]

Percentage of Participants With ≥1.0 log10 Reduction From Baseline in HIV 1 RNA

Defined as HIV-1 RNA levels < 400 copies/mL or at least 1.0 Log 10-decrease from baseline in HIV-1 RNA levels. Baseline value calculated as average of the screening and baseline values if both values were within 1 log10 difference. (NCT00426660)
Timeframe: Baseline up to Week 144

Interventionpercentage of participants (Number)
Day 2-7 (n=26)Week 2 (n=226)Week 4 (n=773)Week 8 (n=761)Week 12 (n=756)Week 16 (n=155)Week 20 (n=131)Week 24 (n=557)Week 32 (n=202)Week 40 (n=227)Week 48 (n=228)Week 60 (n=160)Week 72 (n=125)Week 84 (n=79)Week 96 (n=48)Week 108 (n=18)Week 120 (n=9)Week 132 (n=4)Week 144 (n=1)
Maraviroc30.890.390.291.291.382.689.38990.187.786.485.683.279.785.4100100100100

[back to top]

Percentage of Participants With Acquired Immunodeficiency Syndrome (AIDS)-Defining Illnesses

Treatment-emergent AIDS-defining opportunistic illnesses based on investigator classification guided by a predefined list of clinical Category C adverse events per Center for Disease Control (CDC) HIV Classification System. Includes events occurring up to 30 days after last dose of study drug. (NCT00426660)
Timeframe: Baseline up to Week 144

Interventionpercentage of participants (Number)
AIDS encephalopathyBone tuberculosisCytomegalovirus infectionExtrapulmonary tuberculosisMeningitis cryptococcalMeningitis tuberculousMycobacterium avium complex infectionMycobacterium kansasii infectionOesophageal candidiasisPancreatitis bacterialPneumocystis jiroveci pneumoniaProgressive multifocal leukoencephalopathyPulmonary tuberculosisToxoplasmosisB-cell lymphomaHodgkin's diseaseKaposi's sarcomaEncephalopathy
Maraviroc0.10.10.20.10.10.10.10.10.60.10.10.10.50.10.10.10.20.1

[back to top]

Median Time to Virologic Failure

Computed as time from the first dose of study medication to the loss of virologic response. Virologic failure defined as: failure to achieve a reduction from baseline (BL) in HIV 1 RNA ≥ 0.5 log10 copies /mL by the second viral load determination (unless viral load was below the lower limit level of quantification [LLOQ]); or a ≥ 0.5 log10 increase from nadir in HIV 1 RNA after achieving a HIV 1 RNA reduction from BL >0.5 log10 copies/mL; or a HIV 1 RNA level of >1000 copies/mL after having achieved a HIV 1 RNA level below LLOQ. (NCT00426660)
Timeframe: Day 1 up to Week 144

Interventiondays (Median)
Maraviroc86.5

[back to top]

Number of Participants With Emergence of Resistance to Maraviroc as Defined by Genotypic Changes in the V3 Loop of Glycoprotein 120 (gp 120)

Virus from participants who experienced virologic failure was analyzed for resistance to maraviroc. Resistance testing was performed on archived samples of participants which were available pre--treatment at time of virologic failure. For participants who met definition of virologic failure during the trial, the sequencing of the V3 loop of HIV--1 viral envelope gp 120 was evaluated to identify any amino acid changes concomitant with decreased susceptibility to maraviroc. (NCT00426660)
Timeframe: Baseline up to Week 144

Interventionparticipants (Number)
MaravirocNA

[back to top]

Number of Participants With Reduced Maraviroc Susceptibility as Defined by Change From Baseline to Time of Virologic Failure in Inhibitory Concentration of 50% (IC 50) and Presence of Plateau

Resistance to maravroc in viruses from participants failing therapy with R5 virus was investigated using the in vitro phenotypic (drug susceptibility) assay. The number of participants who failed with R5 virus were assessed successfully for maraviroc susceptibility at Baseline and Last on--treatment (Week 144). Samples were analyzed for change from Baseline to time of virologic failure in IC 50 and presence of plateau. A maximal percent inhibition (MPI) <95% established as a plateau in inhibition at high concentrations of maraviroc was used to identify viruses which had reduced phenotypic susceptibility to maraviroc. (NCT00426660)
Timeframe: Baseline up to Week 144

Interventionparticipants (Number)
Maraviroc14

[back to top]

Change From Baseline in CD4 Cell Count

Change from baseline in cluster of differentiation 4 helper T cells (CD4) cell count. If baseline value was not available, it was taken from immediate preceding non-missing value. (NCT00426660)
Timeframe: Baseline up to Week 144

Interventioncells/uL (Mean)
Day 2-7 (n=6)Week 2 (n=213)Week 4 (n=762)Week 8 (n=739)Week 12 (n=738)Week 16 (n=149)Week 20 (n=130)Week 24 (n=552)Week 32 (n=197)Week 40 (n=232)Week 48 (n=222)Week 60 (n=161)Week 72 (n=124)Week 84 (n=72)Week 96 (n=50)Week 108 (n=19)Week 120 (n=8)Week 132 (n=4)Week 144 (n=1)
Maraviroc28.549.768.994.595.785.9106.6127.8124.1129.2140.5134.0147.0141.1160.3242.9117.9179.093.0

[back to top]

Change From Baseline in CD4 Cell Count Percent

Change from baseline in CD4 cell count percent . If baseline value was not available, it was taken from immediate preceding non-missing value. (NCT00426660)
Timeframe: Baseline up to Week 144

Interventionpercentage of lymphocytes (Mean)
Days 2-7 (n=6)Week 2 (n=206)Week 4 (n=733)Week 8 (n=722)Week 12 (n=708)Week 16 (n=141)Week 20 (n=125)Week 24 (n=531)Week 32 (n=191)Week 40 (n=218)Week 48 (n=210)Week 60 (n=148)Week 72 (n=116)Week 84 (n=64)Week 96 (n=41)Week 108 (n=19)Week 120 (n=7)Week 132 (n=4)Week 144 (n=1)
Maraviroc-1.31.81.61.41.61.92.22.83.23.73.54.55.05.73.96.35.78.51.0

[back to top]

Percentage of Participants With Changes in HIV-1 RNA Level in Participants Meeting the Definition of Virologic Failure

Reasons for virologic failure: A) failure to achieve a reduction in HIV-1 RNA>=0.5 log10 copies/ml from baseline (BL) by second viral load determination (unless below level of quantification [LOQ]); B) >=0.5 log10 increase from nadir in HIV-1 RNA after achieving an HIV-1 RNA reduction from BL >0.5 log10 copies/ml ; C) HIV-1 RNA >1000 copies/ml after having achieved an HIV-1 RNA below LOQ. (NCT00426660)
Timeframe: Baseline up to Week 144

Interventionpercentage of participants (Number)
Reduction in HIV-1 RNA<0.5 log10 copies/ml>=0.5 log10 increase from nadir in HIV-1 RNAHIV-1 RNA >1000 copies/mlReduction <0.5 log10 + HIV-1 RNA >1000 copies/ml>=0.5 log10 increase from nadir + HIV-1 RNA >1000
Maraviroc43.7531.7718.753.132.60

[back to top]

Percentage of Participants With Grade 3 and Grade 4 Adverse Events (AE)

AEs as defined by the Division of AIDS (DAIDS) toxicity grading scale: Grade 3 = severe: interrupted usual daily activity and traditionally required systemic drug therapy or other treatment. Grade 4 = very severe: events that were unacceptable and intolerable or were irreversable or caused imminent danger of death. If same participant had more than 1 occurrence in the same preferred term event category, only the most severe (grade 4) occurrence was taken. Treatment-related = investigator assessment of a reasonable possibility that the investigational product caused or contributed to the AE. (NCT00426660)
Timeframe: Baseline up to Week 144

Interventionpercentage of participants (Number)
Grade 3 AE: all causalityGrade 3 AE: treatment relatedGrade 4 AE: all causalityGrade 4 AE: treatment related
Maraviroc13.22.46.31.6

[back to top]

Percentage of Participants With Grade 3 Laboratory Abnormalities Without Regards to Baseline Abnormalities

Laboratory abnormalities as defined by the Division of AIDS (DAIDS) toxicity grading scale: Grade 3, Severe =events that interrupted participants usual daily activity and traditionally required systemic drug therapy or other treatment. (NCT00426660)
Timeframe: Baseline up to Week 144

Interventionpercentage of participants (Number)
Alanine aminotransferase (ALT) (n=1013)Aspartate aminotransferase (AST) (n=1013)Absolute Neutrophil Count (n=1004)Alkaline Phosphatase (n=1014)Creatinine (n=1014)Gamma-glutamyl transpeptidase (GGT) (n=1014)Hemoglobin (n=1005)Hyperbilirubinemia (n=1013)Hyperuricemia (n=1014)Hypophosphatemia (n=1014)Lipase (n=409)Platelets (n=997)Potassium (hyperkalemia) (n=1014)Serum Amylase (n=1014)Sodium (hypernatremia) (n=1014)Triglycerides (n=1014)
Maraviroc2.172.070.700.390.494.730.201.280.690.204.401.500.104.830.104.73

[back to top]

Number of Subjects With Categorical Absolute Vital Signs and Vital Sign Changes Compared to Baseline

Baseline was defined to be the latest non-missing value from a range of pre-treatment visits. Maximum increase from baseline in supine and standing systolic BP was > = 30 mmHg, and maximum increase from baseline in supine and standing diastolic BP was > = 20 mmHg. (NCT00427934)
Timeframe: Baseline, 16 weeks

,,,
InterventionParticipants (Number)
Maximum Increase in Supine Systolic BPMaximum Increase in Standing Systolic BPMaximum Increase in Supine Diastolic BPMaximum Increase in Standing Diastolic BP
Maraviroc 150 mg BID (PK)2110
Maraviroc 300 mg BID (PK)1110
Maraviroc 300 mg BID (POC)4283
Placebo (POC)3202

[back to top]

Number of Subjects With Withdrawal From Study Due to Lack of Efficacy

Withdrawal is the total number of withdrawals from the study. Withdrawal due to lack of efficacy was collected based on the investigator's judgement. (NCT00427934)
Timeframe: 16 weeks

,
Interventionparticipants (Number)
Overall WithdrawalWithdrawal due to Lack of Efficacy
Maraviroc 300 mg BID (POC)225
Placebo (POC)142

[back to top]

Survival Analysis of Time to Withdrawal: Proportion of Subjects Who Did Not Withdraw From the Study Due to Lack of Efficacy.

Withdrawal due to lack of efficacy was collected based on the investigator's judgement. Time to withdrawal was measured by the probability that a subject did not withdraw due to lack of efficacy by a particular visit. This was a statistical estimate (Kaplan-Meier Survival Analysis) of the probability that a participant would not withdraw due to lack of efficacy. (NCT00427934)
Timeframe: Weeks 1 to 12

,
Interventionproportion (Number)
Week 1 (n=77, 33)Week 2 (n=77, 32)Week 4 (n=76, 32)Week 8 (n=69, 31)Week 12 (n=58, 21)
Maraviroc 300 mg BID (POC)1.001.001.000.990.93
Placebo (POC)1.001.001.001.000.92

[back to top]

Change From Baseline in 12-Lead Electrocardiogram (ECG) Parameters (RR Interval, PR Interval, QRS Complex, QT Interval, Corrected QT [QTc] Interval, QTcB Interval [Bazett's Correction], QTcF Interval [Fridericia's Correction]).

Baseline was defined to be the latest non-missing value from a range of pre-treatment visits. Means of replicate values were used. QTc interval was not measured for the PK populations. (NCT00427934)
Timeframe: Baseline, 16 weeks

,,,
Interventionmsec (Mean)
RR IntervalPR IntervalQRS ComplexQT IntervalQTc IntervalQTcB IntervalQTcF Interval
Maraviroc 150 mg BID (PK)137.019.77.635.0023.122.9
Maraviroc 300 mg BID (PK)114.313.07.322.5023.317.3
Maraviroc 300 mg BID (POC)140.016.211.331.919.627.928.3
Placebo (POC)117.615.410.228.720.330.229.8

[back to top]

Change From Baseline in 12-Lead Electrocardiogram (ECG) Parameters (Heart Rate).

Baseline was defined to be the latest non-missing value from a range of pre-treatment visits. Means of replicate values were used. (NCT00427934)
Timeframe: Baseline, 16 weeks

Interventionbpm (Mean)
Maraviroc 150 mg BID (PK)11.2
Maraviroc 300 mg BID (PK)8.8
Maraviroc 300 mg BID (POC)12.0
Placebo (POC)10.5

[back to top]

American College of Rheumatology (ACR) 20% Responders at Week 12

A subject was an ACR 20 responder if: the counts for both tender and swollen joints had reduced by 20% or more from baseline; and 3 out of the following 5 assessments showed reduction of 20% or more from baseline assessment: Patient's Assessment of Arthritis Pain (Visual Analogue Scale [VAS]), Patient's Global Assessment of Arthritis (VAS), Physician's Global Assessment of Arthritis (Categorical), Health Assessment Questionnaire - Disability Index (HAQ-DI), and C-Reactive Protein (CRP). (NCT00427934)
Timeframe: Week 12

InterventionParticipants (Number)
Maraviroc 300 mg BID (POC)21
Placebo (POC)6

[back to top]

ACR 70% Responders at Weeks 1, 2, 4, 8, and 12

A subject was an ACR 70 responder if: the counts for both tender and swollen joints had reduced by 70% or more from baseline; and 3 out of the following 5 assessments showed reduction of 70% or more from baseline assessment: Patient's Assessment of Arthritis Pain, Patient's Global Assessment of Arthritis, Physician's Global Assessment of Arthritis, HAQ-DI, and CRP. The Week 16 visit (follow-up) was designed for safety rather than efficacy, thus Week 16 ACR 70% data were collected, but not analyzed. (NCT00427934)
Timeframe: Weeks 1, 2, 4, 8, and 12

,
InterventionParticipants (Number)
Week 1Week 2Week 4Week 8Week 12
Maraviroc 300 mg BID (POC)00020
Placebo (POC)00101

[back to top]

Change From Baseline in CRP at Weeks 1, 2, 4, 8, and 12

Change from baseline at each visit were analyzed for CRP. The Week 16 visit (follow-up) was designed for safety rather than efficacy, thus Week 16 CRP data were collected, but not analyzed. (NCT00427934)
Timeframe: Baseline, Weeks 1, 2, 4, 8, and 12

,
Interventionmg/L (Least Squares Mean)
Week 1Week 2Week 4Week 8Week 12
Maraviroc 300 mg BID (POC)1.203.143.362.332.35
Placebo (POC)3.401.81-1.30-1.141.93

[back to top]

Change From Baseline in Disease Activity Score Using CRP (DAS28-4[CRP]) at Weeks 1, 2, 4, 8, and 12

"DAS28-4 (CRP) was calculated using the following formula:~DAS28- 4(CRP) = 0.56 √28 Tender Joint Count + 0.28 √28 Swollen Joint Count + 0.36*natural logarithm(CRP + 1) + 0.014*Patient Global Assessment + 0.96. DAS28 provides a number on a scale (0 to 10) indicating current disease activity. A score above 5.1 means high disease activity and a score below 3.2 indicates low disease activity. Change from baseline at each visit was analyzed for DAS28-4 (CRP). The Week 16 visit (follow-up) was designed for safety rather than efficacy, thus Week 16 DAS28-4 (CRP) data were collected, but not analyzed." (NCT00427934)
Timeframe: Baseline, Weeks 1, 2, 4, 8, and 12

,
Interventionmg/L (Least Squares Mean)
Week 1Week 2Week 4Week 8Week 12
Maraviroc 300 mg BID (POC)-0.23-0.41-0.64-0.82-0.73
Placebo (POC)-0.17-0.33-0.65-0.51-0.63

[back to top]

Change From Baseline in HAQ-DI at Weeks 1, 2, 4, 8, and 12

HAQ-DI assesses degree of difficulty experienced in daily activity categories (dressing/grooming, arising, eating, walking, hygiene, reach, grip and other activities) over the past week. There are 20 questions and difficulty is scored from 0 (none), 1 (some), 2 (much) and 3 (unable to do). Scores were then averaged to give the disability index (scale of 0 to 3). Change from baseline at each visit was analyzed. The Week 16 visit (follow-up) was designed for safety rather than efficacy, thus Week 16 HAQ-DI data were collected but not analyzed. (NCT00427934)
Timeframe: Baseline, Weeks 1, 2, 4, 8, and 12

,
Interventionscores on scale (Least Squares Mean)
Week 1Week 2Week 4Week 8Week 12
Maraviroc 300 mg BID (POC)-0.08-0.22-0.24-0.25-0.18
Placebo (POC)-0.03-0.04-0.18-0.03-0.06

[back to top]

Change From Baseline in Mean Heart Rate

Heart rate = standing and supine at the same time the orthostatic BP measurements were obtained. Baseline was defined to be the latest non-missing value from a range of pre-treatment visits. Means of replicate values were not used. (NCT00427934)
Timeframe: Baseline, 16 weeks

,,,
Interventionbeats per minute (bpm) (Mean)
Standing Heart RateSupine Heart Rate
Maraviroc 150 mg BID (PK)14.615.0
Maraviroc 300 mg BID (PK)8.110.4
Maraviroc 300 mg BID (POC)11.811.6
Placebo (POC)11.49.2

[back to top]

Change From Baseline in Mean Orthostatic Blood Pressure (BP)

Supine BP was recorded after 5 minutes lying down; subjects then sat for 2 minutes then stood for 2 minutes and standing BP was recorded. Orthostatic BP = either a systolic BP drop > 20 mmHg, or diastolic BP drop > 10 mmHg and/or drop in systolic BP < 90 mmHg. If a subject met the orthostatic criteria, they were required to complete 2 additional readings to provide a triplicate reading. The means of replicate BP values were used in the analysis. Baseline = the latest non-missing value from a range of pre-treatment visits. Change from baseline to Week 16 was analyzed for orthostatic BP. (NCT00427934)
Timeframe: Baseline, 16 weeks

,,,
InterventionmmHg (Mean)
Standing Systolic BPSupine Systolic BPStanding Diastolic BPSupine Diastolic BP
Maraviroc 150 mg BID (PK)20.920.913.010.3
Maraviroc 300 mg BID (PK)18.018.38.310.9
Maraviroc 300 mg BID (POC)17.817.511.712.2
Placebo (POC)17.518.011.510.8

[back to top]

Change From Baseline in Patient's Assessment of Arthritis Pain at Weeks 1, 2, 4, 8, and 12

The severity of arthritis was scored by the subject between 0 (no pain) and 100 (most severe pain) on a 100 mm VAS. Change from baseline at each visit was analyzed for Patient's Assessment of Arthritis Pain. The Week 16 visit (follow-up) was designed for safety rather than efficacy, thus Week 16 Patient's Assessment of Arthritis Pain data were collected, but not analyzed. (NCT00427934)
Timeframe: Baseline, Weeks 1, 2, 4, 8, and 12

,
Interventionscores on scale (Least Squares Mean)
Week 1Week 2Week 4Week 8Week 12
Maraviroc 300 mg BID (POC)-3.96-8.46-8.35-10.30-8.30
Placebo (POC)-3.62-3.20-9.08-8.67-6.09

[back to top]

Change From Baseline in Patient's Global Assessment of Arthritis Pain at Weeks 1, 2, 4, 8, and 12

"Subjects answered: Considering all the ways your arthritis affects you, how are you feeling today? Subjects responded by using a 0 - 100 mm VAS where 0=very well and 100=very poorly. Change from baseline at each visit was analyzed for Patient's Global Assessment. The Week 16 visit (follow-up) was designed for safety rather than efficacy, thus Week 16 Patient's Global Assessment of Arthritis Pain data were collected, but not analyzed." (NCT00427934)
Timeframe: Baseline, Weeks 1, 2, 4, 8, and 12

,
Interventionscores on scale (Least Squares Mean)
Week 1Week 2Week 4Week 8Week 12
Maraviroc 300 mg BID (POC)-4.70-8.85-10.00-11.12-8.55
Placebo (POC)-2.34-4.88-9.02-3.44-6.78

[back to top]

Change From Baseline in Physician's Global Assessment of Arthritis Pain at Weeks 1, 2, 4, 8, and 12

Physician's evaluation based on subject's disease signs, functional capacity and physical exam. Response recorded using 5-point scale: 1=Very Good, 2=Good, 3=Fair, 4=Poor and 5=Very Poor. Change from baseline at each visit was analyzed for Physician's Global Assessment. The Week 16 visit (follow-up) was designed for safety rather than efficacy thus Week 16 Physician's Global Assessment of Arthritis Pain data were collected but not analyzed. (NCT00427934)
Timeframe: Baseline, Weeks 1, 2, 4, 8, and 12

,
Interventionscores on scale (Least Squares Mean)
Week 1Week 2Week 4Week 8Week 12
Maraviroc 300 mg BID (POC)-0.30-0.44-0.49-0.63-0.49
Placebo (POC)-0.22-0.39-0.44-0.37-0.36

[back to top]

Area Under the Plasma Concentration-Time Profile From Time Zero to Four Hours Postdose (AUC 0-4) for MTX at Screening and Week 1 and Maraviroc at Week 1

Effect of maraviroc on the PK of MTX (comparison of AUC0-4 of MTX at screening versus at Week 1 after coadministration with 150 mg or 300 mg of maraviroc). PK was assessed at screening (MTX) and at Week 1 (maraviroc and MTX). (NCT00427934)
Timeframe: Screening (1, 2, 3, and 4 hours post-dose), Week 1 (0.5, 1, 2, 3, and 4 hours post-dose)

,
Interventionng.hr/mL (Geometric Mean)
Plasma MTX (Screening)Plasma MTX (Week 1)Plasma Maraviroc (Week 1)
Maraviroc 150 mg BID (PK)909.41045.6451.6
Maraviroc 300 mg BID (PK)888.9844.81106.8

[back to top]

Change From Baseline in SF-36 Mental Component Summary at Weeks 4 and 12

The SF-36 v.2 (Acute version) [12] is a 36-item generic health status measure that measures 8 general health concepts: Physical functioning, role-physical, bodily pain, general health, vitality, social functioning, role-emotional, and mental health. Each domain of the eight domains and the summary concept (mental component score) are scored to yield values between 0 (worst) and 100 (best). Change from baseline at Weeks 4 and 12 were analyzed for SF-36. Due to the termination of the study, SF-36 results for the PK component group were not analyzed. (NCT00427934)
Timeframe: Baseline, Weeks 4 and 12

,
Interventionscores on a scale (Least Squares Mean)
Week 4Week 12
Maraviroc 300 mg BID (POC)1.421.17
Placebo (POC)0.720.61

[back to top]

Change From Baseline in Short Form-36 (SF-36) Physical Component Summary at Weeks 4 and 12

The SF-36 v.2 (Acute version) is a 36-item generic health status measure that measures 8 general health concepts: Physical functioning, role-physical, bodily pain, general health, vitality, social functioning, role-emotional, and mental health. Each domain of the eight domains and the summary concept (physical component score) are scored to yield values between 0 (worst) and 100 (best). Change from baseline at Weeks 4 and 12 were analyzed for SF-36. Due to the termination of the study, SF-36 results for the PK component group were not analyzed. (NCT00427934)
Timeframe: Baseline, Weeks 4 and 12

,
Interventionscores on a scale (Least Squares Mean)
Week 4Week 12
Maraviroc 300 mg BID (POC)4.333.14
Placebo (POC)3.764.81

[back to top]

Change From Baseline in Swollen Joint Count at Weeks 1, 2, 4, 8, and 12

Change from baseline at each visit was analyzed for swollen joint count. Twenty-eight tender and swollen joint scores included the same joints: shoulders, elbows, wrists, MCP joints, PIP joints, and the knees. The Week 16 visit (follow-up) was designed for safety rather than efficacy, thus Week 16 swollen joint count data were collected, but not analyzed. (NCT00427934)
Timeframe: Baseline, Weeks 1, 2, 4, 8, and 12

,
Interventionjoint count (Least Squares Mean)
Week 1Week 2Week 4Week 8Week 12
Maraviroc 300 mg BID (POC)-1.39-2.25-3.93-4.48-3.48
Placebo (POC)-1.07-2.51-4.08-3.03-3.43

[back to top]

Change From Baseline in Tender/Painful Joint Count at Weeks 1, 2, 4, 8, and 12

Change from baseline at each visit was analyzed for tender/painful joint count. Twenty-eight tender and swollen joint scores included the same joints: shoulders, elbows, wrists, metacarpophalangeal joints (MCP), proximal interphalangeal joints (PIP), and the knees. The Week 16 visit (follow-up) was designed for safety rather than efficacy, thus Week 16 tender/painful joint count data were collected, but not analyzed. (NCT00427934)
Timeframe: Baseline, Weeks 1, 2, 4, 8, and 12

,
Interventionjoint count (Least Squares Mean)
Week 1Week 2Week 4Week 8Week 12
Maraviroc 300 mg BID (POC)-0.81-2.08-4.00-4.85-4.89
Placebo (POC)-1.91-2.38-4.01-3.24-3.41

[back to top]

Maximum Observed Concentration (Cmax) During the Dosing Interval for MTX at Screening and Week 1 and Maraviroc at Week 1

Effect of maraviroc on the PK of MTX (comparison of Cmax of MTX at screening versus at Week 1 after coadministration with 150 mg or 300 mg of maraviroc). PK was assessed at screening (MTX) and at Week 1 (maraviroc and MTX). (NCT00427934)
Timeframe: Screening (1, 2, 3, and 4 hours post-dose), Week 1 (0.5, 1, 2, 3, and 4 hours post-dose)

,
Interventionng/mL (Geometric Mean)
Plasma MTX (Screening)Plasma MTX (Week 1)Plasma Maraviroc (Week 1)
Maraviroc 150 mg BID (PK)338.7403.8199.59
Maraviroc 300 mg BID (PK)352.1322.8461.03

[back to top]

Number of Subjects With Categorical Absolute ECG Parameters and ECG Changes Compared to Baseline

Maximum QTcB, QTcF, and QTc intervals were defined as 450 to < 480 msec, 480 to < 500 msec, or > = 500 msec. (NCT00427934)
Timeframe: Baseline, 16 weeks

,,,
InterventionParticipants (Number)
Maximum QTc Interval 450 to < 480 msecMaximum QTc Interval 480 to < 500 msecMaximum QTc Interval > = 500 msecMaximum QTcB Interval 450 to < 480 msecMaximum QTcB Interval 480 to < 500 msecMaximum QTcB Interval > = 500 msecMaximum QTcF Interval 450 to < 480 msecMaximum QTcF Interval 480 to < 500 msecMaximum QTcF Interval > = 500 msec
Maraviroc 150 mg BID (PK)000100000
Maraviroc 300 mg BID (PK)000300200
Maraviroc 300 mg BID (POC)2001320900
Placebo (POC)200811500

[back to top]

Time for Cmax (Tmax) for MTX at Screening and Week 1 and Maraviroc at Week 1

PK was assessed at screening (MTX) and at Week 1 (maraviroc and MTX). (NCT00427934)
Timeframe: Screening (1, 2, 3, and 4 hours post-dose), Week 1 (0.5, 1, 2, 3, and 4 hours post-dose)

,
Interventionhr (Median)
Plasma MTX (Screening)Plasma MTX (Week 1)Plasma Maraviroc (Week 1)
Maraviroc 150 mg BID (PK)2.0001.0002.000
Maraviroc 300 mg BID (PK)1.0001.5002.500

[back to top]

ACR 50% Responders at Weeks 1, 2, 4, 8, and 12

A subject was an ACR 50 responder if: the counts for both tender and swollen joints had reduced by 50% or more from baseline; and 3 out of the following 5 assessments showed reduction of 50% or more from baseline assessment: Patient's Assessment of Arthritis Pain, Patient's Global Assessment of Arthritis, Physician's Global Assessment of Arthritis, HAQ-DI, and CRP. The Week 16 visit (follow-up) was designed for safety rather than efficacy, thus Week 16 ACR 50% data were collected, but not analyzed. (NCT00427934)
Timeframe: Weeks 1, 2, 4, 8, and 12

,
InterventionParticipants (Number)
Week 1Week 2Week 4Week 8Week 12
Maraviroc 300 mg BID (POC)12268
Placebo (POC)00233

[back to top]

ACR 20% Responders at Weeks 1, 2, 4, and 8

A subject was an ACR 20 responder if: the counts for both tender and swollen joints had reduced by 20% or more from baseline; and 3 out of the following 5 assessments showed reduction of 20% or more from baseline assessment: Patient's Assessment of Arthritis Pain, Patient's Global Assessment of Arthritis, Physician's Global Assessment of Arthritis, HAQ-DI, and CRP. The Week 16 visit (follow-up) was designed for safety rather than efficacy, thus Week 16 ACR 20% data were collected, but not analyzed. (NCT00427934)
Timeframe: Weeks 1, 2, 4, and 8

,
InterventionParticipants (Number)
Week 1Week 2Week 4Week 8
Maraviroc 300 mg BID (POC)8122123
Placebo (POC)4587

[back to top]

Time to Virologic Failure (VF)

Virologic failure was defined as failing to achieve a reduction in HIV-1 RNA of at least 0.5 log10 copies/ml from baseline by the second viral load determination; or experiencing at least 0.5 log10 increase from nadir in HIV-1 RNA after achieving an HIV-1 RNA reduction from baseline more than 0.5 log10 copies/ml; or experiencing an HIV-1 RNA more than 1000 copies/ml after having achieved an HIV-1 RNA below level of quantification. (NCT00478231)
Timeframe: Baseline to Week 96 or EOT

InterventionDays (Median)
Maraviroc255.0

[back to top]

Change From Baseline in CD4 Percent at Week 4, 8, 12, 24, 36, 48, 60, 72, 84 and Week 96 or EOT

(NCT00478231)
Timeframe: Baseline, Week 4, Week 8, Week 12, Week 24, Week 36, Week 48, Week 60, Week 72, Week 84 and Week 96 or EOT

InterventionPercent CD4 (Mean)
BaselineChange at Week 4 (n= 190)Change at Week 8 (n= 190)Change at Week 12 (n= 186)Change at Week 24 (n= 184)Change at Week 36 (n= 165)Change at Week 48 (n= 154)Change at Week 60 (n= 134)Change at Week 72 (n= 129)Change at Week 84 (n= 124)Change at Week 96 or EOT (n= 107)Change at LOCF (n= 201)
Maraviroc12.21.81.32.12.93.64.34.55.25.36.64.9

[back to top]

Change From Baseline in CD8 Percent at Week 4, 8, 12, 24, 36, 48, 60, 72, 84 and Week 96 or EOT

(NCT00478231)
Timeframe: Baseline, Week 4, Week 8, Week 12, Week 24, Week 36, Week 48, Week 60, Week 72, Week 84 and Week 96 or EOT

InterventionPercent CD8 (Mean)
BaselineChange at Week 4 (n= 190)Change at Week 8 (n= 190)Change at Week 12 (n= 186)Change at Week 24 (n= 185)Change at Week 36 (n= 165)Change at Week 48 (n= 155)Change at Week 60 (n= 135)Change at Week 72 (n= 130)Change at Week 84 (n= 124)Change at Week 96 or EOT (n= 107)Change at LOCF (n= 201)
Maraviroc59.80.40.9-0.1-2.5-3.6-4.3-4.7-6.1-6.8-7.7-5.4

[back to top]

Change From Baseline in Human Immunodeficiency Virus (HIV) -1 Viral Load (Ribonucleic Acid [RNA]) at Week 4, 8, 12, 24, 36, 48, 60, 72, 84 and Week 96 or EOT

Change from baseline in log 10-transformed plasma viral load (HIV-1 RNA) levels (log 10 copies per milliliter [log10 copies/ml]). (NCT00478231)
Timeframe: Baseline, Week 4, Week 8, Week 12, Week 24, Week 36, Week 48, Week 60, Week 72, Week 84 and Week 96 or EOT

Interventionlog 10 copies/ml (Mean)
BaselineChange at week 4 (n= 202)Change at week 8 (n= 196)Change at week 12 (n= 190)Change at week 24 (n= 185)Change at week 36 (n= 171)Change at week 48 (n= 158)Change at week 60 (n= 141)Change at week 72 (n= 137)Change at week 84 (n= 125)Change at week 96 or EOT (n= 109)Change at LOCF (n= 206)
Maraviroc4.8-1.5-1.5-1.6-1.6-1.6-1.6-1.6-1.4-1.5-1.7-1.4

[back to top]

Change From Baseline in Lymphocyte Cluster of Differentiation 4 (CD4) Count at Week 4, 8, 12, 24, 36, 48, 60, 72, 84 and Week 96 or EOT

(NCT00478231)
Timeframe: Baseline, Week 4, Week 8, Week 12, Week 24, Week 36, Week 48, Week 60, Week 72, Week 84 and Week 96 or EOT

Interventioncells per microliter (cells/µL) (Mean)
BaselineChange at Week 4 (n= 190)Change at Week 8 (n= 190)Change at Week 12 (n= 186)Change at Week 24 (n= 184)Change at Week 36 (n= 165)Change at Week 48 (n= 154)Change at Week 60 (n= 134)Change at Week 72 (n= 129)Change at Week 84 (n= 124)Change at Week 96 or EOT (n= 107)Change at LOCF (n= 201)
Maraviroc215.693.5110.0132.1137.3157.8172.2187.2199.8198.2225.2174.1

[back to top]

Change From Baseline in Lymphocyte Cluster of Differentiation 8 (CD8) Count at Week 4, 8, 12, 24, 36, 48, 60, 72, 84 and Week 96 or EOT

(NCT00478231)
Timeframe: Baseline, Week 4, Week 8, Week 12, Week 24, Week 36, Week 48, Week 60, Week 72, Week 84 and Week 96 or EOT

Interventioncells/µL (Mean)
BaselineChange at Week 4 (n= 190)Change at Week 8 (n= 190)Change at Week 12 (n= 186)Change at Week 24 (n= 185)Change at Week 36 (n= 165)Change at Week 48 (n= 155)Change at Week 60 (n= 135)Change at Week 72 (n= 130)Change at Week 84 (n= 124)Change at Week 96 or EOT (n= 107)Change at LOCF (n= 201)
Maraviroc1016.4387.3518.3507.2358.1352.3348.6349.1272.4246.7214.5223.0

[back to top]

Number of Participants With C-X-C Chemokine Receptor Type 4 {CXCR4} [X4] Tropism Status

Virus tropism was done by the Monogram Biosciences Trofile assay. (NCT00478231)
Timeframe: Time of virologic failure (VF) and Week 96 or EOT

InterventionParticipants (Number)
VF X4 (n= 134)EOT X4 (n= 146)
Maraviroc78

[back to top] [back to top]

Number of Participants With Division of Acquired Immunodeficiency Syndrome (DAIDS) Grade 3 and Grade 4 Laboratory Abnormalities

Grade 3 or severe events included those that interrupted participant's usual daily activity and traditionally required systemic drug therapy or other treatment. Grade 4 or very severe events included those that were unacceptable and intolerable or which were irreversible or caused the participant to be in imminent danger of death. (NCT00478231)
Timeframe: Baseline to 30 days post-week 96 or ET

InterventionParticipants (Number)
Grade 3: Alanine aminotransferase (ALT)Grade 3: Aspartate aminotransferase (AST)Grade 3: Absolute neutrophil count (ANC)Grade 3: Alkaline phosphataseGrade 3: Gamma-glutamyl transpeptidase (GGT)Grade 3: HyperbilirubinemiaGrade 3: HyperuricemiaGrade 3: Lipase (n= 101)Grade 3: PlateletsGrade 3: Potassium (Hyperkalemia)Grade 3: Serum amylaseGrade 4: Alanine aminotransferase (ALT)Grade 4: Aspartate aminotransferase (AST)Grade 4: Absolute neutrophil count (ANC)Grade 4: Alkaline phosphataseGrade 4: Gamma-glutamyl transpeptidase (GGT)Grade 4: HyperbilirubinemiaGrade 4: HyperuricemiaGrade 4: Lipase (n= 101)Grade 4: PlateletsGrade 4: Potassium (Hyperkalemia)Grade 4: Serum amylase
Maraviroc0471151738301210408612021

[back to top]

Number of Participants With Genotype Resistance

Evolution in resistance to OBT was shown by emergence of new primary or secondary resistance mutations to nucleoside reverse transcriptase inhibitor (NRTI), non-nucleoside reverse transcriptase inhibitor (NNRTI) and protease inhibitor (PI). (NCT00478231)
Timeframe: Baseline through Week 96

InterventionParticipants (Number)
VF: NRTIVF: NNRTIVF: PIVF: NRTI and NNRTIVF: NRTI and PIVF: NNRTI and PIVF: NRTI and NNRTI and PIVF: no NRTI, no NNRTI and no PIVF: no test result data
Maraviroc0016032764048

[back to top]

Number of Participants With Grade 3 and Grade 4 Adverse Events (AEs) and Serious Adverse Events (SAEs)

AEs: any untoward medical occurrence/worsening of pre-existing medical condition, whether or not related to study drug. SAE: any AE that resulted in death; was life threatening; resulted in persistent/significant disability/incapacity; resulted in/prolonged an existing in-patient hospitalization; was congenital anomaly. Grade 3: Events that interrupted participant's usual daily activity and traditionally required systemic drug therapy or other treatment. Grade 4: Events which were unacceptable and intolerable or which were irreversible or caused participant to be in imminent danger of death. (NCT00478231)
Timeframe: Baseline to 30 days post-week 96 or early termination (ET)

InterventionParticipants (Number)
Grade 3 AEsGrade 4 AEsSAEs
Maraviroc40934

[back to top]

Number of Participants With Laboratory Test Abnormalities

Pre-defined criteria based on upper limit normal (ULN) and lower limit normal (LLN) were established for each laboratory test to define the values that would be identified as laboratory test abnormality. (NCT00478231)
Timeframe: Baseline to 30 days post-week 96 or ET

InterventionParticipants (Number)
Hematocrit (<0.8*LLN)Red blood cell count (<0.8*LLN)White blood cell count (<0.6*LLN)White blood cell count (>1.5*ULN)Absolute lymphocytes (<0.8*LLN)Absolute lymphocytes (>1.2*ULN)Percent lymphocytes (<0.8*LLN) (n= 1)Percent lymphocytes (>1.2*ULN) (n= 1)Percent neutrophils (<0.8*LLN) (n= 1)Percent neutrophils (>1.2*ULN) (n= 1)Absolute basophils (>1.2*ULN)Percent basophils (>1.2*ULN) (n= 1)Absolute eosinophils (>1.2*ULN)Percent eosinophils (>1.2*ULN) (n= 1)Absolute monocytes (>1.2*ULN)Percent monocytes (>1.2*ULN) (n= 1)Direct Bilirubin (>1.5*ULN)Indirect bilirubin (>1.5*ULN)Lactose dehydrogenase (LDH) (>3.0*ULN)Total protein (<0.8*LLN)Total protein (>1.2*ULN)Albumin (<0.8*LLN)Albumin (>1.2*ULN)Blood urea nitrogen (BUN) (>1.3*ULN)Cholesterol (>1.3*ULN)High density lipoprotein cholesterol (<0.8*LLN)Low density lipoprotein cholesterol (>1.2*ULN)Triglycerides (>1.3*ULN)Calcium (<0.9*LLN)Calcium (>1.1*ULN)Creatine kinase (CK) (>2.0*ULN)
Maraviroc1335741260011010032025067251119307261151011456050

[back to top]

Number of Participants With Treatment Emergent Malignancies

(NCT00478231)
Timeframe: Baseline to 30 days post-week 96 or ET

InterventionParticipants (Number)
Carcinoma in situHodgkin's diseaseIntestinal T-cell lymphomaNeoplasm of appendix
Maraviroc1111

[back to top]

Percentage of Participants Achieving HIV-1 RNA Below Limit of Quantification

Below limit of quantification was defined as less than 400 copies/milliliter (mL) (NCT00478231)
Timeframe: Baseline, Week 4, Week 8, Week 12, Week 24, Week 36, Week 48, Week 60, Week 72, Week 84 and Week 96 or EOT

InterventionPercentage of participants (Number)
BaselineWeek 4 (n= 202)Week 8 (n= 196)Week 12 (n= 190)Week 24 (n= 185)Week 36 (n= 171)Week 48 (n= 158)Week 60 (n= 141)Week 72 (n= 137)Week 84 (n= 125)Week 96 or EOT (n= 109)LOCF (n= 206)
Maraviroc2.429.743.942.641.147.441.136.224.823.233.926.7

[back to top]

Percentage of Participants With at Least 0.5 Log 10 Reduction in Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA)

(NCT00478231)
Timeframe: Week 4, Week 8, Week 12, Week 24, Week 36, Week 48, Week 60, Week 72, Week 84 and Week 96 or end of treatment (EOT)

InterventionPercentage of participants (Number)
Week 4 (n= 202)Week 8 (n= 196)Week 12 (n= 190)Week 24 (n= 185)Week 36 (n= 171)Week 48 (n= 158)Week 60 (n= 141)Week 72 (n= 137)Week 84 (n= 125)Week 96 or EOT (n= 109)LOCF (n= 206)
Maraviroc78.778.680.579.583.682.384.481.082.489.976.2

[back to top]

Percentage of Participants With at Least 1.0 Log 10 Reduction in HIV-1 RNA

(NCT00478231)
Timeframe: Week 4, Week 8, Week 12, Week 24, Week 36, Week 48, Week 60, Week 72, Week 84 and Week 96 or EOT

InterventionPercentage of participants (Number)
Week 4 (n= 202)Week 8 (n= 196)Week 12 (n= 190)Week 24 (n= 185)Week 36 (n= 171)Week 48 (n= 158)Week 60 (n= 141)Week 72 (n= 137)Week 84 (n= 125)Week 96 or EOT (n= 109)LOCF (n= 206)
Maraviroc67.868.971.168.669.070.370.264.271.272.561.2

[back to top]

The Primary Outcome of This Study is the Proportion of Patients Having Detectable HIV-1 RNA Using the Single Copy Assay After 48 Weeks of Treatment and the Study Hypothesis is That New Treatment is Better Than the Control Group.

(NCT00525733)
Timeframe: 48 weeks

Intervention# subjects without detectable viremia (Number)
3-drug Standard Therapy3
5-drug Experimental Therapy9

[back to top]

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 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 Plasma HIV-1 Viral Load From Baseline to Week 1

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

Change in Cardiovascular Risk Score From Baseline

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

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

[back to top]

Change in 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]

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]

Density Rate Per 1000 Participant-Years for Incidence of Myocardial Infarction or Ischemia: Events Adjudicated as 'Definite' or 'Possible'

Density rate per 1000 participant-years for incidence of myocardial infarction or ischemia (definite + possible) was reported. Events of myocardial infarction or ischemia were adjudicated as definite or possible; where definite= an event had definitely occurred; possible =an event had possibly occurred. Density rate was calculated as the number of events (n) per 1000 participant-years at risk. Poisson regressions were fit to the categorical events with only treatment in the model to obtain the crude rate and CI. In this outcome measure, density rate per 1000 participant-years for incidence of myocardial infarction or ischemia (which included sum of definite + possible events) was reported. (NCT00665561)
Timeframe: Up to 5 years following enrollment

Interventionevents per 1000 participant-years (Number)
Maraviroc Exposed4.80
Maraviroc Unexposed5.34

[back to top]

Density Rate Per 1000 Participant-Years for Incidence of Centers for Disease Control and Prevention Category C AIDS -Defining Opportunistic Infections

Density rate per 1000 participant-years for incidence of centers for disease control and prevention category C acquired immunodeficiency syndrome (AIDS) -defining opportunistic infections was reported. Density rate was calculated as the number of events (n) per 1000 participant-years at risk. Poisson regressions were fit to the categorical events with only treatment in the model to obtain the crude rate and confidence interval (CI). (NCT00665561)
Timeframe: Up to 5 years following enrollment

Interventionevents per 1000 participant-years (Number)
Maraviroc Exposed22.28
Maraviroc Unexposed41.77

[back to top]

Adjusted Density Rate Per 1000 Participant-Years for Incidence of Myocardial Infarction or Ischemia: Events Adjudicated as 'Definite' or 'Possible' or 'Insufficient Data'

Adjusted density rate per 1000 participant-years for incidence of myocardial infarction or ischemia (definite + possible + insufficient data) was reported. Events of myocardial infarction or ischemia were adjudicated as definite or possible or insufficient data; where definite= an event had definitely occurred; possible =an event had possibly occurred; insufficient =insufficient data to determine whether an event had occurred. Density rate was calculated as the number of events (n) per 1000 participant-years at risk. Poisson regressions were fit to the categorical events with treatment as the main effect, PS quartile and imputed FS as covariates in the model to obtain the adjusted rate and CI. In this outcome measure, adjusted density rate per 1000 participant-years for incidence of myocardial infarction or ischemia (which included sum of definite + possible events + insufficient data) was reported. (NCT00665561)
Timeframe: Up to 5 years following enrollment

Interventionevents per 1000 participant-years (Number)
Maraviroc Exposed5.65
Maraviroc Unexposed5.99

[back to top]

Adjusted Density Rate Per 1000 Participant-Years for Incidence of Myocardial Infarction or Ischemia: Events Adjudicated as 'Definite' or 'Possible'

Adjusted density rate per 1000 participant-years for incidence of myocardial infarction or ischemia (definite + possible) was reported. Events of myocardial infarction or ischemia were adjudicated as definite or possible; where definite= an event had definitely occurred; possible =an event had possibly occurred. Density rate was calculated as the number of events (n) per 1000 participant-years at risk. Poisson regressions were fit to the categorical events with treatment as the main effect, PS quartile and imputed FS as covariates in the model to obtain the adjusted rate and CI. In this outcome measure, adjusted density rate per 1000 participant-years for incidence of myocardial infarction or ischemia (which included sum of definite + possible events) was reported. (NCT00665561)
Timeframe: Up to 5 years following enrollment

Interventionevents per 1000 participant-years (Number)
Maraviroc Exposed2.08
Maraviroc Unexposed3.37

[back to top]

Adjusted Density Rate Per 1000 Participant-Years for Incidence of Death Due to Any Cause

Adjusted density rate per 1000 participant-years for incidence of death due to any cause was reported. Density rate was calculated as the number of events (n) per 1000 participant-years at risk. Poisson regressions were fit to the categorical events with treatment as the main effect, PS quartile and imputed FS as covariates in the model to obtain the adjusted rate and CI. (NCT00665561)
Timeframe: Up to 5 years following enrollment

Interventionevents per 1000 participant-years (Number)
Maraviroc Exposed14.39
Maraviroc Unexposed15.64

[back to top]

Density Rate Per 1000 Participant-Years for Incidence of Death Due to Any Cause

Density rate per 1000 participant-years for incidence of death due to any cause was reported. Density rate was calculated as the number of events (n) per 1000 participant-years at risk. Poisson regressions were fit to the categorical events with only treatment in the model to obtain the crude rate and CI. (NCT00665561)
Timeframe: Up to 5 years following enrollment

Interventionevents per 1000 participant-years (Number)
Maraviroc Exposed14.41
Maraviroc Unexposed18.33

[back to top] [back to top]

Adjusted Density Rate Per 1000 Participant-Years for Incidence of Centers for Disease Control and Prevention Category C Aids-Defining Opportunistic Infections

Adjusted density rate per 1000 participant-years for incidence of centers for disease control and prevention category c aids-defining opportunistic infections was reported. Density rate was calculated as the number of events (n) per 1000 participant-years at risk. Poisson regressions were fit to the categorical events with treatment as the main effect, propensity score (PS) quartile and imputed Framingham score (FS) as covariates in the model to obtain the adjusted rate and CI. (NCT00665561)
Timeframe: Up to 5 years following enrollment

Interventionevents per 1000 participant-years (Number)
Maraviroc Exposed22.36
Maraviroc Unexposed28.70

[back to top]

Density Rate Per 1000 Participant-Years for Incidence of Liver Failure

Density rate per 1000 participant-years for incidence of liver failure was reported. Density rate was calculated as the number of events (n) per 1000 participant-years at risk. Poisson regressions were fit to the categorical events with only treatment in the model to obtain the crude rate and CI. (NCT00665561)
Timeframe: Up to 5 years following enrollment

Interventionevents per 1000 participant-years (Number)
Maraviroc Exposed2.50
Maraviroc Unexposed2.55

[back to top]

Density Rate Per 1000 Participant-Years for Incidence of Myocardial Infarction or Ischemia: Events Adjudicated as 'Definite' or 'Possible' or 'Insufficient Data'

Density rate per 1000 participant-years for incidence of myocardial infarction or ischemia (definite + possible + insufficient data) was reported. Events of myocardial infarction or ischemia were adjudicated as definite or possible or insufficient data; where definite= an event had definitely occurred; possible =an event had possibly occurred; insufficient =insufficient data to determine whether an event had occurred. Density rate was calculated as the number of events (n) per 1000 participant-years at risk. Poisson regressions were fit to the categorical events with only treatment in the model to obtain the crude rate and CI. In this outcome measure, density rate per 1000 participant-years for incidence of myocardial infarction or ischemia (which included sum of definite + possible events + insufficient data) was reported. (NCT00665561)
Timeframe: Up to 5 years following enrollment

Interventionevents per 1000 participant-years (Number)
Maraviroc Exposed8.26
Maraviroc Unexposed7.43

[back to top]

Density Rate Per 1000 Participant-Years for Incidence of Rhabdomyolysis

Density rate per 1000 participant-years for incidence of rhabdomyolysis was reported. Density rate was calculated as the number of events (n) per 1000 participant-years at risk. Poisson regressions were fit to the categorical events with only treatment in the model to obtain the crude rate and CI. (NCT00665561)
Timeframe: Up to 5 years following enrollment

Interventionevents per 1000 participant-years (Number)
Maraviroc Exposed0.77
Maraviroc Unexposed0.70

[back to top]

Density Rate Per 1000 Participant-Years for Incidence of Viral Encephalitis

Density rate per 1000 participant-years for incidence of viral encephalitis was reported. Density rate was calculated as the number of events (n) per 1000 participant-years at risk. Poisson regressions were fit to the categorical events with only treatment in the model to obtain the crude rate and CI. (NCT00665561)
Timeframe: Up to 5 years following enrollment

Interventionevents per 1000 participant-years (Number)
Maraviroc Exposed0.38
Maraviroc Unexposed0.46

[back to top]

Percentage of Participants With All-Cause Mortality

All-cause death was defined as the death due to any cause during the course of study. (NCT00665561)
Timeframe: Up to 5 years following enrollment

InterventionPercentage of participants (Number)
Maraviroc Exposed5.7
Maraviroc Unexposed7.0

[back to top]

Adjusted Density Rate Per 1000 Participant-Years for Incidence of All Malignancies (AIDS Defining Malignancies and Non-AIDS Defining Malignancies)

Adjusted density rate per 1000 participant-years for incidence of all malignancies as well as its types (categorized as AIDS defining malignancies and non-AIDS defining malignancies) were reported. AIDS defining malignancies included malignancies due to any of these: cervical cancer, Kaposi's sarcoma or lymphoma; whereas all other malignancies (except AIDS-defining) were non-AIDS defining malignancies. Density rate was calculated as the number of events (n) per 1000 participant-years at risk. Poisson regressions were fit to the categorical events with treatment as the main effect, PS quartile and imputed FS as covariates in the model to obtain the adjusted rate and CI. (NCT00665561)
Timeframe: Up to 5 years following enrollment

,
Interventionevents per 1000 participant-years (Number)
All MalignanciesAIDS defining MalignanciesNon-AIDS defining Malignancies
Maraviroc Exposed12.492.319.87
Maraviroc Unexposed13.543.319.86

[back to top]

Density Rate Per 1000 Participant-Years for Incidence of All Malignancies (AIDS Defining Malignancies and Non-AIDS Defining Malignancies)

Density rate per 1000 participant-years for incidence of all malignancies as well as its types (categorized as: AIDS defining malignancies and non-AIDS defining malignancies) were reported. AIDS defining malignancies included malignancies due to any of these: cervical cancer, Kaposi's sarcoma or lymphoma; whereas all other malignancies (except AIDS-defining) were non-aids defining malignancies. Density rate was calculated as the number of events (n) per 1000 participant-years at risk. Poisson regressions were fit to the categorical events with only treatment in the model to obtain the crude rate and CI. (NCT00665561)
Timeframe: Up to 5 years following enrollment

,
Interventionevents per 1000 participant-years (Number)
All MalignanciesAIDS defining MalignanciesNon-AIDS defining Malignancies
Maraviroc Exposed12.872.3110.56
Maraviroc Unexposed13.693.719.98

[back to top]

Maraviroc Pharmacokinetic (PK) Parameter: 12-Hour Trough Concentration (C12)

Effect of raltegravir on pharmacokinetics of maraviroc (comparison of C12 of raltegravir co-administered with maraviroc (Test) versus maraviroc administered alone (Reference)). Pharmacokinetics were assessed on Day 11 (maraviroc) and Day 14 (maraviroc and raltegravir). C12 is the 12-hour trough concentration. (NCT00666705)
Timeframe: Days 11 and 14

Interventionng/mL (Mean)
Maraviroc + Raltegravir (Test)48.34
Maraviroc (Reference)53.36

[back to top]

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

Effect of raltegravir on pharmacokinetics of maraviroc (comparison of Cmax of raltegravir co-administered with maraviroc (Test) versus maraviroc administered alone (Reference)). Pharmacokinetics were assessed on Day 11 (maraviroc) and Day 14 (maraviroc and raltegravir). Cmax is the maximum plasma concentration. (NCT00666705)
Timeframe: Days 11 and 14

Interventionng/mL (Mean)
Maraviroc + Raltegravir (Test)691.6
Maraviroc (Reference)851.4

[back to top]

Raltegravir Pharmacokinetics (PK) Parameter: 12-Hour Trough Concentration (C12)

Effect of maraviroc on pharmacokinetics of raltegravir (comparison of C12 of raltegravir co-administered with maraviroc (Test) vs. raltegravir administered alone (Reference)). Pharmacokinetics were assessed on Day 3 (raltegravir) and Day 14 (maraviroc and raltegravir). C12 is the 12-hour trough concentration. (NCT00666705)
Timeframe: Days 3 and 14

Interventionng/mL (Mean)
Maraviroc + Raltegravir (Test)51.16
Raltegravir (Reference)73.69

[back to top]

Raltegravir Pharmacokinetics (PK) Parameter: Area Under the Plasma Concentration-time Profile Over the Dosing Interval (AUCτ)

Effect of maraviroc on pharmacokinetics of raltegravir (comparison of AUCτ of raltegravir co-administered with maraviroc (Test) versus raltegravir administered alone (Reference)). Pharmacokinetics were assessed on Day 3 (raltegravir) and Day 14 (maraviroc and raltegravir). (NCT00666705)
Timeframe: Days 3 and 14

Interventionng.hr/mL (Mean)
Maraviroc + Raltegravir (Test)6287.5
Raltegravir (Reference)9122.4

[back to top]

Maraviroc Pharmacokinetic (PK) Parameter: Area Under the Plasma Concentration-time Profile Over the Dosing Interval (AUCτ)

Effect of raltegravir on pharmacokinetics of maraviroc (comparison of AUCτ of raltegravir co-administered with maraviroc (Test) versus maraviroc administered alone (Reference)). Pharmacokinetics were assessed on Day 11 (maraviroc) and Day 14 (maraviroc and raltegravir). (NCT00666705)
Timeframe: Days 11 and 14

Interventionng.hr/mL (Mean)
Maraviroc + Raltegravir (Test)2551.9
Maraviroc (Reference)2971.6

[back to top]

Raltegravir Pharmacokinetics (PK) Parameter: Maximum Concentration (Cmax)

Effect of maraviroc on pharmacokinetics of raltegravir (comparison of Cmax of raltegravir co-administered with maraviroc (Test) versus raltegravir administered alone (Reference)). Pharmacokinetics assessed on Day 3 (raltegravir) and Day 14 (maraviroc and raltegravir). Cmax is the maximum plasma concentration. (NCT00666705)
Timeframe: Days 3 and 14

Interventionng/mL (Mean)
Maraviroc + Raltegravir (Test)2169.6
Raltegravir (Reference)3026.6

[back to top]

Change in IL-6, MCP-1, MCP-2, and Plasma CD40L

Change was calculated as week 36 result minus the week 24 result (average of the week 22 and week 24 values). (NCT00709111)
Timeframe: From week 24 to week 36

Interventionpg/ml (Median)
IL-6MCP-1MCP-2CD40L
Maraviroc-0.9-6.90.10

[back to top]

Change in IL-6, MCP-1, MCP-2, and Plasma CD40L

Change was calculated as week 48 result (average of week 46 and week 48) minus the week 24 result (average of the week 22 and week 24 values). (NCT00709111)
Timeframe: From week 24 to week 48

Interventionpg/ml (Median)
IL-6MCP-1MCP-2CD40L
Maraviroc-0.57.70.232.3

[back to top]

Change in Intercellular Cell Adhesion Molecule (ICAM)-1, Plasma P-selectin, Soluble TNFRII (sTNFRII), and Matrix Metalloproteinase (MMP)-9

Change was calculated as the week 24 result (average of the week 22 and week 24 values) minus the baseline result (average of pre-entry and entry values). (NCT00709111)
Timeframe: From baseline to week 24

Interventionng/ml (Median)
ICAM-1P-selectinsTNFRIIMMP-9
Maraviroc-25.7-11.70.18-12.5

[back to top]

Change in Interleukin (IL)-6, Monocyte Chemoattractant Protein (MCP)-1, MCP-2, and Plasma CD40 Ligand (CD40L)

Change was calculated as the week 24 result (average of the week 22 and week 24 values) minus the baseline result (average of pre-entry and entry values). (NCT00709111)
Timeframe: From baseline to week 24

Interventionpg/ml (Median)
IL-6MCP-1MCP-2CD40L
Maraviroc0.744.6-1.4-1.8

[back to top]

Change in Percentage of CD4+ T-cells That Are: naïve (%CD45RA+CCR7+), Central Memory (%CD45RA-CCR7+), Effector Memory (%CD45RA-CCR7-), Effector (%CD45RA+CCR7-), %HLA-DR+CD38+, %CD38+, %Ki67+, %caspase3+, %Bcl-2-, and %CD57+

Change was calculated as the week 24 result (average of the week 22 and week 24 values) minus the baseline result (average of pre-entry and entry values). (NCT00709111)
Timeframe: From baseline to week 24

Intervention% of CD4+ T-cells (Median)
naïve (%CD45RA+CCR7+)central memory (%CD45RA-CCR7+)effector memory (%CD45RA-CCR7-)effector (%CD45RA+CCR7-)%HLA-DR+CD38+%CD38+%Ki67+%caspase3+%Bcl-2-%CD57+
Maraviroc-1.3-4.85.80.7-1.3-14.8-1.0-1.10.71.8

[back to top]

Change in Percentage of CD4+ T-cells That Are: naïve (%CD45RA+CCR7+), Central Memory (%CD45RA-CCR7+), Effector Memory (%CD45RA-CCR7-), Effector (%CD45RA+CCR7-), %HLA-DR+CD38+, %CD38+, %Ki67+, %caspase3+, %Bcl-2-, and %CD57+

Change was calculated as week 36 result minus the week 24 result (average of the week 22 and week 24 values). (NCT00709111)
Timeframe: From week 24 to week 36

Intervention% of CD4+ T-cells (Median)
naïve (%CD45RA+CCR7+)central memory (%CD45RA-CCR7+)effector memory (%CD45RA-CCR7-)effector (%CD45RA+CCR7-)%HLA-DR+CD38+%CD38+%Ki67+%caspase3+%Bcl-2-%CD57+
Maraviroc3.5-1.4-3.70.1-0.22.80.10.3-0.6-0.9

[back to top]

Change in Percentage of CD4+ T-cells That Are: naïve (%CD45RA+CCR7+), Central Memory (%CD45RA-CCR7+), Effector Memory (%CD45RA-CCR7-), Effector (%CD45RA+CCR7-), %HLA-DR+CD38+, %CD38+, %Ki67+, %caspase3+, %Bcl-2-, and %CD57+

Change was calculated as week 48 result (average of week 46 and week 48) minus the week 24 result (average of the week 22 and week 24 values). (NCT00709111)
Timeframe: From week 24 to week 48

Intervention% of CD4+ T-cells (Median)
naïve (%CD45RA+CCR7+)central memory (%CD45RA-CCR7+)effector memory (%CD45RA-CCR7-)effector (%CD45RA+CCR7-)%HLA-DR+CD38+%CD38+%Ki67+%caspase3+%Bcl-2-%CD57+
Maraviroc2.1-3.10.41.20.47.10.10.7-0.5-0.4

[back to top]

Change in Percentage of CD8+ T-cells That Are: naïve (%CD45RA+CCR7+), Central Memory (%CD45RA-CCR7+), Effector Memory (%CD45RA-CCR7-), Effector (%CD45RA+CCR7-), %HLA-DR+CD38+, %CD38+, %Ki67+, %caspase3+, %Bcl-2-, and %CD57+

Change was calculated as the week 24 result (average of the week 22 and week 24 values) minus the baseline result (average of pre-entry and entry values). (NCT00709111)
Timeframe: From baseline to week 24

Intervention% of CD8+ T-cells (Median)
naïve (%CD45RA+CCR7+)central memory (%CD45RA-CCR7+)effector memory (%CD45RA-CCR7-)effector (%CD45RA+CCR7-)%HLA-DR+CD38+%CD38+%Ki67+%caspase3+%Bcl-2-%CD57+
Maraviroc-3.3-1.02.52.0-1.4-14.2-0.1-0.70.53.6

[back to top]

Change in Percentage of CD8+ T-cells That Are: naïve (%CD45RA+CCR7+), Central Memory (%CD45RA-CCR7+), Effector Memory (%CD45RA-CCR7-), Effector (%CD45RA+CCR7-), %HLA-DR+CD38+, %CD38+, %Ki67+, %caspase3+, %Bcl-2-, and %CD57+

Change was calculated as week 36 result minus the week 24 result (average of the week 22 and week 24 values). (NCT00709111)
Timeframe: From week 24 to week 36

Intervention% of CD8+ T-cells (Median)
naïve (%CD45RA+CCR7+)central memory (%CD45RA-CCR7+)effector memory (%CD45RA-CCR7-)effector (%CD45RA+CCR7-)%HLA-DR+CD38+%CD38+%Ki67+%caspase3+%Bcl-2-%CD57+
Maraviroc2.4-0.3-5.12.4-0.8-1.60.10.2-0.3-2.8

[back to top]

Change in Percentage of CD8+ T-cells That Are: naïve (%CD45RA+CCR7+), Central Memory (%CD45RA-CCR7+), Effector Memory (%CD45RA-CCR7-), Effector (%CD45RA+CCR7-), %HLA-DR+CD38+, %CD38+, %Ki67+, %caspase3+, %Bcl-2-, and %CD57+

Change was calculated as week 48 result (average of week 46 and week 48) minus the week 24 result (average of the week 22 and week 24 values). (NCT00709111)
Timeframe: From week 24 to week 48

Intervention% of CD8+ T-cells (Median)
naïve (%CD45RA+CCR7+)central memory (%CD45RA-CCR7+)effector memory (%CD45RA-CCR7-)effector (%CD45RA+CCR7-)%HLA-DR+CD38+%CD38+%Ki67+%caspase3+%Bcl-2-%CD57+
Maraviroc1.1-0.5-0.10.40.14.20.20.50-1.4

[back to top]

Drug Adherence Assessed as Number of Missed Doses Over a 4-day Recall

Self-reported MVC adherence data were based on a four-day (8 expected doses) recall. Based on the wording of the Self Report case report form (CRF), participants reporting that they were currently taking MVC that then failed to complete the record of the number of missed doses were assumed to have no missed doses to report. Missing adherence assessments at a time point of interest were ignored and only those participants completing an adherence assessment at least one time point of interest were included. (NCT00709111)
Timeframe: At weeks 4, 12, and 24

Interventiondoses missed (Median)
Week 4 (N=30)Week 12 (N=28)Week 24 (N=27)
Maraviroc000

[back to top]

Number of Subjects Who Experience a Grade 2, 3 or 4 Signs and Symptoms, Grade 3 or 4 Laboratory Abnormalities, or Death.

Events with date of onset or specimen date prior to first dose of MVC or after the last dose of MVC were excluded. Signs and symptoms with a date of onset the same as the first dose of MVC were excluded if confirmed by the site to be before the first dose. Lab abnormalities with the date of specimen the same as the date of the first dose of MVC were excluded on the assumption that the specimen was drawn before the first dose. Grading used the Division of AIDS (DAIDS) 2004 Severity of Adverse Events Tables, where Grade 1=Mild, 2=Moderate, 3=Severe, 4=Potentially life-threatening. (NCT00709111)
Timeframe: From baseline through week 24

Interventionparticipants (Number)
grade>=2 signs/symptoms or grade>=3 lab abnorm.deaths
Maraviroc150

[back to top]

Proportion of Participants With Detectable HIV-1 Viremia as Measured by Single Copy Assay (SCA)

A subject was considered detectable at a specific week if HIV-1 RNA by SCA >=1 copy/ml. (NCT00709111)
Timeframe: At weeks -1 (pre-entry), 0 (entry), 12, 22, 24, and 36

Interventionproportion of participants (Number)
Week -1 (N=31)Week 0 (N=31)Week 12 (N=30)Week 22 (N=31)Week 24 (N=29)Week 36 (N=30)
Maraviroc0.350.320.430.290.480.43

[back to top]

Change in ICAM-1, Plasma P-selectin, sTNFRII, and MMP-9

Change was calculated as week 36 result minus the week 24 result (average of the week 22 and week 24 values). (NCT00709111)
Timeframe: From week 24 to week 36

Interventionng/ml (Median)
ICAM-1P-selectinsTNFRIIMMP-9
Maraviroc-20.9-13.7-0.0511.3

[back to top]

Change in D-dimer

Change was calculated as the week 24 result (average of the week 22 and week 24 values) minus the baseline result (average of pre-entry and entry values). (NCT00709111)
Timeframe: From baseline to week 24

Interventionmcg/ml (Median)
Maraviroc0.09

[back to top]

Change From Within-subject Pre-treatment CD4+ T-cell Count Slopes to Corresponding Within-subject CD4+ T-cell Count Slopes From Baseline Through Week 24

The estimated mean change in slopes was summarized across the population by using generalized estimating equations. Pre-treatment CD4+ T-cell counts (from at least 48 weeks prior to study entry) were recorded at screening from patient source documentation. Baseline was defined as the average of pre-entry and entry values. Week 24 was defined as the average of the week 22 and week 24 values. (NCT00709111)
Timeframe: From pre-treatment through week 24

Interventioncells/mm^3/year (Mean)
Maraviroc25.2

[back to top]

Change in CD4 Percentage

Change was calculated as the week 24 CD4 percentage (average of the week 22 and week 24 values) minus the baseline CD4 percentage (average of pre-entry and entry values). (NCT00709111)
Timeframe: From baseline to week 24

Intervention% of total lymphocytes (Median)
Maraviroc0.5

[back to top]

Change in CD4 Percentage

Change was calculated as week 36 CD4 percentage minus the week 24 CD4 percentage (average of the week 22 and week 24 values). (NCT00709111)
Timeframe: From week 24 to week 36

Intervention% of total lymphocytes (Median)
Maraviroc0.2

[back to top]

Change in CD4 Percentage

Change was calculated as week 48 CD4 percentage (average of week 46 and week 48) minus the week 24 CD4 percentage (average of the week 22 and week 24 values). (NCT00709111)
Timeframe: From week 24 to week 48

Intervention% of total lymphocytes (Median)
Maraviroc0.5

[back to top]

Change in ICAM-1, Plasma P-selectin, sTNFRII, and MMP-9

Change was calculated as week 48 result (average of week 46 and week 48) minus the week 24 result (average of the week 22 and week 24 values). (NCT00709111)
Timeframe: From week 24 to week 48

Interventionng/ml (Median)
ICAM-1P-selectinsTNFRIIMMP-9
Maraviroc-32.4-17.30.0312.8

[back to top]

Change in CD4+ T-cell Count

Change was calculated as the week 24 CD4+ T-cell count (average of the week 22 and week 24 values) minus the baseline CD4+ T-cell count (average of pre-entry and entry values). (NCT00709111)
Timeframe: From baseline to week 24

Interventioncells/mm^3 (Median)
Maraviroc12

[back to top]

Change in CD4+ T-cell Count

Change was calculated as week 36 CD4+ T-cell count minus the week 24 CD4+ T-cell count (average of the week 22 and week 24 values). (NCT00709111)
Timeframe: From week 24 to week 36

Interventioncells/mm^3 (Median)
Maraviroc-3

[back to top]

Change in CD4+ T-cell Count

Change was calculated as week 48 CD4+ T-cell count (average of week 46 and week 48) minus the week 24 CD4+ T-cell count (average of the week 22 and week 24 values). (NCT00709111)
Timeframe: From week 24 to week 48

Interventioncells/mm^3 (Median)
Maraviroc7

[back to top]

Change in D-dimer

Change was calculated as week 36 result minus the week 24 result (average of the week 22 and week 24 values). (NCT00709111)
Timeframe: From week 24 to week 36

Interventionmcg/ml (Median)
Maraviroc0.01

[back to top]

Change in D-dimer

Change was calculated as week 48 result (average of week 46 and week 48) minus the week 24 result (average of the week 22 and week 24 values). (NCT00709111)
Timeframe: From week 24 to week 48

Interventionmcg/ml (Median)
Maraviroc-0.02

[back to top]

Change in High Sensitivity C-reactive Protein (Hs-CRP)

Change was calculated as the week 24 result (average of the week 22 and week 24 values) minus the baseline result (average of pre-entry and entry values). (NCT00709111)
Timeframe: From baseline to week 24

Interventionmg/dl (Median)
Maraviroc0.01

[back to top]

Change in Hs-CRP

Change was calculated as week 36 result minus the week 24 result (average of the week 22 and week 24 values). (NCT00709111)
Timeframe: From week 24 to week 36

Interventionmg/dl (Median)
Maraviroc-0.01

[back to top]

Change From Within-subject Pre-treatment CD4 Percentage Slopes to Corresponding Within-subject CD4 Percentage Slopes From Baseline Through Week 24

The estimated mean change in slopes was summarized across the population by using generalized estimating equations. Pre-treatment CD4 percentage (from at least 48 weeks prior to study entry) were recorded at screening from patient source documentation. Baseline was defined as the average of pre-entry and entry values. Week 24 was defined as the average of the week 22 and week 24 values. (NCT00709111)
Timeframe: From pre-treatment through week 24

Intervention% of total lymphocytes/year (Mean)
Maraviroc-1.0

[back to top]

Change in Hs-CRP

Change was calculated as week 48 result (average of week 46 and week 48) minus the week 24 result (average of the week 22 and week 24 values). (NCT00709111)
Timeframe: From week 24 to week 48

Interventionmg/dl (Median)
Maraviroc0.01

[back to top]

Change in Soluble CD14

Change was calculated as the week 24 result (average of the week 22 and week 24 values) minus the baseline result (average of pre-entry and entry values). Soluble CD14 is a marker of gut microbial translocation. (NCT00709111)
Timeframe: From baseline to week 24

Interventionmcg/ml (Median)
Maraviroc-0.03

[back to top]

Change in Soluble CD14

Change was calculated as week 36 result minus the week 24 result (average of the week 22 and week 24 values). Soluble CD14 is a marker of gut microbial translocation. (NCT00709111)
Timeframe: From week 24 to week 36

Interventionmcg/ml (Median)
Maraviroc-0.17

[back to top]

Change in Soluble CD14

Change was calculated as week 48 result (average of week 46 and week 48) minus the week 24 result (average of the week 22 and week 24 values). Soluble CD14 is a marker of gut microbial translocation. (NCT00709111)
Timeframe: From week 24 to week 48

Interventionmcg/ml (Median)
Maraviroc-0.16

[back to top]

Proportion of Participants Achieving a 50-cell Increase in CD4+ T-cell Count

Baseline was defined as the average of pre-entry and entry values. Week 24 was defined as the average of the week 22 and week 24 values. (NCT00709111)
Timeframe: From baseline to week 24

Interventionproportion of participants (Number)
Maraviroc0.06

[back to top]

Within-subject CD4 Percentage Slopes

The estimated mean slope was summarized across the population by using generalized estimating equations. Baseline was defined as the average of pre-entry and entry values. Week 24 was defined as the average of the week 22 and week 24 values. (NCT00709111)
Timeframe: From baseline through week 24

Intervention% of total lymphocytes/year (Mean)
Maraviroc-0.3

[back to top]

Within-subject CD4+ T-cell Count Slopes

The estimated mean slope was summarized across the population by using generalized estimating equations. Baseline was defined as the average of pre-entry and entry values. Week 24 was defined as the average of the week 22 and week 24 values. (NCT00709111)
Timeframe: From baseline through week 24

Interventioncells/mm^3/year (Mean)
Maraviroc24.7

[back to top]

Renal Clearance (CLR) in Subjects With Normal, Mild, Moderate and Severe Renal Function

Renal clearance (CLR) measured in milliliters per minute (mL/min). (NCT00717067)
Timeframe: Hour 0 (prior to MVC dosing [single dose] or prior to last MVC dose [multiple dose]) to 72 hours post-dose ; hours 0, 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48, and 72.

InterventionmL/min (Geometric Mean)
Healthy Subjects: Multiple Dose105.7
Mild Renal Impairment: Multiple Dose77.2
Moderate Renal Impairment: Multiple Dose62.5
Healthy Subjects: Single Dose110.0
Severe Renal Impairment: Single Dose26.6

[back to top]

Maximum Observed Plasma Concentration (Cmax)

Maximum observed plasma concentration (Cmax) within the dosing interval; measured in nanograms per milliliter (ng/mL). (NCT00717067)
Timeframe: Pre-dose, post-dose hours 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48, and 72.

Interventionng/mL (Geometric Mean)
Healthy Subjects: Multiple Dose950.91
Mild Renal Impairment: Multiple Dose1150.74
Moderate Renal Impairment: Multiple Dose674.20
Healthy Subjects: Single Dose335.60
Severe Renal Impairment: Single Dose801.16
ESRD: Single Dose; After Dialysis576.7
ESRD: Single Dose; Before Dialysis478.5

[back to top]

Hemodialysis Clearance of Maraviroc (MVC) in Subjects With End Stage Renal Disease (ESRD) Undergoing Hemodialysis: CLdD

CLdD: dialysate clearance before dialysis; measured in milliliters per minute. (NCT00717067)
Timeframe: Before dialysis

InterventionmL/min (Geometric Mean)
ESRD: Single Dose; Before Dialysis36.42

[back to top]

Half-life (t1/2)

Elimination half-life (t1/2) measured in hours: time required for half the quantity of maraviroc to be metabolized or eliminated by normal biological processes. (NCT00717067)
Timeframe: Pre-dose, post-dose hours 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48, and 72.

Interventionhour (Mean)
Healthy Subjects: Multiple Dose14.22
Mild Renal Impairment: Multiple Dose16.84
Moderate Renal Impairment: Multiple Dose16.99
Healthy Subjects: Single Dose14.36
Severe Renal Impairment: Single Dose17.29
ESRD: Single Dose; After Dialysis15.03
ESRD: Single Dose; Before Dialysis13.86

[back to top]

Derivation of Renal Clearance in Subjects With Normal, Mild, Moderate and Severe Renal Function: Ae

Ae: amount of drug excreted unchanged in the urine; measured in milligrams (mg). (NCT00717067)
Timeframe: Hour 0 (prior to MVC dosing [single dose] or prior to last MVC dose [multiple dose]) to 72 hours post-dose ; hours 0, 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48, and 72.

Interventionmg (Mean)
Healthy Subjects: Multiple Dose41.6
Mild Renal Impairment: Multiple Dose39.1
Moderate Renal Impairment: Multiple Dose24.9
Healthy Subjects: Single Dose10.8
Severe Renal Impairment: Single Dose8.4

[back to top]

AUCtau

AUCtau: area under the plasma concentration-time profile from time zero to the end of the dosing interval (tau); measured in nanograms * hours divided by milliliters (ng.hr/mL). (NCT00717067)
Timeframe: Pre-dose, post-dose hours 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48, and 72.

Interventionng*hr/mL (Geometric Mean)
Healthy Subjects: Multiple Dose5341.4
Mild Renal Impairment: Multiple Dose8118.7
Moderate Renal Impairment: Multiple Dose6193.3

[back to top]

Safety and Tolerability of Maraviroc in the Absence and Presence of a Potent CYP3A4 Inhibitor in Subjects With Various Degrees of Renal Impairment or Undergoing Hemodialysis: Number of Subjects With Maximum EGC QTC, QTCB and QTCF Intervals

Single 12-lead ECG: number of subjects with maximum QTC interval, maximum QTCB interval (Bazett's correction), and maximum QTCF interval (Friderica's correction) measured in milliseconds (msec); range: 450 to <480 msec, 480 to <500 msec, and >500 msec. Maximum QTC interval increase from Baseline; citeria: change = ≥ 30 msec to < 60 msec, and change = ≥ 60 msec. (NCT00717067)
Timeframe: Normal renal function: screening, Day -3 and Day -1; normal renal function, mild and moderate RI: Day 7 to Day 9 and follow-up; severe RI: screening, Day 1, Day 3, Day 4, and follow-up; ESRD: screening, Day 1, Day 3, Day 4, and follow-up

,,,,,,
Interventionsubjects (Number)
Maximum QTC Interval: 450 to < 480 msecMaximum QTC Interval: 480 to < 500 msecMaximum QTC Interval: > 500 msecMaximum QTCB Interval: 450 to < 480 msecMaximum QTCB Interval: 480 to < 500 msecMaximum QTCB Interval: > 500 msecMaximum QTCF Interval: 450 to 480 msecMaximum QTCF Interval: 480 to < 500 msecMaximum QTCF Interval: > 500 msecMax. QTC Interval Increase from BL: change ≥30 <60QTC Interval Increase from BL: change ≥ 60
ESRD: Single Dose; After Dialysis10000000000
ESRD: Single Dose; Before Dialysis00000000010
Healthy Subjects: Multiple Dose10010010000
Healthy Subjects: Single Dose10010000000
Mild Renal Impairment: Multiple Dose00000000000
Moderate Renal Impairment: Multiple Dose00000000000
Severe Renal Impairment: Single Dose20010010000

[back to top]

Safety and Tolerability of Maraviroc in the Absence and Presence of a Potent CYP3A4 Inhibitor in Subjects With Various Degrees of Renal Impairment or Undergoing Hemodialysis: Number of Subjects With Maximum Increase and Decrease in Supine Blood Pressure

Number of subjects with absolute values of supine systolic blood pressure (BP) measured in millimeters of mercury (mm/Hg), range: <90 mmHg; and supine diastolic blood pressure, range: <50 mmHg. Number of subjects with a maximum increase and decrease from Baseline in supine systolic BP ≥ 30 mmHg. Number of subjects with a maximum increase and decrease from Baseline in supine diastolic BP ≥ 20 mmHg. (NCT00717067)
Timeframe: Normal renal function: screening, Day -3 to Day -1; normal, mild and moderate RI: Day 7 to Day 10 and follow-up; severe RI: Day 1 to Day 4 and follow-up; ESRD: Day 1, Day 4, and follow-up

,,,,,,
Interventionsubjects (Number)
BL Supine Systolic Blood Pressure (BP) <90 mmHgMaximum Increase from BL: Supine Systolic BP≥ 30Maximum Decrease from BL: Supine Systolic BP ≥ 30BL Supine Diastolic Blood Pressure (BP) <50 mmHgMaximum Increase from BL: Supine Diastolic BP≥ 20Maximun Decrease from BL: Supine Diastolic BP ≥20
ESRD: Single Dose; After Dialysis001000
ESRD: Single Dose; Before Dialysis000000
Healthy Subjects: Multiple Dose000000
Healthy Subjects: Single Dose000000
Mild Renal Impairment: Multiple Dose000000
Moderate Renal Impairment: Multiple Dose000010
Severe Renal Impairment: Single Dose000000

[back to top]

Area Under the Plasma Concentration Time-curve From Zero to the Last Measured Concentration (AUClast)

Area under the plasma concentration time-curve from zero to the last measured concentration (AUClast) measured in nanograms * hour divided by milliliters (ng*hr/mL). (NCT00717067)
Timeframe: Pre-dose, post-dose hours 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48, and 72.

Interventionng*hr/mL (Geometric Mean)
Healthy Subjects: Multiple Dose7356.3
Mild Renal Impairment: Multiple Dose9502.1
Moderate Renal Impairment: Multiple Dose6496.0
Healthy Subjects: Single Dose1320.7
Severe Renal Impairment: Single Dose4255.5
ESRD: Single Dose; After Dialysis2636.5
ESRD: Single Dose; Before Dialysis2770.1

[back to top]

Safety and Tolerability of Maraviroc in the Absence and Presence of a Potent CYP3A4 Inhibitor in Subjects With Various Degrees of Renal Impairment or Undergoing Hemodialysis: Number of Subjects With Pulse Rate < 40 and > 120 Beats Per Minute

Number of subjects with pulse rate < 40 beats per minute (BPM), number of subjects with pulse rate > 120 BPM. (NCT00717067)
Timeframe: Normal renal function: screening, Day -3 to Day -1; normal, mild and moderate RI: Day 7 to Day 10 and follow-up; severe RI: Day 1 to Day 4 and follow-up; ESRD: Day 1, Day 4, and follow-up

,,,,,,
Interventionbpm (Number)
Supine Pulse Rate <40 BPMSupine Pulse Rate >120 BPM
ESRD: Single Dose; After Dialysis00
ESRD: Single Dose; Before Dialysis00
Healthy Subjects: Multiple Dose00
Healthy Subjects: Single Dose00
Mild Renal Impairment: Multiple Dose00
Moderate Renal Impairment: Multiple Dose00
Severe Renal Impairment: Single Dose00

[back to top]

Plasma Protein Binding

Percent protein binding (protein unbound maraviroc (MVC) fraction [percent free]) was determined by rapid equilibrium dialysis. Percent free = 100 - percent bound. (NCT00717067)
Timeframe: 2 hours post-dose; normal Day -3 and Day 7; mild moderate: Day 7; severe and ESRD: Day 1

,,,,,
Interventionpercent free (Number)
minimum protein unbound MVC fractionmaximum protein unbound MVC fraction
ESRD18.227.8
Healthy Subjects: Multiple Dose19.726.6
Healthy Subjects: Single Dose14.628.2
Mild Renal Impairment: Multiple Dose15.329.1
Moderate Renal Impairment: Multiple Dose18.131.6
Severe Renal Impairment: Single Dose19.228.1

[back to top]

Time of First Occurrence (Tmax)

Time (hours) of first occurrence (Tmax); time after dosing when Cmax (maximum plasma concentration) occured. (NCT00717067)
Timeframe: Pre-dose, post-dose hours 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48, and 72.

Interventionhours (Median)
Healthy Subjects: Multiple Dose1.000
Mild Renal Impairment: Multiple Dose1.500
Moderate Renal Impairment: Multiple Dose2.000
Healthy Subjects: Single Dose2.500
Severe Renal Impairment: Single Dose2.500
ESRD: Single Dose; After Dialysis3.000
ESRD: Single Dose; Before Dialysis2.000

[back to top]

Area Under the Time Curve From 0 to Infinity (AUCinf)

Area under the plasma concentration-time profile from time zero to the time infinate in subjects who received single dose treatment; measured in nanograms * hour divided by millilters (ng*hr/mL). (NCT00717067)
Timeframe: Pre-dose, post-dose hours 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48, and 72

Interventionng*hr/mL (Geometric Mean)
Healthy Subjects: Single Dose1348.4
Severe Renal Impairment: Single Dose4367.7
ESRD: Single Dose; After Dialysis2677.4
ESRD: Single Dose; Before Dialysis2805.5

[back to top]

Change in CD4+ T Cell Count

(NCT00735072)
Timeframe: Baseline and Week 24

Interventioncells/mm^3 over 24 weeks (Mean)
Maraviroc17
Placebo17

[back to top]

Week 24 Change in Percentage of CD8+ T Cells That Co-express CD38 and HLA DR (Week 24 %CD38+HLA-DR+ CD8+ T Cells Minus Baseline %CD38+HLA-DR+ CD8+ T Cells)

(NCT00735072)
Timeframe: Baseline and Week 24

Intervention%CD38+ HLA-DR+ CD8+ T cells (Median)
Maraviroc2.2
Placebo-0.7

[back to top]

Change in Ultra-sensitive Plasma HIV RNA Level (Single Copy/ml Assay)

(NCT00735072)
Timeframe: Baseline and Week 24

Interventionpercent change from baseline (Mean)
Maraviroc-52
Placebo-48

[back to top]

Change in Gut-associated Lymphoid Tissue HIV RNA Level (UCSF Site Only)

(NCT00735072)
Timeframe: Baseline and Week 24

Interventionpercent change from baseline (Median)
Maraviroc-11
Placebo-3

[back to top]

Change in Brachial Artery Flow-mediated Dilatation (UCSF Site Only)

(NCT00735072)
Timeframe: Baseline and Week 24

Interventionpercent change from baseline (Mean)
Maraviroc1.0
Placebo0.3

[back to top]

AUC: Steady-state Plasma MVC PK Following Administration of RTV

Amprenavir (APV) is the active ingredient/ metabolite of Fosamprenavir (FPV). MVC minimum concentration (Cmin), maximum concentration (Cmax), and area under the plasma concentration-time curve (AUC), as determined from MVC concentrations observed in blood samples obtained at baseline, and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours during the period when MVC 300mg BID was administered with the FPV-Containing BID regimens (FPV 1400mg BID, FPV 700mg/RTV 100 mg BID), and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 24 hours during the period when MVC 300mg BID was administered with the FPV QD regimen (FPV 1400mg/RTV 100mg QD). As Groups A and B received the same regimens (albeit in different order), PK data for these two groups were collated, then assessed. For the same reason, PK data from Groups C and D regimens were collated before assessment, as were the PK data from Groups E and F. (NCT00764465)
Timeframe: Day 7 of the MVC 300mg BID regimen and Day 14 of the MVC 300mg/FPV 1400mg BID, MVC 300mg/FPV 700mg/RTV 100mg BID, and MVC 300mg BID Plus FPV 1400mg/RTV 100mg QD regimens

Interventionng•h/mL (Mean)
Group A & B0.87
Group C & D0.34
Group E & F0.98

[back to top]

AUC: Steady-state Plasma Amprenavir (APV) Pharmacokinetics ( PK) Following Admin of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent (Maraviroc) MVC 300mg BID.

Amprenavir (APV) is the active ingredient/metabolite of Fosamprenavir (FPV). (NCT00764465)
Timeframe: Day 14 of the FPV 1400mg BID, FPV 1400mg/MVC 300mg BID, FPV 700mg/RTV 100mg BID, FPV 700mg/RTV 100mg/MVC 300mg BID, FPV 1400mg/RTV 100mg QD, and FPV 1400mg/RTV 100mg QD plus MVC 300mg BID regimens

Interventionng•h/mL (Mean)
Group A & B0.56
Group C & D0.74
Group E & F0.79

[back to top]

Number of Participants Who Experienced an Adverse Event

"Safety/tolerability data collected included all adverse events (AEs) reported within the time frame of each regimen evaluated. The intent was to compare adverse events for each sequence and not for each regimen. The regimens for which AE information was culled were:~MVC 300mg BID alone~FPV 1400mg BID alone~FPV 700mg/RTV 100 mg BID alone~FPV 1400mg/RTV 100mg QD alone~FPV 1400mg BID combined with MVC 300mg BID~FPV 700mg/RTV 100 mg BID combined with MVC 300mg BID~FPV 1400mg/RTV 100mg QD combined with MVC 300mg BID The severity of reported AEs was graded according to DAIDS criteria, Version 1.0 (National Institute of Allergy and Infectious Diseases (NIAID). Table for Grading the Severity of Adult and Pediatric Adverse Events, Version 1.0. Division of Acquired Immunodeficiency Syndrome (DAIDS), Washington D.C.; 2004" (NCT00764465)
Timeframe: Day 0 through Day 49

Interventionparticipants (Number)
Group A4
Group B1
Group C3
Group D6
Group E4
Group F7

[back to top]

Cmin/Cmax: Steady-state Plasma Amprenavir (APV) Pharmacokinetics ( PK) Following Admin of Fosamprenavir (FPV) 1400mg BID, FPV 700mg/Ritonavir (RTV) 100 mg BID, or FPV 1400mg/RTV 100mg QD With and Without Concurrent (Maraviroc) MVC 300mg BID.

Amprenavir (APV) is the active ingredient/metabolite of Fosamprenavir (FPV). (NCT00764465)
Timeframe: Day 14 of the FPV 1400mg BID, FPV 1400mg/MVC 300mg BID, FPV 700mg/RTV 100mg BID, FPV 700mg/RTV 100mg/MVC 300mg BID, FPV 1400mg/RTV 100mg QD, and FPV 1400mg/RTV 100mg QD plus MVC 300mg BID regimens

,,
Interventionng/mL (Mean)
CminCmax
Group A & B0.990.49
Group C & D0.760.69
Group E & F0.680.68

[back to top]

Cmin/Cmax: Steady-state Plasma MVC PK Following Administration of RTV

Amprenavir (APV) is the active ingredient/ metabolite of Fosamprenavir (FPV). MVC minimum concentration (Cmin), maximum concentration (Cmax), and area under the plasma concentration-time curve (AUC), as determined from MVC concentrations observed in blood samples obtained at baseline, and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 12 hours during the period when MVC 300mg BID was administered with the FPV-Containing BID regimens (FPV 1400mg BID, FPV 700mg/RTV 100 mg BID), and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, and 24 hours during the period when MVC 300mg BID was administered with the FPV QD regimen (FPV 1400mg/RTV 100mg QD). As Groups A and B received the same regimens (albeit in different order), PK data for these two groups were collated, then assessed. For the same reason, PK data from Groups C and D regimens were collated before assessment, as were the PK data from Groups E and F. (NCT00764465)
Timeframe: Day 7 of the MVC 300mg BID regimen and Day 14 of the MVC 300mg/FPV 1400mg BID, MVC 300mg/FPV 700mg/RTV 100mg BID, and MVC 300mg BID Plus FPV 1400mg/RTV 100mg QD regimens

,,
Interventionng/mL (Mean)
Cmin (ng/mL)Cmax (ng/mL)
Group A & B0.720.89
Group C & D0.460.30
Group E & F0.770.93

[back to top]

Percentage of Participants With HIV-1 RNA Levels < 48 Copies/mL at Weeks 24 and 48 Using MD=F Approach

Participants who have been discontinued from the study, have been lost to follow-up, or have missing HIV-1 RNA data prior to the time point of interest were considered to have HIV-1 RNA levels > lower limit of quantification (LLOQ) . This referred as [non-completer = failure; NC=F] or [missing, discontinuation = failure; MD=F]. (NCT00791700)
Timeframe: Week 24 and Week 48 post-treatment

,,,
InterventionPercentage of participants (Number)
Week 24Week 48
>=12 - <18 Years of Age, MVC Tablet Formulation48.839.5
>=2 - <6 Years of Age, MVC Liquid Formulation18.850.0
>=6 - <12 Years of Age, MVC Liquid Formulation61.553.8
>=6 - <12 Years of Age, MVC Tablet Formulation64.554.8

[back to top]

Percentage of Participants With HIV-1 RNA Levels <400 Copies/mL at Weeks 24 and 48 Using Missing, Discontinuation = Failure (MD=F)Approach

Participants who have been discontinued from the study, have been lost to follow-up, or have missing HIV-1 RNA data prior to the time point of interest were considered to have HIV-1 RNA levels > lower limit of quantification (LLOQ) . This referred to as [non-completer = failure; NC=F] or [missing, discontinuation = failure; MD=F]. (NCT00791700)
Timeframe: Week 24 and Week 48 post-treatment

,,,
InterventionPercentage of participants (Number)
Week 24Week 48
>=12 - <18 Years of Age, MVC Tablet Formulation62.851.2
>=2 - <6 Years of Age, MVC Liquid Formulation62.575.0
>=6 - <12 Years of Age, MVC Liquid Formulation69.269.2
>=6 - <12 Years of Age, MVC Tablet Formulation87.1074.2

[back to top]

Percentage of Participants With Optimized Background Treatment Susceptibility Scores

Data was summarized by the total ARV activity of the background regimen using simple and weighted total optimized background treatment susceptibility scores as well as by screening genotype. Simple total optimized background treatment (OBT) susceptibility scores were categorized as 0, 1, >=2 and weighted total OBT susceptibility scores were categorized as 0 to 0.5, 1 to 1.5 and >=2. However, net susceptibility scores were imputed for simple analysis based on genotype. Susceptibility scores indicate the level resistance to the study medication. Scores ranged from 0 to 1 as 1 = susceptible and potential low-level resistance; 0.5 = low and intermediate-level resistance; 0 = high-level resistance, where higher scores indicated lower resistance. (NCT00791700)
Timeframe: 48 weeks

,,
InterventionPercentage of participants (Number)
Simple score 0Simple score 1.0Simple score >=2.0Weighted score 0-0.5Weighted score 1.0-1.5Weighted score >=2.0
Other Failure/Remainder0096.829.029.038.7
PDVF04.395.730.465.24.3
Response08.281.66.153.128.6

[back to top]

Pharmacokinetic(PK): Maraviroc PK Parameter, Average Plasma Concentration (Cavg), Trough Concentration(Cmin), Maximum Plasma Concentration (Cmax)

Cavg: calculated as area under the curve divided by a dosing interval of 12 hours. Cmin: directly observed plasma concentration prior to the next dose. Geometric Coefficient of Variation is defined as the geometric standard deviation to the power of the reciprocal of the geometric mean. (NCT00791700)
Timeframe: Week 2 and Week 48 (0, 1, 2, 4, 6, 8, 12 hours post-dose)

,,,
Interventionnanogram per milliliter (ng/mL) (Geometric Mean)
Cavg-Week2Cavg-Week 48Cmax-Week2Cmax-Week 48Cmin-Week2Cmin-Week 48
>=12 - <18 Years of Age, MVC Tablet Formulation239.85199.12530.80423.3256.1766.51
>=2 - <6 Years of Age, MVC Liquid Formulation237.34163.73581.47334.6818.9748.11
>=6 - <12 Years of Age, MVC Liquid Formulation264.45168.62444.37284.96115.8460.03
>=6 - <12 Years of Age, MVC Tablet Formulation260.65289.69546.80593.68100.0282.21

[back to top]

Time to Reach Maximum Plasma Concentration (Tmax)

(NCT00791700)
Timeframe: Week 2 and Week 48 (0, 1, 2, 4, 6, 8, 12 hours post-dose)

,,,
Interventionhour (Median)
Tmax - Week 2Tmax - Week 48
>=12 - <18 Years of Age, MVC Tablet Formulation2.0002.000
>=2 - <6 Years of Age, MVC Liquid Formulation2.0002.000
>=6 - <12 Years of Age, MVC Liquid Formulation2.0003.000
>=6 - <12 Years of Age, MVC Tablet Formulation4.0002.000

[back to top]

Percentage of Participants With HIV-1 RNA <400 Copies/mL and <48 Copies/mL Using the Time to Loss of Virologic Response Algorithm (TLOVR) at Week 48

TLOVR is defined as the time from first dose of study medication (Day 1) until the time of virologic failure using the a TLOVR algorithm. (NCT00791700)
Timeframe: Week 48

,,,
InterventionPercentage of participants (Number)
<400 copies/mL; TLOVR Responder<48 copies/mL; TLOVR Responder
>=12 - <18 Years of Age, MVC Tablet Formulation48.844.2
>=2 - <6 Years of Age, MVC Liquid Formulation62.543.8
>=6 - <12 Years of Age, MVC Liquid Formulation69.246.2
>=6 - <12 Years of Age, MVC Tablet Formulation74.254.8

[back to top]

Percentage of Participants With HIV-1 RNA <400 Copies/mL Through Week 48 Using Missing, Discontinuation = Failure (MD=F) Approach

"The proportion of participants who achieved HIV-1 RNA <400 copies/mL at week 24 or 48 was assessed according to Food and Drug Administration's (FDA's) Missing, Switch, Discontinuation'=Failure (MSDF) Snapshot algorithm. The algorithm uses the plasma HIV-1 RNA in the Week 24 or 48 visit window, follows the virology-first principle and considers a participant who has a missing plasma HIV-1 RNA, or switches to prohibited ARV regimen or discontinues from the study or study drug for any reason, or dies, as a failure." (NCT00791700)
Timeframe: Week 24 and Week 48 post-treatment

,,,
Interventionpercentage of participants (Number)
Week 24Week 48
>=12 - <18 Years of Age, MVC Tablet Formulation62.851.2
>=2 - <6 Years of Age, MVC Liquid Formulation68.875.0
>=6 - <12 Years of Age, MVC Liquid Formulation69.269.2
>=6 - <12 Years of Age, MVC Tablet Formulation90.377.4

[back to top] [back to top]

Area Under the Curve at Steady State (AUCtau)

AUCtau is the area under the plasma concentration time curve (AUC) at steady state from time zero (pre-dose) to end of dosing interval (tau), here dosing interval is 12 hours. (NCT00791700)
Timeframe: Week 2 and Week 48 (0, 1, 2, 4, 6, 8, 12 hours post-dose)

,,,
Interventionng*hr/mL (Geometric Mean)
AUCtau - Week 2AUCtau - Week 48
>=12 - <18 Years of Age, MVC Tablet Formulation2878.22389.4
>=2 - <6 Years of Age, MVC Liquid Formulation2848.11964.7
>=6 - <12 Years of Age, MVC Liquid Formulation3173.42023.5
>=6 - <12 Years of Age, MVC Tablet Formulation3127.73476.3

[back to top]

Change From Baseline in Cluster of Differentiation 4 (CD4+) Cell Count at Weeks 24 and 48

Change from baseline in CD4 cell count to Week 24 and Week 48 were tabulated in aggregated and broken down by age cohort using summary statistics. (NCT00791700)
Timeframe: Baseline, Week 24, Week 48 post-treatment

,,,
Interventioncells/mm^3 (Mean)
Week 24Week 48
>=12 - <18 Years of Age, MVC Tablet Formulation173.6168.6
>=2 - <6 Years of Age, MVC Liquid Formulation232.7275.9
>=6 - <12 Years of Age, MVC Liquid Formulation213.9167.3
>=6 - <12 Years of Age, MVC Tablet Formulation355.8362.7

[back to top]

Change From Baseline in HIV-1 RNA (Log10 Copies/mL)

Plasma HIV-1 RNA was determined using the Roche COBAS AmpliPrep/COBAS TaqMan HIV-1 Test (lower limit of quantification [LLOQ] <48 copies/mL). Blood samples were taken at the time points indicated in the participant evaluation schedule. Screening HIV-1 RNA >1000 copies/ml was used to determine eligibility for the study. (NCT00791700)
Timeframe: Baseline, Week 24, Week 48 post-treatment

,,,
InterventionLog10 Copies/mL (Mean)
Change from Baseline - Log10 - Week 24Change from Baseline - Log10 - Week 48
>=12 - <18 Years of Age, MVC Tablet Formulation-1.6482-1.4591
>=2 - <6 Years of Age, MVC Liquid Formulation-2.4853-2.5831
>=6 - <12 Years of Age, MVC Liquid Formulation-2.1756-2.0549
>=6 - <12 Years of Age, MVC Tablet Formulation-2.2324-1.9579

[back to top]

Change From Baseline in HIV-1 RNA (Original)

Plasma HIV-1 RNA was determined using the Roche COBAS AmpliPrep/COBAS TaqMan HIV-1 Test (lower limit of quantification [LLOQ] <48 copies/mL). Blood samples were taken at the time points indicated in the participant evaluation schedule. Screening HIV-1 RNA >1000 copies/ml was used to determine eligibility for the study. (NCT00791700)
Timeframe: Baseline, Week 24, Week 48 post-treatment

,,,
Interventioncopies/mL (Mean)
Change from Baseline - Original - Week 24Change from Baseline - Original - Week 48
>=12 - <18 Years of Age, MVC Tablet Formulation-57325.7-55321.1
>=2 - <6 Years of Age, MVC Liquid Formulation-271974.6-267834.2
>=6 - <12 Years of Age, MVC Liquid Formulation-58081.0-56351.7
>=6 - <12 Years of Age, MVC Tablet Formulation-38764.0-34787.7

[back to top]

Change From Baseline in Percentage (%) of CD4+ Cells at Weeks 24 and 48

Change from baseline in CD4 % to Week 24 and Week 48 were tabulated in aggregated and broken down by age cohort using summary statistics. (NCT00791700)
Timeframe: Baseline, Week 24 and Week 48 post-treatment

,,,
Interventionpercentage of CD4+ cells (Mean)
Week 24Week 48
>=12 - <18 Years of Age, MVC Tablet Formulation3.84.6
>=2 - <6 Years of Age, MVC Liquid Formulation7.37.5
>=6 - <12 Years of Age, MVC Liquid Formulation3.52.5
>=6 - <12 Years of Age, MVC Tablet Formulation3.86.0

[back to top]

Incidence and Severity of Grade 3 and Grade 4 Treatment-Emergent Adverse Events(AEs) (All Causality)

Incidence is reported in terms of number of events of AEs. The investigator used the Division of AIDS (DAIDS) Table for Grading the Severity of Adult and Pediatric AEs as follows: Grade 1= Symptoms causing no or minimal interference with usual social and functional activities; Grade 2= Symptoms causing greater than minimal interference with usual social and functional activities; Grade 3= Symptoms causing inability to perform usual social and functional activities; Grade 4= Symptoms causing inability to perform basic self-care functions or medical or operative intervention indicated to prevent permanent impairment, persistent disability, or death. Data have been reported in the measure for Grade 3 and 4 as per system organ class and preferred term. (NCT00791700)
Timeframe: Baseline up to 5 years

,,,,,,,
Interventionevents (Number)
Gastrointestinal disorders - VomitingHepat. disorders - Drug-induced liver injuryInfections and infestations - H1N1 influenzaInfections and infestations - PneumoniaInvestigations - Lipase increasedPyschiatric disorder - Bipolar disorderGastrointestinal disorders - GastritisInvestigations - Hepatic enzyme abnormalInvestigations - Transaminases increasedPyschiatric disorder - AggressionBlood and lymphatic system disorders - AnaemiaInfections and infestations - MeningitisInfections and infestations -Otitis media
Cohort 1 (Grade 3)1000000000000
Cohort 1 (Grade 4)0000100000000
Cohort 2 (Grade 3)1000011111001
Cohort 2 (Grade 4)0000000000000
Cohort 3 (Grade 3)0001000000000
Cohort 3 (Grade 4)0000000000000
Cohort 4 (Grade 3)0011000000100
Cohort 4 (Grade 4)0100000000010

[back to top]

Number of Participants With Emergence of Reverse Transcriptase Inhibitor (RTI) and Protease Inhibitor (PI) Resistance Associated Mutations (RAMs) Between Screening and On-Treatment Confirmed PDVF

Phenotypic and genotypic susceptibility to reverse transcriptase and protease inhibitors was evaluated at screening using the Monogram Biosciences PhenoSense™ GT (PSGT) assay. Samples from a confirmatory PDVF visit or early termination of MVC were planned to be analyzed if the plasma HIV-1 RNA was ≥400 copies/mL. Data for participants with respective gene mutation category has been reported. Participants with more than one mutation are counted more than once. (NCT00791700)
Timeframe: 48 weeks

,,,
Interventionparticipants (Number)
With valid on-treatment resultsPI Minor L10L/FPI Minor L89L/I/MPI Minor V77V/INNRTI K103K/NNNRTI K103NNRTI M184VPI Minor K20K/RTotal with emergence
>=12 - <18 Years of Age, MVC Tablet Formulation1111101004
>=2 - <6 Years of Age, MVC Liquid Formulation200000000
>=6 - <12 Years of Age, MVC Liquid Formulation300001123
>=6 - <12 Years of Age, MVC Tablet Formulation400010001

[back to top]

Number of Participants With Protocol Defined Virologic Failure

The occurrence of any one of the following criteria would constitute Virologic failure: Criteria A=Decrease from Baseline plasma HIV-1 RNA <1 log10 and plasma HIV-1 RNA >400 copies/mL starting at Week 12 and confirmed at consecutive Week 16; Criteria B=Decrease from Baseline plasma HIV-1 RNA <2.0 log10 and plasma HIV-1 RNA >400 copies/mL at Week 24 OR plasma HIV-1 RNA >10,000 copies/mL on and after Week 24, and confirmed within 14 to 21 days; Criteria C=Increase from nadir plasma HIV-1 RNA of >=1 log10 (>=1,000 copies/mL if nadir plasma HIV-1 RNA <48 copies/mL) at any time, and confirmed within 14 to 21 days. (NCT00791700)
Timeframe: Week 48

,,,
Interventionparticipants (Number)
Criteria ACriteria BCriteria C
>=12 - <18 Years of Age, MVC Tablet Formulation508
>=2 - <6 Years of Age, MVC Liquid Formulation003
>=6 - <12 Years of Age, MVC Liquid Formulation102
>=6 - <12 Years of Age, MVC Tablet Formulation203

[back to top]

Number of Participants With Viral Tropism Between Screening and Confirmed Protocol Defined Virologic Failure (PDVF) Prior to Week 48

Virus tropism was determined using the Monogram Biosciences Trofile™ viral tropism assay. Change in detected tropism from screening to the time of failure prior to Week 48 was reported. X4=CXCR4 tropic virus; R5=CCR5-tropic virus; X4=CXCR4-tropic virus. Number of participants as per tropism to respective virus has been reported. (NCT00791700)
Timeframe: Screening to Week 48

,,,
Interventionparticipants (Number)
With valid on-treatment resultsTropism at Confirmed PDVF R5Tropism at Confirmed PDVF DMTropism at Confirmed PDVF X4Tropism at Confirmed PDVF Not Reportable
>=12 - <18 Years of Age, MVC Tablet Formulation119201
>=2 - <6 Years of Age, MVC Liquid Formulation22001
>=6 - <12 Years of Age, MVC Liquid Formulation32100
>=6 - <12 Years of Age, MVC Tablet Formulation43100

[back to top]

Percentage of Participants With >= 1.0 log10 Reduction in HIV-1RNA Concentration From Baseline to Week 24 and Week 48

Percentage of participants with at least a 1.0 log10 reduction in HIV-1 RNA from baseline to Week 24 and Week 48 were tabulated. (NCT00791700)
Timeframe: Baseline to Week 24, Week 48 post-treatment

,,,
Interventionpercentage of participants (Number)
Baseline to Week 24Baseline to Week 48
>=12 - <18 Years of Age, MVC Tablet Formulation93.188.0
>=2 - <6 Years of Age, MVC Liquid Formulation92.3100.0
>=6 - <12 Years of Age, MVC Liquid Formulation100.0100.0
>=6 - <12 Years of Age, MVC Tablet Formulation100.096.2

[back to top]

Percentage of Participants With HIV-1 RNA <48 Copies/mL Through Week 48 Using Missing, Discontinuation = Failure (MD=F)Approach

"The proportion of participants who achieved HIV-1 RNA <48 copies/mL at week 24 or 48 was assessed according to Food and Drug Administration's (FDA's) Missing, Switch, Discontinuation'=Failure (MSDF) Snapshot algorithm. The algorithm uses the plasma HIV-1 RNA in the Week 24 or 48 visit window, follows the virology-first principle and considers a participant who has a missing plasma HIV-1 RNA, or switches to prohibited ARV regimen or discontinues from the study or study drug for any reason, or dies, as a failure." (NCT00791700)
Timeframe: Week 24 and Week 48 post-treatment

,,,
Interventionpercentage of participants (Number)
Week 24Week 48
>=12 - <18 Years of Age, MVC Tablet Formulation48.839.5
>=2 - <6 Years of Age, MVC Liquid Formulation18.7550.0
>=6 - <12 Years of Age, MVC Liquid Formulation61.553.8
>=6 - <12 Years of Age, MVC Tablet Formulation64.554.8

[back to top]

HIV-1 RNA Levels at Baseline

(NCT00827112)
Timeframe: Baseline

Interventioncopies/mL (Mean)
Maraviroc+ Atazanavir / Ritonavir84982
Atazanavir / Ritonavir + Emtricitabine / Tenofovir114827

[back to top]

Maximum Observed Plasma Concentration (Cmax) of Maraviroc

(NCT00827112)
Timeframe: Day 14 (0, 0.5, 1, 2, 3, 4, 6, 8, 12 and 24 hours post dose)

Interventionnanogram (ng)/mL (Median)
Maraviroc+ Atazanavir / Ritonavir650

[back to top]

Number of Participants With Phenotypic Resistance

Phenotypic resistance was assessed for all participants at screening and was evaluated for PIs, NRTIs, and NNRTIs using Monogram GenoSeq and/or PhenoSenseGT assays. This was then repeated for all participants with HIV-1 viral load more than 500 copies/mL either at treatment failure or at early termination, up to Week 96. (NCT00827112)
Timeframe: Week 96 or Time of treatment failure

InterventionParticipants (Number)
Maraviroc+ Atazanavir / Ritonavir0
Atazanavir / Ritonavir + Emtricitabine / Tenofovir0

[back to top]

Change From Baseline in Cluster of Differentiation 8+T Lymphocyte (CD8) Cell Count at Weeks 16, 24, 48 and 96

(NCT00827112)
Timeframe: Baseline, Week 16, Week 24, Week 48, Week 96

,
Interventioncells/mcL (Mean)
Baseline (n= 59, 61)Change at Week 16 (n= 54, 58)Change at Week 24 (n= 54, 57)Change at Week 48 (n= 52, 53)Change at Week 96 (n= 50, 51)
Atazanavir / Ritonavir + Emtricitabine / Tenofovir1125.60-153.80-178.00-267.60-231.40
Maraviroc+ Atazanavir / Ritonavir931.1063.706.20-76.80-63.00

[back to top]

Percentage of Participants With Less Than 400 Copies/mL of HIV-1 RNA

(NCT00827112)
Timeframe: Week 2, Week 4, Week 8, Week 12, Week 16, Week 20, Week 24, Week 32, Week 40, Week 48, Week 60, Week 72, Week 84, Week 96

,
InterventionPercentage of participants (Number)
Week 2 (n= 55, 60)Week 4 (n= 57, 60)Week 8 (n= 57, 59)Week 12 (n= 55, 59)Week 16 (n= 54, 58)Week 20 (n= 56, 57)Week 24 (n= 56, 58)Week 32 (n= 55, 57)Week 40 (n= 54, 55)Week 48 (n= 53, 54)Week 60 (n= 52, 53)Week 72 (n= 52, 53)Week 84 (n= 50, 52)Week 96 (n= 49, 51)
Atazanavir / Ritonavir + Emtricitabine / Tenofovir34.4352.4677.0588.5291.8093.4493.4493.4490.1686.8986.8985.2585.2583.61
Maraviroc+ Atazanavir / Ritonavir27.1250.8579.6689.8388.1489.8391.5389.8391.5389.8386.4486.4481.3677.97

[back to top]

Change From Baseline in Cluster of Differentiation 4+T Lymphocyte (CD4) Cell Counts at Weeks 16, 24, 48 and 96

(NCT00827112)
Timeframe: Baseline, Week 16, Week 24, Week 48, Week 96

,
Interventioncells/microliter (cells/mcL) (Mean)
Baseline (n= 59, 61)Change at Week 16 (n= 54, 58)Change at Week 24 (n= 54, 57)Change at Week 48 (n= 52, 53)Change at Week 96 (n= 50, 51)
Atazanavir / Ritonavir + Emtricitabine / Tenofovir390.00139.80173.30226.60298.50
Maraviroc+ Atazanavir / Ritonavir357.70169.60188.90215.70287.50

[back to top]

Time to Loss of Virological Response (TLOVR)

TLOVR (virological failure) was defined as the time from first dose of study treatment (Day 1) until the time of virologic failure using the time to loss of virologic response algorithm. (NCT00827112)
Timeframe: Baseline through Week 96

InterventionDays (Mean)
Maraviroc+ Atazanavir / Ritonavir436.2
Atazanavir / Ritonavir + Emtricitabine / Tenofovir463.8

[back to top]

Percentage of Participants With Plasma Human Immuno Deficiency Virus-1 Ribonucleic Acid (HIV-1 RNA) Levels Less Than 50 Copies/Milliliter (mL)

(NCT00827112)
Timeframe: Week 48

InterventionPercentage of participants (Number)
Maraviroc+ Atazanavir / Ritonavir74.60
Atazanavir / Ritonavir + Emtricitabine / Tenofovir83.60

[back to top]

Time-Averaged Difference (TAD) in log10 Viral Load

TAD was calculated as area under the curve of HIV divided by time period minus baseline HIV where HIV was denoted as HIV-1 RNA (log10 copies/mL). (NCT00827112)
Timeframe: Week 16, Week 24, Week 48, Week 96

,
Interventionlog10 copies/mL (Mean)
Week 16Week 24Week 48Week 96
Atazanavir / Ritonavir + Emtricitabine / Tenofovir-2.402-2.626-2.868-3.001
Maraviroc+ Atazanavir / Ritonavir-2.459-2.663-2.897-2.998

[back to top]

Average Observed Plasma Concentration (Cavg) of Maraviroc

Cavg was described as area under the plasma concentration-time profile from time zero to time 24 hours (AUC24) divided by the dosing interval (AUC24/ 24). (NCT00827112)
Timeframe: Day 14 (0, 0.5, 1, 2, 3, 4, 6, 8, 12 and 24 hours post dose)

Interventionng/mL (Mean)
Maraviroc+ Atazanavir / Ritonavir185.10

[back to top]

Number of Participants With Genotypic Resistance

Genotypic resistance was assessed for all participants at screening and was evaluated for protease inhibitors (PIs), Nucleotide reverse transcriptase inhibitors (NRTIs), and non-NRTIs (NNRTIs) using Monogram GenoSeq and/or PhenoSenseGT assays. This was then repeated for all participants with HIV-1 viral load more than 500 copies/mL either at treatment failure or at early termination, up to Week 96. (NCT00827112)
Timeframe: Week 96 or Time of treatment failure

InterventionParticipants (Number)
Maraviroc+ Atazanavir / Ritonavir0
Atazanavir / Ritonavir + Emtricitabine / Tenofovir0

[back to top]

Change From Baseline in Plasma log10 Viral Load at Weeks 16, 24, 48 and 96

(NCT00827112)
Timeframe: Baseline, Week 16, Week 24, Week 48, Week 96

,
Interventionlog10 copies/ml (Mean)
Baseline (n= 59, 61)Change at Week 16 (n= 54, 58)Change at Week 24 (n= 56, 58)Change at Week 48 (n= 53, 54)Change at Week 96 (n= 49, 51)
Atazanavir / Ritonavir + Emtricitabine / Tenofovir114827-107684.6-110498.1-115582.9-99662.6
Maraviroc+ Atazanavir / Ritonavir84982-89859.1-87241.2-82343.4-80117.7

[back to top]

Number of Participants With HIV-1 RNA Tropism Status Using Trofile Assay

Viral tropism was determined using the trofile assay with enhanced sensitivity for participants with HIV-1 RNA greater than equal to 1000 copies/mL. The enhanced trofile assay had the sensitivity to detect 100 percent of spiked samples when C-X-C chemokine receptor type 4 {CXCR4} [X4]-using HIV-1 RNA represented 0.3 percent of the total viral population. (NCT00827112)
Timeframe: Baseline to Week 96 or Time of treatment Failure

,
InterventionParticipants (Number)
BaselineWeek 96 or Time of treatment Failure
Atazanavir / Ritonavir + Emtricitabine / Tenofovir610
Maraviroc+ Atazanavir / Ritonavir600

[back to top]

Percentage of Participants With Less Than 50 Copies/mL of HIV-1 RNA

(NCT00827112)
Timeframe: Week 2, Week 4, Week 8, Week 12, Week 16, Week 20, Week 24, Week 32, Week 40, Week 48, Week 60, Week 72, Week 84, Week 96

,
InterventionPercentage of participants (Number)
Week 2 (n= 55, 60)Week 4 (n= 57, 60)Week 8 (n= 57, 59)Week 12 (n= 55, 59)Week 16 (n= 54, 58)Week 20 (n= 56, 57)Week 24 (n= 56, 58)Week 32 (n= 55, 57)Week 40 (n= 54, 55)Week 48 (n= 53, 54)Week 60 (n= 52, 53)Week 72 (n= 52, 53)Week 84 (n= 50, 52)Week 96 (n= 49, 51)
Atazanavir / Ritonavir + Emtricitabine / Tenofovir6.6021.3042.6062.3073.8083.6188.5288.5288.5283.6185.2581.9783.6181.97
Maraviroc+ Atazanavir / Ritonavir08.5047.5061.0072.9071.2081.3679.6681.3674.5867.8074.5876.2767.80

[back to top]

Minimum Observed Plasma Concentration (Cmin) of Maraviroc

(NCT00827112)
Timeframe: Day 14 (0, 0.5, 1, 2, 3, 4, 6, 8, 12 and 24 hours post dose)

Interventionng/mL (Median)
Maraviroc+ Atazanavir / Ritonavir37.0

[back to top]

Change From Baseline in HIV-1 RNA Levels of First 15 Participants at Days 4, 7, 10 and 14

Plasma HIV-1 RNA levels were evaluated for first 15 participants enrolled at United States (U.S) sites only. (NCT00827112)
Timeframe: Baseline , Days 4, 7, 10 and 14

,
Interventioncopies/mL (Mean)
Change at Day 4Change at Day 7Change at Day 10Change at Day 14
Atazanavir / Ritonavir + Emtricitabine / Tenofovir-46479.40-52137.10-54925.90-55449.90
Maraviroc+ Atazanavir / Ritonavir1800.00-36947.90-58595.80-47271.60

[back to top]

Percent Change in FMD

endothelial function as assessed by measured flow-mediated vasodilation (FMD) of the brachial artery (NCT00844519)
Timeframe: Baseline, 24 weeks

Interventionpercent change in FMD (Mean)
Maraviroc0.57
Placebo-0.05

[back to top]

Change From Baseline in Cluster of Differentiation 4 (CD4+) Cell Counts at Month 6

(NCT00850395)
Timeframe: Baseline, Month 6

Interventioncells/mcL (Mean)
Maraviroc91.88

[back to top]

Change From Baseline in Log 10 Transformed Human Immunodeficiency Virus-1 Ribonucleic Acid (HIV-1 RNA) at Month 3

(NCT00850395)
Timeframe: Baseline, Month 3

Interventioncopies/mL (Mean)
Maraviroc-1.10

[back to top]

Change From Baseline in Log 10 Transformed Human Immunodeficiency Virus-1 Ribonucleic Acid (HIV-1 RNA) at Month 6

(NCT00850395)
Timeframe: Baseline, Month 6

Interventioncopies/mL (Mean)
Maraviroc-1.15

[back to top]

Number of Participants With Human Immunodeficiency Virus (HIV) Response

Response was defined as a HIV-1 RNA count of less than 50 copies/mL. (NCT00850395)
Timeframe: Month 12

Interventionparticipants (Number)
Maraviroc51

[back to top]

Change From Baseline in Acquired Immune Deficiency Syndrome (AIDS) Clinical Trials Group (ACTG) Symptom Distress Module (SDM) Overall Score at Months 6 and 12

SDM consists of the 20 items questionnaire, each item rated from 0 to 4 where 0 (complete absence of symptom) and 4 (very bothersome symptom). Overall score calculated as the sum of the scores for each of the 20 items of the questionnaire and ranged from 0 (best health) and 80 (worst health). A positive change from baseline indicates a decline in a participant's quality of life over that period. (NCT00850395)
Timeframe: Baseline, Months 6, 12

Interventionunits on a scale (Mean)
Change at Month 6 (n=51)Change at Month 12 (n=79)
Maraviroc-4.87-2.14

[back to top]

Number of Participants Taking Concomitant Therapy

Participants taking HIV/AIDS concomitant medication at Month 12, at Baseline and Month 12 were reported. It included Emtricitabine/tenofovir disoproxil fumarate(FTC/TDF),Raltegravir(RAL), Ritonavir (RTV), Darunavir(DRV), Kaletra, Atazanavir sulfate(ATV), Abacavir sulfate/lamivudine(ABC/LAM), Tenofovir disoproxil fumarate(TDF), Etravirine(ETR), Lamivudine (LAM), Zidovudine W/lamivudine(ZDV W/LAM), Nevirapine(NVP), Saquinavir mesilate(SQV), Trizivir(TZV), Zidovudine(ZDV), Abacavir sulfate(ABC), Emtricitabine(FTC),Entecavir(ETV). (NCT00850395)
Timeframe: Baseline, Month 12

Interventionparticipants (Number)
ABC, FTC/TDF (Month 12)ABC, FTC/TDF (Baseline, Month 12)ABC/LAM (Month 12)ABC/LAM (Baseline, Month 12)ABC/LAM, DRV, RAL (Month 12)ABC/LAM, DRV, RAL (Baseline, Month 12)ABC/LAM, RAL (Month 12)ABC/LAM, RAL (Baseline, Month 12)ABC/LAM, RAL, TDF (Month 12)ABC/LAM, RAL, TDF (Baseline, Month 12)ATV (Month 12)ATV (Baseline, Month 12)ATV, FTC/TDF (Month 12)ATV, FTC/TDF (Baseline, Month 12)ATV, FTC/TDF, RTV (Month 12)ATV, FTC/TDF, RTV (Baseline, Month 12)ATV, LAM, RAL, RTV (Month 12)ATV, LAM, RAL, RTV (Baseline, Month 12)ATV, RAL (Month 12)ATV, RAL (Baseline, Month 12)ATV, RAL, RTV (Month 12)ATV, RAL, RTV (Baseline, Month 12)ATV, RTV (Month 12)ATV, RTV (Baseline, Month 12)DRV, FTC/TDF (Month 12)DRV, FTC/TDF (Baseline, Month 12)DRV, FTC/TDF, ETR (Month 12)DRV, FTC/TDF, ETR (Baseline, Month 12)DRV, FTC/TDF, RTV (Month 12)DRV, FTC/TDF, RTV (Baseline, Month 12)DRV, ETV, ETR, RTV (Month 12)DRV, ETV, ETR, RTV (Baseline, Month 12)DRV, LAM, RAL (Month 12)DRV, LAM, RAL (Baseline, Month 12)DRV, LAM, RAL, RTV (Month 12)DRV, LAM, RAL, RTV (Baseline, Month 12)DRV, LAM, TDF (Month 12)DRV, LAM, TDF (Baseline, Month 12)DRV, NVP, RAL, RTV (Month 12)DRV, NVP, RAL, RTV (Baseline, Month 12)DRV, RAL, RTV (Month 12)DRV, RAL, RTV (Baseline, Month 12)DRV, RTV (Month 12)DRV, RTV (Baseline, Month 12)DRV, RTV, TZV (Month 12)DRV, RTV, TZV (Baseline, Month 12)FTC/TDF (Month 12)FTC/TDF (Baseline, Month 12)FTC/TDF, Kaletra (Month 12)FTC/TDF, Kaletra (Baseline, Month 12)FTC/TDF, Kaletra, RAL (Month 12)FTC/TDF, Kaletra, RAL (Baseline, Month 12)FTC/TDF, Kaletra, SQV (Month 12)FTC/TDF, Kaletra, SQV (Baseline, Month 12)FTC/TDF, RAL (Month 12)FTC/TDF, RAL (Baseline, Month 12)FTC/TDF, RAL, SQV (Month 12)FTC/TDF, RAL, SQV (Baseline, Month 12)ETR, RAL, RTV (Month 12)ETR, RAL, RTV (Baseline, Month 12)Kaletra (Month 12)Kaletra (Baseline, Month 12)LAM, RAL, TDF, ZDV (Month 12)LAM, RAL, TDF, ZDV (Baseline, Month 12)RAL (Month 12)RAL (Baseline, Month 12)RAL, TDF (Month 12)RAL, TDF (Baseline, Month 12)RAL, ZDV W/LAM (Month 12)RAL, ZDV W/LAM (Baseline, Month 12)TDF, TZV (Month 12)TDF, TZV (Baseline, Month 12)TDF, ZDV (Month 12)TDF, ZDV (Baseline, Month 12)
Maraviroc1122102211111111102211111111651010101111442211131121331154111022102211111111

[back to top]

Number of Participants With Centers for Disease Control and Prevention (CDC) Classification at Month 12

Participants were classified based on the severity as mild (Category A), moderate (Category B), and severe (Category C). (NCT00850395)
Timeframe: Month 12

Interventionparticipants (Number)
Category ACategory BCategory C
Maraviroc312028

[back to top]

Number of Participants With Centers for Disease Control and Prevention (CDC) Classification at Month 3

Participants were classified based on the severity as mild (Category A), moderate (Category B), and severe (Category C). (NCT00850395)
Timeframe: Month 3

Interventionparticipants (Number)
Category ACategory BCategory C
Maraviroc311926

[back to top]

Number of Participants With Centers for Disease Control and Prevention (CDC) Classification at Month 6

Participants were classified based on the severity as mild (Category A), moderate (Category B), and severe (Category C). (NCT00850395)
Timeframe: Month 6

Interventionparticipants (Number)
Category ACategory BCategory C
Maraviroc311627

[back to top]

Physician's Assessment of Efficacy

Number of participants with each grade of efficacy as assessed by the physician was reported on the 5 point categorical scale: excellent, very good, good, fair, poor. (NCT00850395)
Timeframe: Month 12

Interventionparticipants (Number)
ExcellentVery GoodGoodFairPoor
Maraviroc15421411

[back to top]

Change From Baseline in Log 10 Transformed Human Immunodeficiency Virus-1 Ribonucleic Acid (HIV-1 RNA) at Month 12

(NCT00850395)
Timeframe: Baseline, Month 12

Interventioncopies/mL (Mean)
Maraviroc-0.95

[back to top]

Change From Baseline in Cluster of Differentiation 4 (CD4+) Cell Counts at Month 12

(NCT00850395)
Timeframe: Baseline, Month 12

Interventioncells/mcL (Mean)
Maraviroc97.57

[back to top]

Change From Baseline in Cluster of Differentiation 4 (CD4+) Cell Counts at Month 3

(NCT00850395)
Timeframe: Baseline, Month 3

Interventioncells/mcL (Mean)
Maraviroc74.31

[back to top]

Postural Changes in Pulse Rate

Postural change calculated as position 1 (standing) value minus position 2 (supine) value. Baseline was the average of the 3 predose measurements at each period. Means of Replicates were used in calculations. (NCT00853840)
Timeframe: Baseline, 6 and 12 hours post dose on Day 1 from the First Day of each Treatment Leg

,
Interventionbeats per minute (bpm) (Mean)
Baseline6 hours12 hours
Maraviroc + Placebo (Treatment B)14.320.412.0
Maraviroc + Vardenafil (Treatment A)16.022.912.0

[back to top]

Number of Subjects With Postural Hypotension

Postural (orthostatic) hypotension defined as 1) decrease in standing-supine diastolic blood pressure (BP) greater than or equal to 10 mm Hg; 2) decrease in standing-supine systolic BP greater than or equal to 20 mm Hg; or 3) standing systolic BP less than 90 mm Hg. Assessed at Period 1 and Period 2 BP measurement timepoints post maraviroc dose. (NCT00853840)
Timeframe: Period 1 and Period 2 (up to 8 days)

Interventionparticipants (Number)
Maraviroc + Vardenafil (Treatment A)0
Maraviroc + Placebo (Treatment B)0

[back to top]

Standing and Supine Systolic and Diastolic Blood Pressure (BP)

Supine BP was taken after subjects rested for 5 minutes supine. Subjects then sat for 2 minutes and stood for 2 minutes then standing BP taken. Duplicate supine and standing BP measurements were taken per the protocol. The average of the duplicate measurements was calculated prior to data analysis. (NCT00853840)
Timeframe: 1.5, 2, 2.5, 3, 4, 6, 8, 12 hours post Vardenafil or Placebo dose

,
Interventionmm Hg (Least Squares Mean)
Supine Systolic BP: 1.5 hour post Maraviroc doseSupine Diastolic BP: 1.5 hour post Maraviroc doseSupine Systolic BP: 2 hour post Maraviroc doseSupine Diastolic BP: 2 hour post Maraviroc doseSupine Systolic BP: 2.5 hour post Maraviroc doseSupine Diastolic BP: 2.5 hour post Maraviroc doseSupine Systolic BP: 3 hour post Maraviroc doseSupine Diastolic BP: 3 hour post Maraviroc doseSupine Systolic BP: 4 hour post Maraviroc doseSupine Diastolic BP: 4 hour post Maraviroc doseSupine Systolic BP: 6 hour post Maraviroc doseSupine Diastolic BP: 6 hour post Maraviroc doseSupine Systolic BP: 8 hour post Maraviroc doseSupine Diastolic BP: 8 hour post Maraviroc doseSupine Systolic BP: 12 hour post Maraviroc doseSupine Diastolic BP: 12 hour post Maraviroc doseStanding Systolic BP: 1.5 hour post Maraviroc doseStanding Diastolic BP 1.5 hour post Maraviroc doseStanding Systolic BP: 2 hour post Maraviroc doseStanding Diastolic BP: 2 hour post Maraviroc doseStanding Systolic BP: 2.5 hour post Maraviroc doseStanding Diastolic BP 2.5 hour post Maraviroc doseStanding Systolic BP: 3 hour post Maraviroc doseStanding Diastolic BP: 3 hour post Maraviroc doseStanding Systolic BP: 4 hour post Maraviroc doseStanding Diastolic BP: 4 hour post Maraviroc doseStanding Systolic BP: 6 hour post Maraviroc doseStanding Diastolic BP: 6 hour post Maraviroc doseStanding Systolic BP: 8 hour post Maraviroc doseStanding Diastolic BP: 8 hour post Maraviroc doseStanding Systolic BP: 12 hour post Maraviroc doseStanding Diastolic BP: 12 hour post Maraviroc dose
Maraviroc + Placebo (Treatment B)119.4769.99118.4469.27119.5570.80116.1669.05117.9769.24118.7566.52120.8368.85124.7570.27125.1878.74125.4080.69121.3278.94121.3777.38121.4078.38120.5976.77123.9077.32128.6580.91
Maraviroc + Vardenafil (Treatment A)118.3668.06113.4566.31114.6766.98114.1167.28115.8968.37116.4565.67116.6166.34122.8167.95120.8274.81118.4974.90115.5272.29117.1073.73119.3577.29118.2973.34120.4374.06128.3878.65

[back to top]

Standing and Supine Pulse Rate

Supine pulse rate measurement was taken after subject rested for 5 minutes supine. Subject sat for 2 minutes, then stood for 2 minutes and standing measurement taken. Duplicate supine and standing pulse rate measurements taken per protocol. Average of duplicate measurements calculated prior to data analysis. (NCT00853840)
Timeframe: 1.5, 2, 2.5, 3, 4, 6, 8, 12 hours post Vardenafil or Placebo dose

,
Interventionbeats per minute (bpm) (Least Squares Mean)
Supine pulse rate: 1.5 hours post Maraviroc doseSupine pulse rate: 2 hours post Maraviroc doseSupine pulse rate: 2.5 hours post Maraviroc doseSupine pulse rate: 3 hours post Maraviroc doseSupine pulse rate: 4 hours post Maraviroc doseSupine pulse rate: 6 hours post Maraviroc doseSupine pulse rate: 8 hours post Maraviroc doseSupine pulse rate: 12 hours post Maraviroc doseStanding pulse rate: 1.5 hours post Maraviroc doseStanding pulse rate: 2 hours post Maraviroc doseStanding pulse rate: 2.5 hours post Maraviroc doseStanding pulse rate: 3 hours post Maraviroc doseStanding pulse rate: 4 hours post Maraviroc doseStanding pulse rate: 6 hours post Maraviroc doseStanding pulse rate: 8 hours post Maraviroc doseStanding pulse rate: 12 hours post Maraviroc dose
Maraviroc + Placebo (Treatment B)60.6160.6958.0057.5358.1665.9463.5567.8078.4578.3477.2275.3475.9287.1779.3480.59
Maraviroc + Vardenafil (Treatment A)67.5666.2061.6460.4260.2271.3967.6771.2585.7885.1682.2580.0579.9793.5384.4182.47

[back to top]

Postural Changes in Systolic and Diastolic Blood Pressure

Postural change calculated as position 1 (standing) value minus the position 2 (supine) value. Baseline was the average of 3 predose measurements at each period. Means of replicates were used in calculations. (NCT00853840)
Timeframe: Baseline, 6 and 12 hours post dose on Day 1 from the First Day of each Treatment Leg

,
Interventionmm Hg (Mean)
Systolic Blood Pressure: BaselineDiastolic Blood Pressure: BaselineSystolic Blood Pressure: 6 hoursDiastolic Blood Pressure: 6 hoursSystolic Blood Pressure: 12 hoursDiastolic Blood Pressure: 12 hours
Maraviroc + Placebo (Treatment B)2.67.51.59.63.59.9
Maraviroc + Vardenafil (Treatment A)3.58.92.28.45.911.4

[back to top]

Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Mean Daily Dose of Celsentri

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. In this outcome measure percentage of participants with ADRs categorized by mean daily dose of Celsentri are reported to assess mean daily dose of Celsentri as a risk factor for ADR. One tablet of Celsentri had a dose of 150 mg. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
<2 tablets2 tablets>2 tablets
Celsentri (Maraviroc) Tablets016.6735.71

[back to top]

Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Inpatient or Outpatient Status

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. In this outcome measure percentage of participants with ADRs categorized by inpatient or outpatient status are reported to assess inpatient or outpatient status as a risk factor for ADR. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
InpatientOutpatientInpatient and outpatient
Celsentri (Maraviroc) Tablets0.0015.5642.86

[back to top]

Percentage of Participants With Adverse Drug Reactions (ADRs): Factor HIV Infection Duration

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. In this outcome measure percentage of participants with ADRs categorized by HIV infection duration are reported to assess HIV infection duration as a risk factor for ADR. Unknown: participants for which the duration of HIV infection was not known. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
<=1 yeargreater than (>) 4 and <=5 years>5 yearsUnknown
Celsentri (Maraviroc) Tablets0.000.0050.0022.58

[back to top]

Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Gender

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. In this outcome measure percentage of participants with ADRs categorized by gender are reported to assess gender as a risk factor for ADR. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
MalesFemales
Celsentri (Maraviroc) Tablets23.0833.33

[back to top]

Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Ethnicity

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. In this outcome measure percentage of participants with ADRs categorized by ethnicity are reported to assess ethnicity as a risk factor for ADR. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
JapaneseOthers
Celsentri (Maraviroc) Tablets23.880.00

[back to top]

Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Centers for Disease Control and Prevention (CDC) Classification

An ADR:any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by physician.Participants are divided into 3 categories as per CDC classification based on level of HIV infection: Category A= asymptomatic HIV-1 infection, persistent generalized lymphadenopathy and acute(primary)HIV-1 infection with accompanying illness or history of acute HIV-1 infection in adult or adolescent aged>=13 years, Category B: conditions attributed to HIV infection or are indicative of a defect in cell-mediated immunity; or the conditions are considered by physicians to have a clinical course or to require management that is complicated by HIV infection in an HIV-infected adolescent or adult; and Category C: clinical conditions listed in the acquired immunodeficiency syndrome (AIDS) diagnostic criteria, corresponding to conventional AIDS. Unknown: Participants for which CDC classification was not described. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
CDC category ACDC category BCDC category CUnknown
Celsentri (Maraviroc) Tablets20.6920.0029.030.00

[back to top]

Percentage of Participants With Adverse Drug Reactions (ADRs): Factor as Per Presence or Absence of Concomitant Therapies

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. In this outcome measure percentage of participants with ADRs categorized by presence or absence of concomitant therapies are reported to assess presence or absence of concomitant therapies as a risk factor for ADR. Concomitant therapies were the treatments taken by participants to treat comorbid conditions. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
Concomitant Therapies: AbsentConcomitant Therapies: Present
Celsentri (Maraviroc) Tablets20.0038.46

[back to top]

Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Age

"An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. In this outcome measure percentage of participants with ADRs categorized by age are reported to assess age as a risk factor for ADR. >= refers to greater than or equal to." (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
>=15 and less than(<) 65 years>=65 and less than or equal to (<=) 70 years
Celsentri (Maraviroc) Tablets22.2240.00

[back to top]

Mean Number of Plasma Human Immuno-Deficiency Virus-Ribosomal Ribonucleic Acid (HIV-RNA) Copies: Factor The Presence or Absence of Comorbidities

HIV-RNA copy numbers were measured employing the TaqMan assay with the lower limit of detection of 40 copies/mL. The reported data for plasma HIV-RNA copies was calculated after logarithmic conversion. (NCT00864474)
Timeframe: Month 0 (Baseline), 3, 6, 9, 12, 15, 18, 21, 24, 27, 30, 33, 36, 39, 42, 45, 48, 51, 54, 57, 60, 63, 66, 69, 72, 75, 78, 81 and 84

InterventionLog HIV-RNA copies/mL (Mean)
Comorbidities-Present: Month 0 (Baseline)Comorbidities-Present: Month 3Comorbidities-Present: Month 6Comorbidities-Present: Month 9Comorbidities-Present: Month 12Comorbidities-Present: Month 15Comorbidities-Present: Month 18Comorbidities-Present: Month 21Comorbidities-Present: Month 24Comorbidities-Present: Month 27Comorbidities-Present: Month 30Comorbidities-Present: Month 33Comorbidities-Present: Month 36Comorbidities-Present: Month 39Comorbidities-Present: Month 42Comorbidities-Present: Month 45Comorbidities-Present: Month 48Comorbidities-Present: Month 51Comorbidities-Present: Month 54Comorbidities-Present: Month 57Comorbidities-Present: Month 60Comorbidities-Present: Month 63Comorbidities-Present: Month 69Comorbidities-Present: Month 75Comorbidities-Present: Month 78Comorbidities-Present: Month 84Comorbidities-Absent: Month 0 (Baseline)Comorbidities-Absent: Month 3Comorbidities-Absent: Month 6Comorbidities-Absent: Month 9Comorbidities-Absent: Month 12Comorbidities-Absent: Month 15Comorbidities-Absent: Month 18Comorbidities-Absent: Month 21Comorbidities-Absent: Month 27Comorbidities-Absent: Month 33Comorbidities-Absent: Month 36Comorbidities-Absent: Month 39Comorbidities-Absent: Month 42Comorbidities-Absent: Month 45Comorbidities-Absent: Month 48Comorbidities-Absent: Month 51Comorbidities-Absent: Month 57Comorbidities-Absent: Month 63Comorbidities-Absent: Month 69
Celsentri (Maraviroc) Tablets2.81.91.81.52.11.61.82.11.51.51.71.31.41.51.61.31.31.41.31.31.31.31.31.31.31.33.81.81.41.31.31.72.41.51.31.31.31.31.31.31.31.31.31.31.3

[back to top]

Mean Number of Plasma Human Immuno-Deficiency Virus-Ribosomal Ribonucleic Acid (HIV-RNA) Copies: Factor Gender

HIV-RNA copy numbers were measured employing the TaqMan assay with the lower limit of detection of 40 copies per milliliter (copies/mL). The reported data for plasma HIV-RNA copies was calculated after logarithmic conversion. (NCT00864474)
Timeframe: Month 0 (Baseline), 3, 6, 9, 12, 15, 18, 21, 24, 27, 30, 33, 36, 39, 42, 45, 48, 51, 54, 57, 60, 63, 66, 69, 72, 75, 78, 81 and 84

InterventionLog HIV-RNA copies/milliliter(mL) (Mean)
Males: Month 0 (Baseline)Males: Month 3Males: Month 6Males: Month 9Males: Month 12Males: Month 15Males: Month 18Males: Month 21Males: Month 24Males: Month 27Males: Month 30Males: Month 33Males: Month 36Males: Month 39Males: Month 42Males: Month 45Males: Month 48Males: Month 51Males: Month 54Males: Month 57Males: Month 60Males: Month 63Males: Month 69Males: Month 75Males: Month 78Males: Month 84Females: Month 0 (Baseline)Females: Month 3Females: Month 15
Celsentri (Maraviroc) Tablets3.11.91.61.41.91.71.81.91.51.41.71.31.41.41.51.31.31.3219.01294.51.31.31.31.31.31.31.31.31.3

[back to top]

Mean Number of Plasma HIV-RNA Copies: Factor The Presence or Absence of Use of Cytochrome P450 3A4 (CYP3A4) Enzyme Inducer Taken Along With Celsentri

HIV-RNA copy numbers were measured employing the TaqMan assay with the lower limit of detection of 40 copies/mL. CYP3A4 is an important enzyme in the body, mainly found in the liver and in the intestine. This enzyme is responsible for metabolism of majority of drugs. Many of the food substances and commonly used drugs act as inducers of enzyme CYP3A4. The reported data for plasma HIV-RNA copies was calculated after logarithmic conversion. (NCT00864474)
Timeframe: Month 0 (Baseline), 3, 6, 9, 12, 15, 18, 21, 24, 27, 30, 33, 36, 39, 42, 45, 48, 51, 54, 57, 60, 63, 66, 69, 72, 75, 78, 81 and 84

InterventionLog HIV-RNA copies/mL (Mean)
Present: Month 0 (Baseline)Present: Month 3Present: Month 6Present: Month 9Present: Month 12Present: Month 15Present: Month 18Present: Month 21Present: Month 24Present: Month 33Present: Month 36Present: Month 39Present: Month 42Present: Month 45Present: Month 48Present: Month 51Present: Month 57Present: Month 63Present: Month 69Present: Month 75Present: Month 78Present: Month 84Absent: Month 0 (Baseline)Absent: Month 3Absent: Month 6Absent: Month 9Absent: Month 12Absent: Month 15Absent: Month 18Absent: Month 21Absent: Month 24Absent: Month 27Absent: Month 30Absent: Month 33Absent: Month 36Absent: Month 39Absent: Month 42Absent: Month 45Absent: Month 48Absent: Month 51Absent: Month 54Absent: Month 57Absent: Month 60Absent: Month 63Absent: Month 69
Celsentri (Maraviroc) Tablets3.62.41.51.33.91.61.31.31.31.31.31.31.31.31.31.31.31.31.31.31.31.33.01.71.71.51.71.61.92.01.61.41.71.31.41.41.51.31.31.41.31.31.31.31.3

[back to top]

Mean Number of Plasma HIV-RNA Copies: Factor The Presence or Absence of History of Therapies for HIV Infection

HIV-RNA copy numbers were measured employing the TaqMan assay with the lower limit of detection of 40 copies/mL. The reported data for plasma HIV-RNA copies was calculated after logarithmic conversion. (NCT00864474)
Timeframe: Month 0 (Baseline), 3, 6, 9, 12, 15, 18, 21, 24, 27, 30, 33, 36, 39, 42, 45, 48, 51, 54, 57, 60, 63, 66, 69, 72, 75, 78, 81 and 84

InterventionLog HIV-RNA copies/mL (Mean)
Present: Month 0 (Baseline)Present: Month 3Present: Month 6Present: Month 9Present: Month 12Present: Month 15Present: Month 18Present: Month 21Present: Month 24Present: Month 27Present: Month 30Present: Month 33Present: Month 36Present: Month 39Present: Month 42Present: Month 45Present: Month 48Present: Month 51Present: Month 54Present: Month 57Present: Month 60Present: Month 63Present: Month 69Present: Month 75Present: Month 78Present: Month 84Absent: Month 0 (Baseline)Absent: Month 3Absent: Month 6Absent: Month 9Absent: Month 12Absent: Month 15Absent: Month 21Absent: Month 24Absent: Month 27Absent: Month 33Absent: Month 36Absent: Month 39Absent: Month 42
Celsentri (Maraviroc) Tablets2.41.61.71.41.81.61.82.11.51.41.71.31.41.41.51.31.31.31.31.31.31.31.31.31.31.34.72.21.61.62.01.71.31.31.31.31.31.31.3

[back to top]

Mean Number of Plasma HIV-RNA Copies: Factor The Centers for Disease Control and Prevention (CDC) Classification

Participants are divided into 3 categories as per CDC classification based on the level of HIV infection as follows: Category A= asymptomatic HIV-1 infection, persistent generalized lymphadenopathy and acute (primary) HIV-1 infection with accompanying illness or history of acute HIV-1 infection in an adult or adolescent aged >=13 years, Category B: conditions attributed to HIV infection or are indicative of a defect in cell-mediated immunity; or the conditions are considered by physicians to have a clinical course or to require management that is complicated by HIV infection in an HIV-infected adolescent or adult; and Category C: clinical conditions listed in the acquired immunodeficiency syndrome (AIDS) diagnostic criteria, corresponding to conventional AIDS. Once criteria C has occurred, the person will remain in Category C even if symptoms are alleviated. (NCT00864474)
Timeframe: Month 0 (Baseline), 3, 6, 9, 12, 15, 18, 21, 24, 27, 30, 33, 36, 39, 42, 45, 48, 51, 54, 57, 60, 63, 66, 69, 72, 75, 78, 81 and 84

InterventionLog HIV-RNA copies/mL (Mean)
CDC category A: Month 0 (Baseline)CDC category A: Month 3CDC category A: Month 6CDC category A: Month 9CDC category A: Month 12CDC category A: Month 15CDC category A: Month 18CDC category A: Month 21CDC category A: Month 24CDC category A: Month 27CDC category A: Month 33CDC category A: Month 36CDC category A: Month 39CDC category A: Month 42CDC category A: Month 45CDC category A: Month 48CDC category A: Month 51CDC category A: Month 57CDC category A: Month 63CDC category A: Month 69CDC category A: Month 75CDC category A: Month 78CDC category A: Month 84CDC category B: Month 0 (Baseline)CDC category B: Month 3CDC category B: Month 6CDC category B: Month 15CDC category B: Month 18CDC category B: Month 24CDC category B: Month 27CDC category B: Month 30CDC category B: Month 33CDC category B: Month 39CDC category B: Month 42CDC category B: Month 45CDC category B: Month 48CDC category B: Month 51CDC category B: Month 60CDC category B: Month 63CDC category C: Month 0 (Baseline)CDC category C: Month 3CDC category C: Month 6CDC category C: Month 9CDC category C: Month 12CDC category C: Month 15CDC category C: Month 18CDC category C: Month 21CDC category C: Month 24CDC category C: Month 27CDC category C: Month 30CDC category C: Month 33CDC category C: Month 36CDC category C: Month 39CDC category C: Month 42CDC category C: Month 51CDC category C: Month 54CDC category C: Month 57CDC category C: Month 60CDC category C: Month 69
Celsentri (Maraviroc) Tablets3.41.61.51.31.51.51.61.51.31.31.31.31.31.31.31.31.31.31.31.31.31.31.31.61.31.52.11.51.31.31.31.31.31.31.31.31.31.31.33.02.31.91.62.31.72.02.11.61.81.91.41.51.61.91.51.31.31.31.3

[back to top]

Mean Number of Plasma HIV-RNA Copies for Participants Who Took Concomitant Therapies Along With Celsentri

(NCT00864474)
Timeframe: Month 0 (Baseline), 3, 6, 9, 12, 15, 18, 21, 24, 27, 30, 33, 36, 39, 42, 45, 48, 51, 54, 57, 60, 63, 66, 69, 72, 75, 78, 81 and 84

InterventionLog HIV-RNA copies/mL (Mean)
At baselineAt Month 3At Month 6At Month 9At Month 12At Month 15At Month 18At Month 21At Month 24At Month 27At Month 30At Month 33At Month 36At Month 39At Month 42At Month 45At Month 48At Month 51At Month 54At Month 57At Month 60At Month 63At Month 69At Month 75At Month 78At Month 84
Celsentri (Maraviroc) Tablets3.11.81.61.41.91.61.81.91.51.41.71.31.41.41.51.31.31.31.31.31.31.31.31.31.31.3

[back to top]

Mean Number of CD4+ Lymphocyte: Factor The Presence or Absence of Use of Cytochrome P450 3A4 (CYP3A4) Enzyme Inducer Taken Along With Celsentri

CD4 cells are the white blood cells and act as laboratory marker providing an indication of immune functioning. A higher number is associated with better immune functioning. CYP3A4 is an important enzyme in the body, mainly found in the liver and in the intestine. This enzyme is responsible for metabolism of many of drugs. Many of the food substances and commonly used drugs act as inducers of enzyme CYP3A4. (NCT00864474)
Timeframe: Month 0 (Baseline), 3, 6, 9, 12, 15, 18, 21, 24, 27, 30, 33, 36, 39, 42, 45, 48, 51, 54, 57, 60, 63, 66, 69, 72, 75, 78, 81 and 84

InterventionCD4 cells/mm^3 (Mean)
Present: Month 0 (Baseline)Present: Month 3Present: Month 6Present: Month 9Present: Month 12Present: Month 15Present: Month 18Present: Month 21Present: Month 24Present: Month 33Present: Month 36Present: Month 39Present: Month 42Present: Month 45Present: Month 48Present: Month 51Present: Month 57Present: Month 63Present: Month 69Present: Month 75Present: Month 78Present: Month 84Absent: Month 0 (Baseline)Absent: Month 3Absent: Month 6Absent: Month 9Absent: Month 12Absent: Month 15Absent: Month 18Absent: Month 21Absent: Month 24Absent: Month 27Absent: Month 30Absent: Month 33Absent: Month 36Absent: Month 39Absent: Month 42Absent: Month 45Absent: Month 48Absent: Month 51Absent: Month 54Absent: Month 57Absent: Month 60Absent: Month 63Absent: Month 69
Celsentri (Maraviroc) Tablets200.7193.0212.0223.04.0231.3248.017.0182.0367.7478.0581.0334.0526.5589.0609.0519.0712.0583.0969.0787.0934.0320.0342.1342.1474.3426.2507.0406.5554.3525.4612.4376.0477.4508.0550.6887.11119.0607.7665.1219.02070.0652.0178.01429.0

[back to top]

Mean Number of CD4+ Lymphocyte Counts: Fcator The Presence or Absence of Comorbidities

CD4 cells are the white blood cells and act as laboratory marker providing an indication of immune functioning. A higher number is associated with better immune functioning. (NCT00864474)
Timeframe: Month 0 (Baseline), 3, 6, 9, 12, 15, 18, 21, 24, 27, 30, 33, 36, 39, 42, 45, 48, 51, 54, 57, 60, 63, 66, 69, 72, 75, 78, 81 and 84

InterventionCD4 cells/mm^3 (Mean)
Comorbidities-Present: Month 0 (Baseline)Comorbidities-Present: Month 3Comorbidities-Present: Month 6Comorbidities-Present: Month 9Comorbidities-Present: Month 12Comorbidities-Present: Month 15Comorbidities-Present: Month 18Comorbidities-Present: Month 21Comorbidities-Present: Month 24Comorbidities-Present: Month 27Comorbidities-Present: Month 30Comorbidities-Present: Month 33Comorbidities-Present: Month 36Comorbidities-Present: Month 39Comorbidities-Present: Month 42Comorbidities-Present: Month 45Comorbidities-Present: Month 48Comorbidities-Present: Month 51Comorbidities-Present: Month 54Comorbidities-Present: Month 57Comorbidities-Present: Month 60Comorbidities-Present: Month 63Comorbidities-Present: Month 69Comorbidities-Present: Month 75Comorbidities-Present: Month 78Comorbidities-Present: Month 84Comorbidities-Absent: Month 0 (Baseline)Comorbidities-Absent: Month 3Comorbidities-Absent: Month 6Comorbidities-Absent: Month 9Comorbidities-Absent: Month 12Comorbidities-Absent: Month 15Comorbidities-Absent: Month 18Comorbidities-Absent: Month 21Comorbidities-Absent: Month 27Comorbidities-Absent: Month 33Comorbidities-Absent: Month 36Comorbidities-Absent: Month 39Comorbidities-Absent: Month 42Comorbidities-Absent: Month 45Comorbidities-Absent: Month 48Comorbidities-Absent: Month 51Comorbidities-Absent: Month 57Comorbidities-Absent: Month 63Comorbidities -Absent: Month 69
Celsentri (Maraviroc) Tablets294.2282.0284.9361.5303.3422.9401.6308.4427.3558.7376.0357.0415.7476.4813.21017.5603.0714.1219.02070.0652.0178.01429.0969.0787.0934.0325.6388.0401.4523.7707.0419.7301.0777.5693.0605.8755.0788.5758.5628.0603.0406.0519.0712.0583.0

[back to top]

Mean Number of CD4+ Lymphocyte Counts: Factor The Centers for Disease Control and Prevention (CDC) Classification

CD4 cells are the white blood cells and act as a laboratory marker providing an indication of immune functioning. A higher number is associated with better immune functioning. Participants are divided into 3 categories as per CDC classification based on the level of HIV infection as: Category A= asymptomatic HIV-1 infection, persistent generalized lymphadenopathy and acute(primary)HIV-1 infection with accompanying illness or history of acute HIV-1 infection in an adult or adolescent aged>=13 years, Category B: conditions attributed to HIV infection or are indicative of a defect in cell-mediated immunity; or the conditions are considered by physicians to have a clinical course or to require management that is complicated by HIV infection in an HIV-infected adolescent or adult; and Category C: clinical conditions listed in the AIDS diagnostic criteria, corresponding to conventional AIDS. Once criteria C has occurred, the person will remain in Category C even if symptoms are alleviated. (NCT00864474)
Timeframe: Month 0 (Baseline), 3, 6, 9, 12, 15, 18, 21, 24, 27, 30, 33, 36, 39, 42, 45, 48, 51, 54, 57, 60, 63, 66, 69, 72, 75, 78, 81 and 84

InterventionCD4 cells/mm^3 (Mean)
CDC category A: Month 0 (Baseline)CDC category A: Month 3CDC category A: Month 6CDC category A: Month 9CDC category A: Month 12CDC category A: Month 15CDC category A: Month 18CDC category A: Month 21CDC category A: Month 24CDC category A: Month 27CDC category Month 33CDC category A: Month 36CDC category A: Month 39CDC category A: Month 42CDC category A: Month 45CDC category A: Month 48CDC category A: Month 51CDC category A: Month 57CDC category A: Month 63CDC category A: Month 69CDC category A: Month 75CDC category A: Month 78CDC category A : Month 84CDC category B: Month 0 (Baseline)CDC category B: Month 3CDC category B: Month 6CDC category B: Month 15CDC category B: Month 18CDC category B: Month 24CDC category B: Month 27CDC category B: Month 30CDC category B: Month 33CDC category B: Month 39CDC category B: Month 42CDC category B: Month 45CDC category B: Month 48CDC category B: Month 51CDC category B: Month 60CDC category B: Month 63CDC category C: Month 0 (Baseline)CDC category C: Month 3CDC category C: Month 6CDC category C: Month 9CDC category C: Month 12CDC category C: Month 15CDC category C: Month 18CDC category C: Month 21CDC category C: Month 24CDC category C: Month 27CDC category C: Month 30CDC category C: Month 33CDC category C: Month 36CDC category C: Month 39CDC category C: Month 42CDC category C: Month 51CDC category C: Month 54CDC category C: Month 57CDC category C: Month 60CDC category C: Month 69
Celsentri (Maraviroc) Tablets347.0416.2428.3457.4524.8441.2401.3777.5567.5784.8531.7616.5719.3617.0621.3596.0609.0519.0712.0583.0969.0787.0934.0264.3227.0122.0447.5371.0207.0180.0218.0137.0201.01221.01427.0610.0219.0149.0178.0255.9165.0207.0315.3243.2389.2396.1308.4412.3411.7455.0398.8384.5519.1848.81111.2219.02070.01155.01429.0

[back to top]

Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Presence or Absence of Hemophilia

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. In this outcome measure percentage of participants with ADRs categorized by presence or absence of hemophilia are reported to assess presence or absence of hemophilia as a risk factor for ADR. Hemophilia is a bleeding disorder that slows the blood clotting process. Participants with this condition experience prolonged bleeding or oozing following an injury, surgery, or having a tooth pulled. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
Hemophilia: AbsentHemophilia: Present
Celsentri (Maraviroc) Tablets22.9528.57

[back to top]

Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Presence or Absence of Comorbidities

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. In this outcome measure percentage of participants with ADRs categorized by presence or absence of comorbidities are reported to assess presence or absence of comorbidities as a risk factor for ADR. Comorbidity referred to the presence of co-existing or additional diseases along with HIV infection. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
Comorbidities: AbsentComorbidities: Present
Celsentri (Maraviroc) Tablets15.3825.45

[back to top]

Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Presence or Absence of Allergies

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. In this outcome measure percentage of participants with ADRs categorized by presence or absence of allergies are reported to assess presence or absence of allergies as a risk factor for ADR. Unknown: participants for which the presence or absence of allergies was not known. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
Allergies: AbsentAllergies: PresentAllergies: Unknown
Celsentri (Maraviroc) Tablets18.5238.46100.00

[back to top]

Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Number of Concomitant Anti-HIV Drugs Use

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. In this outcome measure percentage of participants with ADRs categorized by number of concomitant anti-HIV treatment are reported to assess number of concomitant anti-HIV treatment as a risk factor for ADR. Concomitant drugs refers to the drugs other than Celsentri. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
1 drug2 drugs3 drugs>=4 drugs
Celsentri (Maraviroc) Tablets0.0023.5325.0027.78

[back to top]

Mean Number of CD4+ Lymphocyte Counts: Factor Presence or Absence of History of Therapies for HIV Infection

CD4 cells are the white blood cells and act as a laboratory marker providing an indication of immune functioning. A higher number is associated with better immune functioning. (NCT00864474)
Timeframe: Month 0 (Baseline), 3, 6, 9, 12, 15, 18, 21, 24, 27, 30, 33, 36, 39, 42, 45, 48, 51, 54, 57, 60, 63, 66, 69, 72, 75, 78, 81 and 84

InterventionCD4 cells/mm^3 (Mean)
Present: Month 0Present: Month 3Present: Month 6Present: Month 9Present: Month 12Present: Month 15Present: Month 18Present: Month 21Present: Month 24Present: Month 27Present: Month 30Present: Month 33Present: Month 36Present: Month 39Present: Month 42Present: Month 45Present: Month 48Present: Month 51Present: Month 54Present: Month 57Present: Month 60Present: Month 63Present: Month 69Present: Month 75Present: Month 78Present: Month 84Absent: Month 0Absent: Month 3Absent: Month 6Absent: Month 9Absent: Month 12Absent: Month 15Absent: Month 21Absent: Month 24Absent: Month 27Absent: Month 33Absent: Month 36Absent: Month 39Absent: Month 42
Celsentri (Maraviroc) Tablets334.8263.0297.6393.7373.2443.2393.3520.6496.2607.1376.0412.1540.0599.9804.7822.8603.0637.1219.01294.5652.0445.01006.0969.0787.0934.0225.6382.2371.0426.8394.8306.5353.014.0660.0801.0382.0236.0746.0

[back to top]

Percentage of Participants With Adverse Drug Reactions (ADRs)

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
Celsentri (Maraviroc) Tablets23.53

[back to top]

Number of Participants With Unknown Adverse Drug Reactions (ADRs)

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. Unknown ADRs were the ADRs those were not listed on the package insert. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

InterventionParticipants (Count of Participants)
Celsentri (Maraviroc) Tablets8

[back to top]

Number of Participants With Tropism Switch From CCR5- to CXCR4-Tropic Variants

CCR5= C-C chemokine receptor type 5 and CXCR4= C-X-C chemokine receptor type 4. Tropism switch is the mutation of CCR5-tropic HIV-1 to a CXCR4-using virus. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

InterventionParticipants (Count of Participants)
Celsentri (Maraviroc) Tablets0

[back to top]

Number of Adverse Drug Reactions (ADRs) Considered to Have Occurred Due to Effect of Celsentri on Immune Function

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

InterventionADRs (Number)
Celsentri (Maraviroc) Tablets0

[back to top]

Number of Adverse Drug Reactions (ADRs) Considered to Have Occurred Due to Effect of Celsentri on Hepatic Function

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

InterventionADRs (Number)
Celsentri (Maraviroc) Tablets3

[back to top]

Number of Adverse Drug Reactions (ADRs) Considered to Have Occurred Due to Effect of Celsentri on Cardiovascular Effects

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

InterventionADRs (Number)
Celsentri (Maraviroc) Tablets0

[back to top]

Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Mean Total Dose of Celsentri

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. In this outcome measure percentage of participants with ADRs categorized by mean total dose of Celsentri are reported to assess mean total dose of Celsentri as a risk factor for ADR. One tablet of Celsentri had a dose of 150 mg. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
4 to <=360 tablets> 360 and <=730 tablets> 730 and <=1460 tablets> 1460 and <=21632 tablets
Celsentri (Maraviroc) Tablets7.353.280.0017.07

[back to top]

Mean Number of Cluster of Differentiation of More Than 4 (CD4+) Lymphocyte Count: Factor Gender

CD4+ Lymphocyte were counted by CD4 cells per cubic millimeter (cells/mm^3). CD4 cells are the white blood cells and act as laboratory marker providing an indication of immune functioning. A higher number is associated with better immune functioning. (NCT00864474)
Timeframe: Month 0 (Baseline), 3, 6, 9, 12, 15, 18, 21, 24, 27, 30, 33, 36, 39, 42, 45, 48, 51, 54, 57, 60, 63, 66, 69, 72, 75, 78, 81 and 84

InterventionCD4 cells/mm^3 (Mean)
Males: Month 0 (Baseline)Males: Month 3Males: Month 6Males: Month 9Males: Month 12Males: Month 15Males: Month 18Males: Month 21Males: Month 24Males: Month 27Males: Month 30Males: Month 33Males: Month 36Males: Month 39Males: Month 42Males: Month 45Males: Month 48Males: Month 51Males: Month 54Males: Month 57Males: Month 60Males: Month 63Males: Month 69Males: Month 75Males: Month 78Males: Month 84Females: Month 0 (Baseline)Females: Month 3Females: Month 15
Celsentri (Maraviroc) Tablets301.2318.2319.2405.7384.0440.9393.3464.7427.3612.4376.0447.5500.5554.4794.9822.8603.0637.1219.01294.5652.0445.01006.0969.0787.0934.0335.0122.0197.0

[back to top]

Primary: Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Presence or Absence of History of Therapies for Human Immuno-Deficiency Virus (HIV) Infection

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. In this outcome measure percentage of participants with ADRs categorized by presence or absence of history of therapies for HIV Infection are reported to assess presence or absence of history of therapies for HIV Infection as a risk factor for ADR. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
History of Therapies for HIV Infection: AbsentHistory of Therapies for HIV Infection: Present
Celsentri (Maraviroc) Tablets18.7525.00

[back to top]

Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Total Number of Days of Administration of Celsentri

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. In this outcome measure percentage of participants with ADRs categorized by total number of days of administration of Celsentri are reported to assess total number of days of administration of Celsentri as a risk factor for ADR. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
2 to <=180 days> 180 and <=365 days> 365 and <=730 days> 730 and <= 2704 days
Celsentri (Maraviroc) Tablets8.821.677.698.57

[back to top]

Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Presence or Absence of Use of Cytochrome P450 3A4 (CYP3A4) Enzyme Inducer Taken Along With Celsentri

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. In this outcome measure percentage of participants with ADRs categorized by presence or absence of concomitant CYP3A4 enzyme inducer use are reported to assess presence or absence of concomitant CYP3A enzyme inducer use as a risk factor for ADR. CYP3A4 is an important enzyme in the body, mainly found in the liver and in the intestine. This enzyme is responsible for metabolism of majority of drugs. Many of the food substances and commonly used drugs act as inducer for enzyme CYP3A4. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
CYP3A Enzyme Inducer Use: AbsentCYP3A Enzyme Inducer Use: Present
Celsentri (Maraviroc) Tablets21.8230.77

[back to top]

Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Presence or Absence of Renal Impairment

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. In this outcome measure percentage of participants with ADRs categorized by presence or absence of renal impairment are reported to assess presence or absence of renal impairment as a risk factor for ADR. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
Renal Impairment: AbsentRenal Impairment: Present
Celsentri (Maraviroc) Tablets20.9750.00

[back to top]

Percentage of Participants With Adverse Drug Reactions (ADRs): Factor Presence or Absence of Hepatic Impairment

An ADR was any untoward medical occurrence attributed to Celsentri tablets in a participant who received Celsentri tablets. Relatedness to Celsentri tablets was assessed by the physician. In this outcome measure percentage of participants with ADRs categorized by presence or absence of hepatic impairment are reported to assess presence or absence of hepatic impairment as a risk factor for ADR. (NCT00864474)
Timeframe: From April 2009 to December 2018 (up to approximately 8 years 8 months)

Interventionpercentage of participants (Number)
Hepatic Impairment: AbsentHepatic Impairment: Present
Celsentri (Maraviroc) Tablets21.7427.27

[back to top]

Trough Plasma and Tissue Drug Levels in Volunteers at the Time of the Upper Endoscopy

The reported drug level is for the primary ART agent for that cohort. For the maraviroc arm, maraviroc plasma and tissue levels are reported. For the maraviroc plus raltegravir arm, the raltegravir plasma and tissue levels are reported. For the efavirenz arm, the efavirenz plasma and tissue levels are reported. HIV negative controls were not on ART and did not have drug levels measured. (NCT00870363)
Timeframe: nine months

,,
Interventionng/mL (Median)
plasma primary ARTduodenal tissue primary ART
Efavirenz or Other NNRTI With 2 NRTIs245911.1
Maraviroc in Combination With 2 NRTIs94.50.7
Maraviroc PLUS Raltegravir in Combination With 2 NRTIs3970.1

[back to top]

Change in HIV DNA Per 10^6 Cells in Duodenal Tissue Versus PBMC by Drug Regimen Received

single-cell suspension of digested duodenal tissue and Ficol-Hypaque separated PBMC underwent HIV-DNA PCR (NCT00870363)
Timeframe: Baseline and nine months

,,
Interventioncopies/10^6 cells (Mean)
PBMC HIV-DNADuodenal HIV-DNA
Efavirenz or Other NNRTI With 2 NRTIs-1330-325
Maraviroc in Combination With 2 NRTIs-1709-16
Maraviroc PLUS Raltegravir in Combination With 2 NRTIs-2500-846

[back to top]

Changes in CD4+ T-cell Numbers by Treatment Regimen

peripheral absolute CD4+ T-cell counts increase from baseline to 9 months of cART by commercial assay (NCT00870363)
Timeframe: Baseline and nine months

Interventioncells/mL (Mean)
Maraviroc in Combination With 2 NRTIs221
Maraviroc PLUS Raltegravir in Combination With 2 NRTIs231
Efavirenz or Other NNRTI With 2 NRTIs194

[back to top]

Change in the Density of CD3+/CD4+ Cells Per Cubic Millimeter at the Effector Sites in the Duodenal Tissues Following Antiretroviral Therapy Regimen

immunohistochemistry for CD3+/CD4+ cells counted manually within the lamina propria (NCT00870363)
Timeframe: Baseline and nine months for 3 treatment cohorts and Baseline for the control group, which was only assessed at one time point

Interventioncells/mm^2 (Mean)
Maraviroc in Combination With 2 NRTIs24
Maraviroc PLUS Raltegravir in Combination With 2 NRTIs89
Efavirenz or Other NNRTI With 2 NRTIs119
HIV Negative Controls Not on ART566

[back to top]

Change in CD4+/CD8+ T-cell Immune Activation, Maturation, Regulatory and Apoptosis Markers at Baseline and Weeks 4 and 24.

To compare the percent change of CD4+/CD8+ T-cell (NCT00925756)
Timeframe: Baseline to Week 24

Interventioncells/mm^3 (Median)
Wk 4 CD4+ totalWk 4 CD4+ naiveWk 4 CD4+ central memoryWk 4 CD4+ effector memoryWk 4 CD4+ effectorWk 4 CD4+ immune senescenceWk 24 CD4+ totalWk 24 CD4+ naiveWk 24 CD4+ central memoryWk 24 CD4+ effector memoryWk 24 CD4+ effectorWk 24 CD4+ immune senescenceWk 4 CD8+ totalWk 4 CD8+ naiveWk 4 CD8+ central memoryWk 4 CD8+ effector memoryWk 4 CD8+ effectorWk 4 CD8+ immune senescenceWk 24 CD8+ totalWk 24 CD8+ naiveWk 24 CD8+ central memoryWk 24 CD8+ effector memoryWk 24 CD8+ effectorWk 24 CD8+ immune senescence
Maraviroc Intensification0.62-0.470.43-0.330.00-0.121.120.52-0.15-0.300.00-0.250.90-0.450.830.73-0.90-1.832.38-0.150.750.80-2.68-3.30

[back to top]

Differences in Gene Expression Profiles Obtained at Baseline and Week 4 and Week 24.

"To determine differential gene expression in T-Cells due to MVC exposure between week 0, 4 and 24 weeks.~Repeated measures (RM) ANOVA was used to identify genes whose expression changed over the course of MVC administration. Multivariate permutation tests under default settings (80% confident no more than 10% false positives) were performed using BRB-Array Tools. Gene assignment to temporal profiles was performed using a non- parametric clustering algorithm in Short Time-series Expression Miner (STEM)" (NCT00925756)
Timeframe: Baseline to Week 24

Intervention-fold TNF downregulation (Number)
Maraviroc Intensification1.44

[back to top]

CD4+ T-cell Absolute Count and Percentage at Baseline, Weeks 4 and 24.

To compare the CD4+/CD8+ T-cell absolute count and percentage change at Weeks 4 and 24 from Baseline. Wilcoxon signed rank test was used to assess changes in T cell counts, percentages, CD4+ T cell recovery slopes and changes in T cell phenotypes measured by flow cytometry. (NCT00925756)
Timeframe: Baseline to Week 24

Interventioncells/mm^3 (Median)
Wk 4 CD4+ cells (absolute)Wk 4 CD8+ cells (absolute)Wk 24 CD4+ cells (absolute)Wk 24 CD8+ cells (absolute)
Maraviroc Intensification6.504636.75100.00

[back to top]

Rate of Early Mortality After Transplant

Number of participants who died without relapse within 1 year of SCT (NCT00948753)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Phase 1: 150mg Maraviroc2
Phase 1: 300mg Maraviroc0
Phase 2: 300mg Maraviroc4

[back to top]

Number of Patients Treated With Maraviroc During SCT That Develop Chronic GVHD

count of how many patients treated with Maraviroc during SCT go on to develop chronic GVHD in 1 year (NCT00948753)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Phase 1: 150mg Maraviroc1
Phase 1: 300mg Maraviroc3
Phase 2: 300mg Maraviroc2

[back to top]

Number of Participants Who Relapsed During Study Period

Number of participants who received Maraviroc during SCT who relapsed within 1 year and 11 months. This was based on a diagnosis made by their physician that their primary cancer had returned. (NCT00948753)
Timeframe: 1 year and 11 months

InterventionParticipants (Count of Participants)
Phase 1: 150mg Maraviroc2
Phase 1: 300mg Maraviroc2
Phase 2: 300mg Maraviroc15

[back to top]

Efficacy of Maraviroc

"Efficacy is measured by number of participants progressing to acute GVHD. If acute GVHD is noted in a participant following exposure to study drug, then efficacy was not achieved.~If no GVHD was noted following exposure, then efficacy was achieved in that participant" (NCT00948753)
Timeframe: 8 weeks

InterventionParticipants (Number)
Phase 1: 150mg Maraviroc0
Phase 1: 300mg Maraviroc0
Phase 2: 300mg Maraviroc0

[back to top]

Pharmacokinetic Profile of Maraviroc in Patients Undergoing Nonmyeloablative Allogeneic SCT

Plasma maraviroc levels were measured in the blood with a target level of 100 ng per milliliter. Blood was drawn on Day 0 and Day 10-12 at pre-dose, 1, 2, 3, 4, 6, and 12 hours post-dose. Data was analyzed looking at the number of patients to achieve the target of 100 ng per milliliter at any time point. (NCT00948753)
Timeframe: pre-dose, 1,2,3,4,6,12 hours post-dose

Interventionnumber of patients to reach target (Number)
Phase 1: 150mg Maraviroc4
Phase 1: 300mg Maraviroc6
Phase 2: 300mg Maraviroc0

[back to top]

Safety of Maraviroc

number of Adverse Events following exposure to Maraviroc (NCT00948753)
Timeframe: 1 year

InterventionNumber of AEs (Number)
Phase 1: 150mg Maraviroc8
Phase 1: 300mg Maraviroc6
Phase 2: 300mg Maraviroc18

[back to top]

Change From Baseline in HIV DNA in Rectal Tissue at Week 56

(NCT00976404)
Timeframe: Week 56

Interventionlog^10 copies per 10^6 cells (Median)
Maraviroc + Raltegravir Intensification0.08
Maraviroc + Raltegravir Intens. Plus DNA + HIV-rAd5 Vaccine-0.02

[back to top]

Serious Adverse Events Attributed to Study Treatments

Grade 3 or 4 serious adverse events related to study treatments (raltegravir, maraviroc, or HIV-recombinant Ad5-based vaccine) (NCT00976404)
Timeframe: 56 weeks

Interventionserious adverse events (Number)
Maraviroc + Raltegravir Intensification1
Maraviroc + Raltegravir Intens. Plus DNA + HIV-rAd5 Vaccine3

[back to top]

HIV Specific T-cell Response to Env

HIV-specific immunity: Interferon gamma ELISpot response to Env (clades A) at week 36 (one month after rAd5 boosting) (NCT00976404)
Timeframe: 36 weeks

Interventionresponse per 10^6 PBMCs (Median)
Maraviroc + Raltegravir Intensification28
Maraviroc + Raltegravir Intens. Plus DNA + HIV-rAd5 Vaccine86

[back to top]

Change From Baseline in HIV DNA in PBMCs at Week 56

(NCT00976404)
Timeframe: 56 weeks

Interventionlog^10 copies per 10^6 PBMCs (Median)
ART Intensification: Maraviroc +Raltegravir0.00
Maraviroc + Raltegravir Plus DNA + HIV-rAd5 Vaccine0.04

[back to top]

Change From Baseline in CD4+ T Cell Count at Week 56

(NCT00976404)
Timeframe: Week 56

Interventioncells per mm^3 (Median)
Maraviroc + Raltegravir Intensification-1
Maraviroc + Raltegravir Intens. Plus DNA + HIV-rAd5 Vaccine23

[back to top]

Number of Participants With Adverse Events

To evaluate the short term safety and tolerability of maraviroc in HIV-1 infected subjects on stable antiretroviral therapy (NCT00982878)
Timeframe: 15 days

InterventionParticipants (Count of Participants)
Maraviroc0

[back to top]

CSF (Cerebrospinal Fluid) : Plasma Ratio of Maraviroc 1H Magnetic Resonance Spectroscopy (1H-MRS)

"To describe the CNS exposure of maraviroc in HIV-1 infected subjects receiving a stable antiretroviral regimen, including maraviroc, at steady state.~It were assessed by in vivo cerebral (1)H magnetic resonance spectroscopy ((1)H-MRS).~Cerebral MRS imaging (T1- and T2-weighted images) was performed on a Phillips Achieva™ 1.5 Tesla magnetic resonance (MR) scanner (Phillips NV, Best, Netherlands) and studied by an experienced neuroradiologist" (NCT00982878)
Timeframe: 15 days

Interventionratio (Mean)
Maraviroc1.01

[back to top]

Change From Baseline to 24 Weeks in Neuropsychological Performance As Measured by Age- and Education-Adjusted Z-Scores

The Z-score represents the number of standard deviations away from the mean, with positive Z-scores representing better neuropsychological performance and negative Z-scores representing poorer neuropsychological performance. Z-scores have been adjusted based on age- and education-matched norms. (NCT00987948)
Timeframe: Baseline to 24 Weeks

InterventionZ-score (Median)
Maraviroc0.57

[back to top]

Change From Baseline to 24 Weeks in HIV DNA (Log-10 Copies/10^6 Cells) as Measured by HIV DNA Within CD14+ Peripheral Blood Mononuclear Cells

Week 24 minus baseline (NCT00987948)
Timeframe: Baseline to 24 weeks

InterventionLog-10 copies/10^6 cells (Median)
Maraviroc0.58

[back to top]

Drug Adherence, Number of Participants With Missed Doses

Drug adherence, assessed as number of participants with missed doses over four-day recall (NCT00993148)
Timeframe: Week 24

Interventionparticipants (Number)
Maraviroc + Darunavir/Ritonavir0

[back to top]

Drug Resistance Mutations and Co-receptor Tropism Assessed by Trofile ES

(NCT00993148)
Timeframe: At study entry and at the time of virologic failure

Interventionparticipants (Number)
Maraviroc + Darunavir/Ritonavir0

[back to top]

Median CD4 Count Change From Baseline

Median changes from baseline in peripheral CD4+ T-cell count (NCT00993148)
Timeframe: 96 weeks

Interventioncells per mm^3 (Median)
Maraviroc + Darunavir/Ritonavir247

[back to top]

Percentage of Participants With Plasma HIV-1 RNA >50

Percentage of participants with confirmed plasma HIV-1 RNA > 50 copies/mL (NCT00993148)
Timeframe: 24 weeks

Interventionpercentage of participants (Number)
Maraviroc + Darunavir/Ritonavir12.5

[back to top]

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

Percentage of participants with confirmed plasma HIV-1 RNA level >50 copies/mL (NCT00993148)
Timeframe: 48 weeks

Interventionpercentage of participants (Number)
Maraviroc + Darunavir/Ritonavir8.3

[back to top]

Percentage of Participants With Virologic Failure or Off Study Treatment Regimen

Percentage of participants with virologic failure (confirmed plasma HIV-1 RNA > 50 copies/mL) or off study treatment regimen (composite end point) (NCT00993148)
Timeframe: 24 weeks

Interventionpercentage of participants (Number)
Maraviroc + Darunavir/Ritonavir12.5

[back to top]

Proportion of Participants With Plasma HIV-1 RNA >50 Copies/mL

Proportion of participants with confirmed plasma HIV-1 RNA level >50 copies/mL (NCT00993148)
Timeframe: 96 weeks

Interventionpercentage of participants (Number)
Maraviroc + Darunavir/Ritonavir10

[back to top]

Signs/Symptoms or Laboratory Toxicities of Grade 3 or Higher

Signs/symptoms or laboratory toxicities of Grade 3 or higher, or of any grade which led to a permanent change or discontinuation of study treatment regimen (NCT00993148)
Timeframe: 96 weeks

Interventionparticipants (Number)
Maraviroc + Darunavir/Ritonavir1

[back to top]

Trough Concentrations (Ctrough) of Maraviroc

Average trough concentration (Ctrough) of maraviroc (NCT00993148)
Timeframe: 24 hours

Interventionng/mL (Mean)
Maraviroc + Darunavir/Ritonavir39.3

[back to top]

Number of Participants With HIV RNA < 50 and <400 Copies/ml.

(NCT01068873)
Timeframe: week 24

InterventionParticipants (Count of Participants)
Open Label Single Arm1

[back to top]

Change in Proviral HIV-1 DNA in Total CD4+ T-cells From Baseline to Week 48 in Participants Randomized to the Intensified Arm Versus the Control Arm Who Received Placebo in Addition to Standard HAART.

The level of HIV Provirus in CD4 T cells obtained from peripheral blood at 48 weeks compared to baseline. A quantitative HIV PCR assay was done. The mean/median values from the standard HAART group is compared to the intensive HAART treatment regimen. (NCT01154673)
Timeframe: Baseline to Week 48

InterventionHIV DNA copies/ million CD4 cells (Median)
Intensive HAART279
Placebo Arm244

[back to top]

Viral Load

Percentage of subjects with HIV-1 viral load < 50 copies/ml (NCT01204905)
Timeframe: 48 weeks

InterventionParticipants (Count of Participants)
Open Label ART4

[back to top]

Viral Suppression

Time to attainment of virologic suppression (NCT01204905)
Timeframe: 48 weeks

Interventionweeks (Number)
Open Label ART6

[back to top]

Percentage of Participants With Plasma Human Immuno Deficiency Virus-1 Ribonucleic Acid (HIV-1 RNA) Load <50 Copies/Milliliter (mL) at 48 Weeks.

Participants' responder status at Week 48 was assessed according to Missing, discontinuation= Failure (MDF) algorithm. This algorithm treats all participants with HIV 1 RNA data missing at the time of interest or discontinuation of study drug as failures or non responders. (NCT01275625)
Timeframe: 48 weeks

InterventionPercentage of participants (Number)
Maraviroc+Combivir77.92

[back to top]

Number of Participants With Genotypic Resistance.

The viral genotypes were captured at Baseline and at treatment failure or Early termination and any resistance-associated mutations summarized descriptively at Week 48 for the Nucleotide reverse transcriptase inhibitors (NRTIs), and non-NRTIs (NNRTIs)drug classes. (NCT01275625)
Timeframe: Screening to Week 48 or Time of treatment Failure

InterventionParticipants (Number)
Participants with genotype reportedParticipants with Emergent NRTI mutations
Maraviroc+Combivir85

[back to top]

Number of Participants With HIV-1 RNA Tropism Status Using Genotyping Assay at Screening and at the Time of Virologic Failure.

Change in tropism were summarized at the time of treatment failure or Early Termination (note: this was performed for participants with viral load > 400 copies/mL only). (NCT01275625)
Timeframe: Screening to Week 48 or Time of treatment Failure

InterventionParticipants (Number)
Tropism change (n= 8)Reduced Maraviroc susceptibility (n= 3)
Maraviroc+Combivir00

[back to top]

Virologic Response: Percentage of Participants With Plasma HIV-1 RNA Load < 400 Copies/mL at Post-baseline Visits.

Participants' responder status at Week 48 was assessed according to MDF algorithm. This algorithm treats all participants with HIV 1 RNA data missing at the time of interest or discontinuation of study drug as failures or non responders. (NCT01275625)
Timeframe: Baseline, Week 4, Week 8, Week 12, Week 20, Week 24, Week 36 and Week 48

InterventionPercentage of participants (Number)
Baseline (N= 77)Week 4 (N= 77)Week 8 (N= 75)Week 12 (N= 76)Week 20 (N= 73)Week 24 (N= 73)Week 36 (N= 68)Week 48 (N= 65)
Maraviroc+Combivir0.0041.5672.0089.4791.7891.7897.0695.38

[back to top]

Virologic Response: Percentage of Participants With Plasma HIV-1 RNA Load <50 Copies/mL at Post-baseline Visits.

Participants' responder status at Week 48 was assessed according to MDF algorithm. This algorithm treats all participants with HIV 1 RNA data missing at the time of interest or discontinuation of study drug as failures or non responders. (NCT01275625)
Timeframe: Baseline, Week 4, Week 8, Week 12, Week 20, Week 24, Week 36 and Week 48

InterventionPercentage of participants (Number)
Baseline (N= 77)Week 4 (N= 77)Week 8 (N= 75)Week 12 (N= 76)Week 20 (N= 73)Week 24 (N= 73)Week 36 (N= 68)Week 48 (N= 65)
Maraviroc+Combivir0.007.7926.6748.6878.0883.5689.7192.31

[back to top]

Virologic Response: Rate of Virologic Failure at Week 48.

Virologic failure defined as: failure to achieve a reduction from baseline in HIV 1 RNA ≥ 0.5 log10 copies /mL by the second viral load determination (unless viral load was below the lower limit level of quantification [LLOQ]); or a ≥ 0.5 log10 increase from nadir in HIV 1 RNA after achieving a HIV 1 RNA reduction from BL >0.5 log10 copies/mL; or a HIV 1 RNA level of >1000 copies/mL after having achieved a HIV 1 RNA level below LLOQ. Participants with Time to loss of virologic response (defined by level of <50 copies/mL) failure were classified as rebounders or non-responders. (NCT01275625)
Timeframe: 48 weeks

InterventionParticipants (Number)
RespondersReboundersNon-responseDeathDiscontinued before Week 48 due to Adverse EventsDiscontinued before Week 48 for other reasons
Maraviroc+Combivir60115149

[back to top]

Immunological Response at Week 48: Absolute Change From Baseline in Absolute Cluster of Differentiation 4 (CD4)

Immunological Response was summarized using absolute change from Baseline to Week 48 in absolute CD4+ cell count (NCT01275625)
Timeframe: Week 48

Interventioncells/microliter (cells/mcL) (Mean)
Maraviroc+Combivir165.53

[back to top]

Immunological Response at Week 48: Absolute Change From Baseline in Absolute Cluster of Differentiation 8 (CD8)

Immunological Response was summarized using absolute change from Baseline to Week 48 in absolute CD8+ cell count. (NCT01275625)
Timeframe: Week 48

Interventioncells/mcL (Mean)
Maraviroc+Combivir-112.96

[back to top]

Immunological Response at Week 48: Change From Baseline in Absolute Cluster of Differentiation 4 (CD4)/ Cluster of Differentiation 8 (CD8) Ratio.

Immunological Response was summarized using absolute change from Baseline to Week 48 in absolute CD4+/ CD8+ ratio. (NCT01275625)
Timeframe: Week 48

InterventionRatio (Mean)
Maraviroc+Combivir0.33

[back to top]

Immunological Response at Week 48: Percentage Change From Baseline in Absolute Cluster of Differentiation 4 (CD4)

Immunological Response was summarized using percentage change from Baseline to Week 48 in absolute CD4+ cell count (NCT01275625)
Timeframe: Week 48

InterventionPercent (Mean)
Maraviroc+Combivir8.07

[back to top]

Immunological Response at Week 48: Percentage Change From Baseline in Absolute Cluster of Differentiation 8 (CD8)

Immunological Response was summarized using percentage change from Baseline to Week 48 in absolute CD8+ cell count. (NCT01275625)
Timeframe: Week 48

InterventionPercent (Mean)
Maraviroc+Combivir-8.59

[back to top]

Change in Edema Grade

The presence and extent of lower extremity edema was assessed and graded on a scale from 0 to 2 in patients with a higher grade indicating a greater level of edema using the Division of AIDS Table for Grading the Severity of Adult and Pediatric Adverse Events (DAIDS Adverse Events (AE) Grading Table), Version 1.0. Edema grade was recorded at baseline was compared with edema grades recorded at week 96, or at last assessment if participant withdrew from study prior to week 96 and an change in grade was calculated to examine whether or not a decrease in overall grade was observed. A negative value would indicate an overall decrease in the grade of lower extremity edema, and positive value would indicate an overall increase in lower extremity edema. (NCT01276236)
Timeframe: Up to 96 weeks

Interventionscores on a scale (Mean)
Treatment Arm (Maraviroc)-0.7

[back to top]

Number of Participants With a Decrease in Kaposi's Sarcoma (KS) Total Surface Area

To assess improvements in disease, up to five bi-dimensionally measurable cutaneous KS lesions were selected as marker lesions. The collective surface area of the marker lesions was evaluated over the course of the study for either an increase or decrease in the total surface area of lesions using the modified AIDS Clinical Trials Group (ACTG) Oncology Committee Staging Criteria. (NCT01276236)
Timeframe: Up to 96 weeks

InterventionParticipants (Count of Participants)
Treatment Arm (Maraviroc)11

[back to top]

Percent Change in CCR5 Levels on CD4+ T-Cells

Maraviroc works by binding to the C-C chemokine receptor 5 (CCR5) on T-cells and thereby blocking viral entry into CD4+ T-cells. Peripheral blood mononuclear cells (PBMC) were obtained from blood draws at each visit and T-cell immunophenotyping was performed from the baseline visit and final follow-up visit to determine the percent change in CCR5 levels on CD4+ T-Cells (NCT01276236)
Timeframe: Up to 96 weeks

Interventionpercentage change (Mean)
Treatment Arm (Maraviroc)22.8

[back to top]

Percent Change in CCR5 Levels on CD8+ T-cells

Maraviroc works by binding to the C-C chemokine receptor 5 (CCR5) on T-cells and thereby blocking viral entry into CD8+ T-cells. Peripheral blood mononuclear cells (PBMC) were obtained from blood draws at each visit and T-cell immunophenotyping was performed from the baseline visit and final follow-up visit to determine the percent change in CCR5 levels on CD8+ T-Cells (NCT01276236)
Timeframe: Up to 96 weeks

Interventionpercentage change (Mean)
Treatment Arm (Maraviroc)21.6

[back to top]

Percent Change in CD69 Expression in a Subset of Double Negative DR-CD38 Negative (DR-CD38-) T-cells

Peripheral blood mononuclear cells (PBMC) were obtained from blood draws at each visit and T-cell immunophenotyping was performed from the baseline visit and final follow-up visit to determine the percent change in CD69 expression in a subset of double negative DR-CD38- T-cells (NCT01276236)
Timeframe: Up to 96 weeks

Interventionpercentage change (Mean)
Treatment Arm (Maraviroc)50.5

[back to top]

Percent Change in CD69 Expression in a Subset of Double Negative DR-CD38 Positive (DR-CD38+) T-cells

Peripheral blood mononuclear cells (PBMC) were obtained from blood draws at each visit and T-cell immunophenotyping was performed from the baseline visit and final follow-up visit to determine the percent change in CD69 expression in a subset of double negative DR-CD38 positive (DR-CD38+) T-cells (NCT01276236)
Timeframe: Up to 96 weeks

Interventionpercentage change (Mean)
Treatment Arm (Maraviroc)89.7

[back to top]

Percent Change in KS Total Surface Area

Up to five bi-dimensionally measurable cutaneous KS lesions were selected as marker lesions and the the collective surface area of the marker lesions was evaluated over the course of the study. The percent decrease or increase in the total surface area of lesions was calculated from comparing measurements at baseline and through week 96, or at the last assessment if participant withdrew from the study prior to week 96. (NCT01276236)
Timeframe: Up to 96 weeks

Interventionpercentage change (Mean)
Treatment Arm (Maraviroc)-28.1

[back to top]

Mean Change From Baseline in the Hepatic Elastography (FibroscanTM) at Week 48, 96 and 144

Participants had transient hepatic elastography using FibroScan technology. It rapidly and non invasively measures hepatic tissue stiffness. Through a probe, a low frequency vibration of low amplitude is transmitted to the liver. The velocity of the wave that is generated during the procedure correlates directly with tissue stiffness as it passes through the liver; the harder or stiffer the liver, the faster the shear wave propagates. Results are reported in kilopascals (kPa). A negative change in the fibroscan values (i.e. decrease in liver stiffness) correlates with a decrease in fibrosis and thus improved outcome. (NCT01327547)
Timeframe: 48, 96 and 144 weeks

,
InterventionkPa (Mean)
Week 48 (n= 25, 28)Week 96 (n= 25, 28)Week 144 (n= 25, 28)
Maraviroc-1.3-0.8-1.7
Placebo0.40.4-0.3

[back to top]

Mean Change From Baseline in Plasma Hepatitis B Virus (HBV) DNA at Week 48, 96 and 144

Plasma samples were used to determine HBV DNA using the Roche COBAS Taqman HBV assay. Baseline value for HCV RNA/HBV DNA is defined as the pre-dose measurement taken at Day 1 visit. (NCT01327547)
Timeframe: 48, 96 and 144 weeks

,
InterventionLog10 values (Mean)
Week 48 (n= 15, 10)Week 96 (n= 15, 14)Week 144 (n= 15, 15)
Maraviroc-2.6-3.3-3.4
Placebo-3.0-3.0-3.0

[back to top]

Mean Change From Baseline in Markers of Immune Activation: Transforming Growth Factor-beta (TGF Beta) - Week 48, 96 and 144

Plasma samples were used to determine markers of immune activation namely TGF beta. (NCT01327547)
Timeframe: 48, 96 and 144 weeks

,
Interventionng/L (Mean)
Week 48 (n= 67, 66)Week 96 (n= 67, 66)Week 144 (n= 67, 66)
Maraviroc64.1-227.5792.0
Placebo-165.0-296.51275.4

[back to top]

Mean Change From Baseline in Markers of Immune Activation: D Dimer - Week 48, 96 and 144

Plasma samples were used to determine markers of immune activation namely D-Dimer. (NCT01327547)
Timeframe: 48, 96 and 144 weeks

,
Interventionng/dL (Mean)
Week 48 (n= 68, 65)Week 96 (n= 68, 65)Week 144 (n= 68, 65)
Maraviroc-101.1-88.1-97.4
Placebo-20.4-23.19.8

[back to top]

Mean Change From Baseline in Markers of Immune Activation: C-reactive Protein (CRP) - Week 48, 96 and 144.

Plasma samples were used to determine markers of immune activation namely CRP. (NCT01327547)
Timeframe: 48, 96 and 144 weeks

,
Interventionmg/dL (Mean)
Week 48 (n=70, 67)Week 96 (n=70, 67)Week 144 (n=70, 67)
Maraviroc0.40.00.6
Placebo3.1-0.7-0.3

[back to top]

Mean Change From Baseline in Log10 Plasma Hepatitis C Virus (HCV) RNA at Week 48, 96 and 144

Plasma samples were used to determine HCV RNA using the Roche COBAS Ampliprep/COBAS HCV Taqman assay, RUO version (LOD=15 IU/mL).Baseline value for HCV RNA/HBV DNA is defined as the pre-dose measurement taken at Day 1 visit. (NCT01327547)
Timeframe: 48, 96 and 144 weeks

,
InterventionLog10 values (Mean)
Week 48 (n= 45, 45)Week 96 (n= 45, 45)Week 144 (n= 45, 45)
Maraviroc-3.2-3.2-3.1
Placebo-3.2-3.4-3.3

[back to top]

Mean Change From Baseline in Enhanced Liver Fibrosis (ELF) Test at Week 48, 96 and 144

"The markers of fibrosis assessed in this test comprised hyaluronic acid (CHA), tissue inhibitor of metalloproteinase (CTIMP1) and procollagen III N-terminal peptide (CP3NP); these are components of the extracellular matrix and basement sinusoidal membrane of the liver and are elevated during activation of the stellate cell. The ELF tests were performed on an ADVIA Centaur XP and the composite score was calculated as follows: ELF score = 2.278 + 0.851 ln(CHA) + 0.751 ln (CP3NP) + 0.394 ln(CTIMP1).~ELF score < 7.7: no to mild fibrosis; ≥ 7.7 - < 9.8: Moderate fibrosis; ≥ 9.8 - < 11.3: Severe fibrosis; ≥ 11.3: Cirrhosis." (NCT01327547)
Timeframe: 48, 96 and 144 weeks

,
InterventionELF score (Mean)
Week 48 (n= 70, 67)Week 96 (n= 70, 67)Week 144 (n= 70, 67)
Maraviroc0.20.40.4
Placebo0.10.40.4

[back to top]

Mean Change From Baseline in CD38 Expression on CD4 and CD8 Cells at Weeks 48, 96 and 144

Plasma samples were used to determine markers of immune activation namely CD38 expression on CD4 and CD8 cells. (NCT01327547)
Timeframe: 48, 96 and 144 weeks

,
Interventioncell/mm³ (Mean)
Week 48 (n=69, 67)Week 96 (n=69, 67)Week 144(n=69, 67)
Maraviroc-12.25.423.4
Placebo43.047.450.7

[back to top]

Change From Baseline in Fibrosis Score (Ishak) in Liver Biopsy Samples at Week 144

Samples were processed and sent to a central reader for scoring for fibrosis and other analyses such as Sirius red and α smooth muscle actin staining for activated stellate cells. Samples were collected, processed, stored and shipped in accordance with the procedure documented in a separate handling document. The Ishak fibrosis scoring system was used to score the fibrosis observed, with a minimum score of 0 and maximum score of 6 (where 0 = no fibrosis, 1 = expansion of some portal areas with or without septa, 2 = expansion of most portal areas with or without septa, 3 = expansion of most portal areas with occasional portal or portal bridging, 4 = expansion of portal areas with marked bridging [portal-portal and/or portal-central], 5 = marked bridging with occasional nodules [incomplete cirrhosis], 6 = cirrhosis, probable or definitive). The scores for liver biopsies were summarized based upon the availability of liver biopsy results. (NCT01327547)
Timeframe: Week 144

,
InterventionNumerical score (Median)
Absolute Values at BaselineAbsolute Values at Post-dose (or Week 144)Change from baseline in fibrosis score at Week 144
Maraviroc320.0
Placebo000.0

[back to top]

Absolute Fibrosis Score (Ishak) in Liver Biopsy Samples at Baseline and at Week 144

Samples were processed and sent to a central reader for scoring for fibrosis and other analyses such as Sirius red and α smooth muscle actin staining for activated stellate cells. Samples were collected, processed, stored and shipped in accordance with the procedure documented in a separate handling document. The Ishak fibrosis scoring system was used to score the fibrosis observed, with a minimum score of 0 and maximum score of 6 (where 0 = no fibrosis, 1 = expansion of some portal areas with or without septa, 2 = expansion of most portal areas with or without septa, 3 = expansion of most portal areas with occasional portal or portal bridging, 4 = expansion of portal areas with marked bridging [portal-portal and/or portal-central], 5 = marked bridging with occasional nodules [incomplete cirrhosis], 6 = cirrhosis, probable or definitive). The scores for liver biopsies were summarized based upon the availability of liver biopsy results. (NCT01327547)
Timeframe: Baseline and Week 144

,
InterventionNumerical score (Mean)
Absolute Values at BaselineAbsolute Values at Post-dose (or Week 144)
Maraviroc2.62.2
Placebo1.51.5

[back to top]

Time to Development of Grade 3 and Grade 4 ALT Abnormalities at Week 144 Associated With a Change From Baseline ALT >100 IU/L

Time to development of Grade 3 and Grade 4 ALT abnormalities associated with a change from baseline ALT >100 IU/L during the 144-week period. Baseline will be defined as the last measurement prior to Day 1 dosing. (NCT01327547)
Timeframe: 144 weeks

InterventionDays (Median)
MaravirocNA
PlaceboNA

[back to top]

Mean Change From Baseline in CD4+ and CD8+ Cell Counts at Week 48, 96 and 144

Immunologic response (magnitude of change in CD4+ and CD8+ cell counts from baseline) was measured. Baseline value for CD4 and CD8 is defined as the pre-dose measurement taken at Day 1 visit. (NCT01327547)
Timeframe: Week 48, 96 and 144

,
InterventionCells/µL (Mean)
CD4+ (week 48, n=69, 67)CD8+ (week 48, n=69, 67)CD4+ (week 96, n=69, 67)CD8+ (week 96, n=69, 67)CD4+ (week 144, n=69, 67)CD8+ (week 144, n=69, 67)
Maraviroc3.17.85.1-2.717.212.6
Placebo42.028.949.762.641.744.1

[back to top]

Time to Development of Grade 3 and Grade 4 ALT Abnormalities

Time taken in days to development of Grade 3 and Grade 4 ALT abnormalities defined as >5x ULN for participants whose baseline ALT ≤ULN, or >3.5x baseline for participants whose baseline ALT >ULN, at Week 144. (NCT01327547)
Timeframe: 144 weeks

InterventionDays (Median)
MaravirocNA
PlaceboNA

[back to top]

Percentage of Participants With Grade 3 and Grade 4 Alanine Aminotransferase (ALT) Abnormalities at Week 48

Percentage of participants with Grade 3 or Grade 4 ALT abnormalities defined as >5x upper limit of normal (ULN) for participants whose baseline ALT ≤ULN, or >3.5x baseline for participants whose baseline ALT >ULN, up to and including Week 48 in the maraviroc arm versus the placebo arm. The baseline was defined as the last measurement prior to Day 1 dosing. (NCT01327547)
Timeframe: 48 weeks

InterventionPercentage of participants (Number)
Maraviroc1.4
Placebo1.5

[back to top]

Number of Participants With Hy's Law Abnormalities Through Week 144

Hy's law was defined as a total bilirubin >2x ULN with a simultaneous ALT or aspartate transaminase (AST)>3x ULN, excluding participants with an alkaline phosphatase>3x ULN (NCT01327547)
Timeframe: 144 weeks

Interventionparticipants (Number)
Maraviroc0
Placebo0

[back to top]

Exposure-response Relationship Between Change From Baseline in Liver Fibrosis Biomarkers Versus MVC Cavg at Week 48

The relationship between change from baseline in liver fibrosis biomarkers (AST, ALT, ALK, BIL, ELF and FSCN) versus MVC Cavg was analyzed using Bayesian methods. P-values were assessed for significance in the relationship between liver fibrosis biomarkers and MVC Cavg. P-value <0.05 was regarded as significantly related. (NCT01327547)
Timeframe: Week 48

Interventionp-value (Number)
Aspartate Transaminase (AST)0.892
Alanine Transaminase (ALT)0.440
Bilirubin (BIL)0.766
Enhanced Liver Fibrosis (ELF)0.795
Fibroscan (FSCN)0.087
Alkaline Phosphatase (ALK)0.071

[back to top]

Summary of Estimated Maraviroc PK Parameters

Week 4 and Week 48 clinic visits were scheduled such that a trough sample may be taken within a time window of 8-16 hours after the previous dose (Ctrough). Blood samples (4mL) were collected from all participants at the Week 4 and 48 visits. (NCT01327547)
Timeframe: Week 48

,,
Interventionng/mL (Median)
CavgCmaxCmin
Maraviroc 150 mg262496127.2
Maraviroc 300 mg16625861.3
Maraviroc 600 mg30991569.2

[back to top]

Percentage of Participants With Plasma Human Immunodeficiency Virus (HIV)-1 Ribonucleic Acid (RNA) Concentration <40 Copies/mL at Week 48, 96 and 144

"The Food and Drug Administration (FDA's) snapshot algorithm was used to derive the efficacy endpoint of the proportion of participants with HIV-1 RNA <40 copies/mL at Week 48. This algorithm included the missing data imputation method and used the plasma HIV-1 RNA concentration in the visit window only, followed the virology-first principle and considered a participant who had a missing plasma HIV-1 RNA concentration, or switched to a prohibited background anti-retroviral regimen or discontinues from the study or study drug as a failure (MSDF)." (NCT01327547)
Timeframe: Week 48, 96 and 144

,
InterventionPercentage of participants (Number)
Week 48Week 96Week 144
Maraviroc77.167.158.6
Placebo79.170.167.2

[back to top]

Percentage of Participants With Grade 3 and Grade 4 ALT Abnormalities Through Week 144

Percentage of participants with Grade 3 or Grade 4 ALT abnormalities defined as >5x upper limit of normal (ULN) for participants whose baseline ALT ≤ULN, or >3.5x baseline for participants whose baseline ALT >ULN, up to and including Week 96 and Week 144 in the maraviroc arm versus the placebo arm. The baseline was defined as the last measurement prior to Day 1 dosing. (NCT01327547)
Timeframe: Week 96 and 144

,
InterventionPercentage of participants (Number)
at Week 96at Week 144
Maraviroc1.42.9
Placebo3.04.5

[back to top]

Percentage of Participants With Grade 3 and Grade 4 ALT Abnormalities Associated With a Change From Baseline ALT >100 IU/L

Percentage of participants who had Grade 3 and Grade 4 ALT abnormalities associated with a change from baseline ALT >100 IU/L during the 144-week period. Baseline will be defined as the last measurement prior to Day 1 dosing. (NCT01327547)
Timeframe: 144 weeks

,
InterventionPercentage of participants (Number)
Grade 3Grade 4
Maraviroc2.81.4
Placebo4.40.0

[back to top]

Percentage of Participants Who Were Hospitalized Due to Hepatic Disease Through Week 144

Healthcare resource utilization data was collected using the Healthcare Resource Utilization Questionnaire at all study visits except Screening and Baseline. Other components of healthcare resource utilization, including length of hospital stay, type of ward, associated investigative and therapeutic procedures and concomitant medications were captured from primary and secondary data sources. (NCT01327547)
Timeframe: 144 Weeks

,
InterventionPercentage of participants (Number)
Not HospitalizedHospitalized due to Hepatic Disease at least onceHospitalized, but not due to Hepatic Disease
Maraviroc71.410.095.0
Placebo70.15.095.0

[back to top]

Number of Participants With Viral Resistance to Maraviroc (Maraviroc Treated Participants Only) in Participants Meeting PDTF Criteria.

For participants meeting the PDTF criteria, viral resistance to maraviroc for maraviroc treated participants was assessed in patients with R5 virus at failure. The resistance level is calculated by reference to a laboratory strain of virus that is analyzed in parallel with the clinical isolate to identify 50% inhibitory concentrations (IC50). The maximal percent inhibition is the percent inhibition that is achieved in a titration of the drug at high concentrations when the addition of more drug does not result in increased inhibition. Maximal percent inhibition is obtained in the same way as the titration for IC50, but the key measure is of the plateau height of percent inhibition, where increased concentration of maraviroc does not result in additional inhibition. This is consistent with the virus developing some ability to use maraviroc-bound CCR5 for entry. A significant change in IC50 is not required for this mechanism. (NCT01345630)
Timeframe: Week 48

,
Interventionparticipants (Number)
Not eligible for analysis (failed tropism test)Not eligible for analysis (non-R5 tropism)Eligible for analysis (R5 virus using ESTA)Results reportedMaximal percent inhibition <95%IC50 FC ≥3.0
FTC/TDF+DRV/r111100
MVC+DRV/r41121200

[back to top]

Percent Change From Baseline in Immune Cell Function at Week 48: Lymphocyte Activation Marker CD4 (%)

The differences in the magnitude of changes in CD4+ from Baseline through Week 48 for maraviroc versus emtricitabine/tenofovir were compared. (NCT01345630)
Timeframe: Baseline, Week 48

,
InterventionPercentage of lymphocytes (Mean)
Baseline (n=396, 401)Week 48 (n=394, 396)Change from Baseline at Week 48 (n=394, 396)
FTC/TDF+DRV/r24.533.79.2
MVC+DRV/r24.231.37.0

[back to top]

Percent Change From Baseline in Immune Cell Function at Week 48: Lymphocyte Activation Marker CD8 (%)

The differences in the magnitude of changes in CD8+ cell counts from Baseline through Week 48 for maraviroc versus emtricitabine/tenofovir were compared. (NCT01345630)
Timeframe: Baseline, Week 48

,
InterventionPercentage of lymphocytes (Mean)
Baseline (n=396, 401)Week 48 (n=394, 396)Change from Baseline at Week 48 (n=394, 396)
FTC/TDF+DRV/r55.843.0-12.6
MVC+DRV/r57.046.0-10.9

[back to top]

Severity of Abnormal Laboratory Values

Number of participants who had clinically significant laboratory abnormalities of Grade 3 and Grade 4 according to DAIDS. Abnormality incidence of highest grade was reported for a labcode for each individual participant. (NCT01345630)
Timeframe: Week 96

,
Interventionparticipants (Number)
Alanine Aminotransferase (ALT) (n=396, 400)Alkaline Phosphatase (n=396, 400)Amylase (n=396, 400)Aspartate Aminotransferase (AST) (n=396, 400)Blood Urea Nitrogen (BUN) (n=396, 400)Calcium (n=396, 400)Creatine Kinase (n=396, 400)Hemoglobin (n=396, 400)LDL Cholesterol (n=396, 400)Lipase (n=116, 122)Lymphocytes (Abs) (n=396, 400)Phosphate (n=396, 400)Platelets (n=396, 400)Potassium (n=396, 400)Sodium (n=396, 400)Total Bilirubin (n=396, 400)Total Neutrophils (Abs) (n=396, 400)Triglycerides (n=396, 400)Uric Acid (n=396, 400)White Blood Cell Count (n=396, 400)Creatinine (n=396, 400)
FTC/TDF+DRV/r601375102222410212120126201
MVC+DRV/r915113718450325532364010

[back to top]

Tropism Change Between Screening or Baseline and PDTF

For participants meeting the PDTF criteria, tropism was assessed using the original randomized and alternate assays (ie, both genotype testing and ESTA). Data reported here corresponds to the timepoint at or after PDTF. (NCT01345630)
Timeframe: Week 48

,,,
Interventionparticipants (Number)
R5 (Randomized Assay)NON R5 (Randomized Assay)NR (Randomized Assay)R5 (Alternate Assay)NON R5 (Alternate Assay)NR (Alternate Assay)
FTC/TDF+DRV/r - Baseline201300
FTC/TDF+DRV/r - Failure102210
MVC+DRV/r - Baseline14121025
MVC+DRV/r - Failure13131034

[back to top]

Virologic Outcomes at Week 48 Using Protocol-Defined Treatment Failure (PDTF).

Per the protocol participants who meet the following criteria were regarded as PDTFs requiring a confirmatory plasma HIV-1 RNA determination: • Decrease in plasma HIV-1 RNA <1 log10 from baseline after Week 4 unless plasma HIV-1 RNA is <50 copies/mL, or • Plasma HIV-1 RNA >1.0 log10 above the nadir value after Week 4 where the nadir is the lowest plasma HIV-1 RNA concentration, or • Plasma HIV-1 RNA ≥50 copies/mL at any time after Week 24, or • Plasma HIV-1 RNA ≥50 copies/mL after suppression to <50 copies/mL on two consecutive visits, or • Decrease in plasma HIV-1 RNA ≤2 log10 from baseline on or after Week 12 unless plasma HIV-1 RNA is <400 copies/mL. Decrease in plasma HIV-1 RNA ≤2 log10 from baseline on or after Week 12 unless plasma HIV-1 RNA is <50 copies/mL (before August 30 2012) or <400 copies/mL (after August 30 2012). (NCT01345630)
Timeframe: Week 48

,
InterventionNumber of participants (Number)
Confirmed PDTFEvaluable PDTF
FTC/TDF+DRV/r133
MVC+DRV/r4017

[back to top]

Change in Bone Turnover Markers From Baseline and at Week 48 - Blood Osteocalcin

Bone turnover marker, osteocalcin, was collected in the subset of participants participating in the DEXA scan sub-study. (NCT01345630)
Timeframe: Week 48

Interventionng/mL (Mean)
MVC+DRV/r5.61
FTC/TDF+DRV/r6.77

[back to top]

Change in Bone Turnover Markers From Baseline and at Week 48 - Type 1 Collagen Peptide (CTX-1)

Bone turnover marker, C-telopeptide of type 1 collagen (CTx), was collected in the subset of participants participating in the DEXA scan sub-study. (NCT01345630)
Timeframe: Week 48

Interventionpg/mL (Mean)
MVC+DRV/r121.13
FTC/TDF+DRV/r223.52

[back to top]

Changes in Bone Mineral Density (Using DEXA Scan and Serum Markers) From Baseline and at Week 48 - AP Lumbar Spine (L1 - L4) BMD

Bone mineral density was evaluated by DEXA scan in a subset of participants who consented to these evaluations. The effects on BMD were addressed by providing LSMs of change from baseline bone mineral density of the lumbar spine (L1-L4) as measured by the DEXA scan. (NCT01345630)
Timeframe: Week 48

Interventiong/cm^2 (Least Squares Mean)
MVC+DRV/r-0.020
FTC/TDF+DRV/r-0.025

[back to top]

Changes in Bone Mineral Density (Using DEXA Scan and Serum Markers) From Baseline and at Week 48 - Femoral Neck BMD

Bone mineral density was evaluated by DEXA scan in a subset of participants who consented to these evaluations. The effects on BMD were addressed by providing LSMs of change from Baseline bone mineral density femoral neck as measured by the DEXA scan. (NCT01345630)
Timeframe: Week 48

Interventiong/cm^2 (Least Squares Mean)
MVC+DRV/r-0.021
FTC/TDF+DRV/r-0.029

[back to top]

Changes in Bone Mineral Density (Using DEXA Scan and Serum Markers) From Baseline and at Week 48 - Total Hip BMD

Bone mineral density was evaluated by DEXA scan in a subset of participants who consented to these evaluations. The effects on BMD were addressed by providing LSMs of change from baseline bone mineral density of the lumbar spine (L1-L4), left total hip and femoral neck as measured by the DEXA scan. (NCT01345630)
Timeframe: Week 48

Interventiong/cm^2 (Least Squares Mean)
MVC+DRV/r-0.014
FTC/TDF+DRV/r-0.028

[back to top]

Changes in Peripheral Fat Distribution Using Dual Energy X-ray Absorptiometry [DEXA] Scan From Baseline and at Week 48.

A sub-study was conducted in which the participants underwent whole-body DEXA scans to evaluate peripheral fat tissue estimates for left and right arms and legs and truncal fat mass and truncal lean mass. Truncal abdominal fat were estimated from the DEXA scan field set on the torso. The effects on estimates of fat mass and lean mass were addressed by providing LSMs of change from baseline. (NCT01345630)
Timeframe: Week 48

Interventiongram (Least Squares Mean)
MVC+DRV/r-181.6
FTC/TDF+DRV/r-257.5

[back to top]

Changes in Trunk to Limb Fat Distribution Using DEXA Scan From Baseline and at Week 48

A sub-study was conducted in which the participants underwent whole-body DEXA scans to evaluate peripheral fat tissue estimates for left and right arms and legs and truncal fat mass and truncal lean mass. Truncal abdominal fat were estimated from the DEXA scan field set on the torso. The effects on estimates of fat mass and lean mass were addressed by providing LSMs of change from baseline. (NCT01345630)
Timeframe: Week 48

Interventionratio (Least Squares Mean)
MVC+DRV/r0.017
FTC/TDF+DRV/r-0.014

[back to top]

Frequency of Adverse Events (AE).

Number of participants with treatment-emergent non serious AEs (NCT01345630)
Timeframe: Week 96

Interventionparticipants (Number)
MVC+DRV/r360
FTC/TDF+DRV/r365

[back to top]

Number of Participants Who Discontinued Due to AEs

Number of participants who discontinued due to AEs are reported here. Three participants (two from the MVC+DRV/r arm and one from the FTC/TDF+DRV/r arm) were not considered as discontinued due to AE because other reasons for discontinuation were prioritized for these participants. (NCT01345630)
Timeframe: Week 96

Interventionparticipants (Number)
MVC+DRV/r22
FTC/TDF+DRV/r23

[back to top]

Number of Participants With Grade 3 or 4 AEs

Number of participants with grade 3 or 4 AEs are presented here. (NCT01345630)
Timeframe: Week 96

Interventionparticipants (Number)
MVC+DRV/r65
FTC/TDF+DRV/r71

[back to top]

Number of Participants With Treatment-emergent Serious Adverse Events

Total number of participants with treatment-emergent serious adverse events are reported (NCT01345630)
Timeframe: Week 96

Interventionparticipants (Number)
MVC+DRV/r41
FTC/TDF+DRV/r40

[back to top] [back to top]

Percentage of Participants With Plasma HIV-1 RNA <50 Copies/mL.

"The proportion of participants who achieved HIV-1 RNA <50 copies/mL at week 48 was assessed according to Food and Drug Administration's (FDA's) Missing, Switch, Discontinuation'=Failure (MSDF) Snapshot algorithm. The algorithm used the plasma HIV-1 RNA in the Week 48 visit window, followed the virology-first principle and considers a participant who has a missing plasma HIV-1 RNA, or switches to prohibited ARV regimen or discontinues from the study or study drug for any reason, or dies, as a failure." (NCT01345630)
Timeframe: Week 48

InterventionPercentage of participants (Number)
MVC+DRV/r77.3
FTC/TDF+DRV/r86.8

[back to top]

The Relationship Between the Proportion of Participants With Plasma HIV-1 RNA <50 Copies/mL at the Week 48 and the Screening Tropism Test (Genotype Test or ESTA).

The relationship of the proportion of participants achieving HIV-1 RNA <50 copies/mL at Week 48 with the screening tropism test for the MVC containing regimen was analyzed. Virologic response for a participant at Week 48 was derived using the FDA's Snapshot MSDF algorithm. Difference in proportions of patients with plasma HIV-1 RNA <50 copies/mL at week 48 between the maraviroc and the emtricitabine/tenofovir treatment arms, with two-sided 95% confidence interval, among patients who are R5 by genotype (including some who were originally randomized to ESTA and are R5 by genotype upon retesting), were calculated via the Maximum Likelihood method. The estimate was adjusted for the screening plasma HIV RNA level (<100,000 vs. ≥100,000) copies/mL via the Mantel Haenszel (MH) method. (NCT01345630)
Timeframe: Week 48

Interventionproportion of participants (Number)
MVC+DRV/r0.8047
FTC/TDF+DRV/r0.8797

[back to top]

Absolute Change From Baseline in Immune Cell Function at Week 48: Lymphocyte Marker Cluster of Differentiation 4 (CD4, Cell/mm^3)

The differences in the magnitude of changes in CD4+ at Baseline and at Week 48 for maraviroc versus emtricitabine/tenofovir were compared. (NCT01345630)
Timeframe: Baseline, Week 48

,
Interventioncell/mm^3 (Mean)
Baseline (n=396, 401)Week 48 (n=394, 396)Change from Baseline at Week 48 (n=394, 396)
FTC/TDF+DRV/r379.5574.6194.2
MVC+DRV/r382.0576.9194.9

[back to top]

Absolute Change From Baseline in Immune Cell Function at Week 48: Lymphocyte Marker Cluster of Differentiation 8 (CD8, Cell/mm^3)

The differences in the magnitude of changes in CD8+ cell counts from baseline through Week 48 for maraviroc versus emtricitabine/tenofovir were compared. (NCT01345630)
Timeframe: Baseline, Week 48

,
Interventioncell/mm^3 (Mean)
Baseline (n=396, 401)Week 48 (n=394, 396)Change from Baseline at Week 48 (n=394, 396)
FTC/TDF+DRV/r914.5751.1-157.9
MVC+DRV/r954.4900.0-49.9

[back to top]

Absolute Change in CD4+/CD8+ Ratio From Baseline to Week 48

The differences in the magnitude of changes in CD4+/CD8+ ratio from Baseline through Weeks 48 for maraviroc versus emtricitabine/tenofovir were compared. (NCT01345630)
Timeframe: Baseline, Week 48

,
InterventionRatio (Mean)
Baseline (n=396, 401)Week 48 (n=394, 396)Change from Baseline at Week 48 (n=394, 396)
FTC/TDF+DRV/r0.480.870.39
MVC+DRV/r0.470.750.28

[back to top]

Number of Participants With Abnormal Laboratory Values

Number of participants with laboratory abnormalities are reported (NCT01345630)
Timeframe: Week 96

,
Interventionparticipants (Number)
Normal BaselineAbnormal Baseline
FTC/TDF+DRV/r205101
MVC+DRV/r210111

[back to top]

Number of Participants With Resistance to Nucleoside/Nucleotide Reverse Transcriptase Inhibitors (NRTI), Non-nucleoside Reverse Transcriptase Inhibitors (NNRTI), and Protease Inhibitors (PI) in Participants Meeting PDTF Criteria

For participants meeting the PDTF criteria, viral resistance (both genotypic and phenotypic) to NRTI, NNRTI, and PI's were assessed at Baseline and on-treatment. The assessment was performed using the overall (i.e. net) susceptibility score provided using the PhenoSense GT assay. The number of participants with successful assessments were 15/17 for the MVC+DRV/r arm and 3/3 for the FTC/TDF+DRV/r arm. (NCT01345630)
Timeframe: Week 48

,
Interventionparticipants (Number)
NRTI - All (Baseline, n=15, 3)NNRTI Delavirdine (Baseline, n=15, 3)NNRTI Nevirapine (Baseline, n=15, 3)NNRTI Efavirenz (Baseline, n=15, 3)PRI - All (Baseline, n=15, 3)NRTI - All (PDTF, n=15, 3)NNRTI Delavirdine (PDTF, n=15, 3)NNRTI Nevirapine (PDTF, n=15, 3)NNRTI Efavirenz (PDTF, n=15, 3)PRI - All (PDTF, n=15, 3)
FTC/TDF+DRV/r0000000000
MVC+DRV/r0111001110

[back to top]

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

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

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

[back to top]

Number of Participants With Changes in Haematology and Biochemistry Laboratory Tests

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

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

[back to top]

Change in Level of IP-10 From Baseline to Week 48

Change in level of Interferon gamma-induced protein 10 (IP-10) from baseline to week 48 (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionpg/ml (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-198
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-170

[back to top]

Percent Change in Expression of CD57+ on CD8+ T Cells From Baseline to Week 48

percentage change is define as [ (week 48 - week 0) / week 0 ] * 100% (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionpercentage change (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-3.5
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-4.6

[back to top]

Percent Change in Expression of CD28+/CD57+ on CD8+ T Cells From Baseline to Week 48

percentage change is define as [ (week 48 - week 0) / week 0 ] * 100% (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionpercentage change (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-9.1
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-11.2

[back to top]

Percent Change in Expression of CD28+ on CD8+ T Cells From Baseline to Week 48

percentage change is define as [ (week 48 - week 0) / week 0 ] * 100% (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionpercentage change (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)11.9
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)14.0

[back to top]

Change in CD4 Count From Baseline to Week 24

Change in CD4 count from baseline (week 0) to week 24 (NCT01400412)
Timeframe: Week 0, week 24

Interventioncells/mm^3 (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)165
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)127

[back to top]

Change in CD4 Count From Baseline to Week 48

Change in CD4 count from baseline (week 0) to week 48 (NCT01400412)
Timeframe: Week 0, week 48

Interventioncells/mm^3 (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)234
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)188

[back to top]

Percent Change in Lumbar Spine Bone Mineral Density (BMD)

The percent change in bone mineral density (BMD) at lumbar spine (as measured by DXA scan) from baseline (week 0) to week 48. (NCT01400412)
Timeframe: Week 0, week 48

Interventionpercentage change (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-0.88
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-2.35

[back to top]

CD8+ T-cell Change From Baseline to Week 48

CD8+ T-cell change from baseline to week 48 (NCT01400412)
Timeframe: At weeks 0 and 48

Interventioncell/mm^3 (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-6
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-109

[back to top]

Percent Change in Expression of RANKL+ on CD8+ T Cells From Baseline to Week 48

percentage change is defined as [ (week 48 - week 0) / week 0 ] * 100% (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionpercentage change (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-20.7
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-17.0

[back to top]

Change in Levels of D-dimer From Baseline

Change in levels of D-dimer from baseline (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionng/ml (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-82
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-61

[back to top]

Percentage Change in Expression of CD38+/HLA-DR+ on CD4+ T Cells From Baseline to Week 48

percentage change is define as [ (week 48 - week 0) / week 0 ] * 100% (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionpercentage change (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-52.1
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-48.6

[back to top]

Change in Levels of IL-6 From Baseline to Week 48

Change in levels of Interleukin 6 (IL-6) from baseline to week 48 (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionpg/ml (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-0.21
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-0.12

[back to top]

Change in Levels of sCD14 From Baseline

Change in levels of soluble CD14 from baseline (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionng/ml (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-103
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-10

[back to top]

Change in Levels of sCD163 From Baseline to Week 48

Change in levels of soluble CD163 from baseline to week 48 (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionng/ml (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-250
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-258

[back to top]

Number of Participants Who Developed Grade 3 or 4 Primary Adverse Events

"Grade 3 or 4 primary adverse events includes primary signs/symptoms, primary laboratory abnormalities, or primary diagnoses.~See DAIDS AE Grading Table Version 1.0, Dec 2004 (Clarification, Aug 2009)" (NCT01400412)
Timeframe: From study treatment initiation to week 48

Interventionparticipants (Number)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)16
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)22

[back to top]

Number of Participants Who Died During the Study

Number of participants who died during the study (NCT01400412)
Timeframe: From study treatment initiation to week 48

Interventionparticipants (Number)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)0
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)0

[back to top]

Cumulative Probability of Virologic Failure by Week 48

"Confirmed virologic failure is defined as confirmed plasma HIV-1 RNA levels > 1000 copies/mL at or after week 16 and before week 24, or confirmed HIV-1 RNA levels> 200 copies/mL at or after week 24. Participants who discontinued the study with an unconfirmed virologic failure (HIV-1 RNA > 1000 copies at 16 weeks or HIV-1 RNA level > 200 copies/mL at or after week 24) are considered as virologic failures at the study visit week of the unconfirmed value. Time to virologic failure is defined as the time from study entry to the planned visit week of the initial failure.~Product-limit estimates for the survival function were used to estimate the cumulative probability of virologic failure over time and its corresponding 95% confidence interval for each treatment group." (NCT01400412)
Timeframe: From study treatment initiation to week 48

Interventioncumulative probability per 100 persons (Number)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)6
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)5

[back to top]

Number of Participants Who Experienced Bone Fractures

Number of participants who experienced bone fractures during the study (NCT01400412)
Timeframe: From study treatment initiation to week 48

Interventionparticipants (Number)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)2
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)2

[back to top]

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

The primary endpoint is the percent change in bone mineral density (BMD) at total hip (as measured by DXA scan) from baseline (week 0) to week 48. (NCT01400412)
Timeframe: Week 0, week 48

Interventionpercentage change (Median)
MVC Arm (DRV/r + MVC + FTC + TDF Placebo)-1.51
TDF Arm (DRV/r + TDF + FTC + MVC Placebo)-2.40

[back to top]

Percentage Change in Expression of CD38+/HLA-DR+ on CD8+ T Cells From Baseline to Week 48

percentage change is defined as [ (week 48 - week 0) / week 0 ] * 100% (NCT01400412)
Timeframe: At weeks 0 and 48

Interventionpercentage change (Median)
MVC Arm: DRV/r + MVC + FTC + TDF Placebo-59.5
TDF Arm: DRV/r + TDF + FTC + MVC Placebo-60.9

[back to top]

Change in MRS Cerebral Metabolite Ratios in Frontal White Matter

Change in major cerebral metabolites in the frontal white matter, as measured by 1H-Magnetic Resonance Spectroscopy (MRS), between baseline and 12-months. Spectra were acquired on a Phillips Achieva 3T MRI scanner using point-resolved spectroscopy (PRESS) sequence with short TE. jMRUI/AMARES algorithm was used to process spectra. Metabolite ratios were calculated for the following metabolites: N-acetyl aspartate (NAA), choline (Cho), creatine (Cr), myo-inositol (mIo), glutamate/glutamine complex (Glx), in relation to internal H2O as standard. (NCT01449006)
Timeframe: Baseline and 12 months

,
Interventionratio (Least Squares Mean)
NAA BaselineNAA 12 MonthsCr BaselineCr 12 MonthsCho BaselineCho 12 MonthsmIo BaselinemIo 12 MonthsGlx BaselineGlx 12 Months
Maraviroc4.164.162.933.052.132.241.071.352.272.18
Standard of Care HAART Regimen4.324.302.742.992.192.191.111.152.162.05

[back to top]

Change in MRS Cerebral Metabolite Ratios in Basal Ganglia

Change in major cerebral metabolites in the basal ganglia, as measured by 1H-Magnetic Resonance Spectroscopy (MRS), between baseline and 12-months. Spectra were acquired on a Phillips Achieva 3T MRI scanner using point-resolved spectroscopy (PRESS) sequence with short echot time (TE). jMRUI/AMARES algorithm was used to process spectra. Metabolite ratios were calculated for the following metabolites: N-acetyl aspartate (NAA), choline (Cho), creatine (Cr), myo-inositol (mIo), in relation to internal water (H20) as standard. (NCT01449006)
Timeframe: Baseline and 12 months

,
Interventionratio (Least Squares Mean)
NAA BaselineNAA 12 MonthsCr BaselineCr 12 MonthsCho BaselineCho 12 MonthsmIo BaselinemIo 12 Months
Maraviroc3.963.892.913.081.451.551.191.07
Standard of Care HAART Regimen3.763.873.093.241.421.591.100.88

[back to top]

Change in CSF Neopterin Concentration

Change in concentration of the CSF neuroinflammatory marker neopterin (measured in nmol/L) from baseline to 12-months. (NCT01449006)
Timeframe: Baseline and 12-months

,
Interventionnmol/L (Least Squares Mean)
Baseline12-months
Maraviroc12.5715.71
Standard of Care HAART Regimen11.513.25

[back to top]

Change in Neurocognitive Functioning

Change in overall neurocognitive performance, defined as a global neurocognitive z-score, over the study time-period (baseline, 6-months, 12-months). To derive this score, 1) raw scores obtained from a 5-domain brief neurocognitive battery were converted to age-corrected z-scores (M=0, SD=1) and 2) the set of individual subtest z-scores were averaged to generate a single composite (global) z-score for each subject. Lower (negative) scores therefore indicate greater levels of cognitive impairment. (NCT01449006)
Timeframe: Baseline, 6-months and 12-months

,
InterventionGlobal Neurocognitive Z-Score (Least Squares Mean)
Baseline6 months12 months
Maraviroc-0.81-0.51-0.56
Standard of Care HAART Regimen-0.94-1.03-0.93

[back to top]

Occurrence of Grade 3 or Higher Adverse Events (AEs)

participants had Occurrence of Grade 3 or higher adverse events (AEs) (NCT01505114)
Timeframe: Through Week 48

InterventionParticipants (Count of Participants)
Arm 118
Arm 224
Arm 320
Arm 428

[back to top]

Day +180 Rate of Grade II-IV Acute GVHD

The cumulative incidence of grade II-IV acute GVHD by day 180 after the stem-cell infusion. This is based on consensus conference criteria. (NCT01785810)
Timeframe: 180 days

InterventionParticipants (Count of Participants)
Maraviroc22

[back to top]

Safety/Tolerability of the Treatments

description and frequency of adverse events for all participants during the study. (NCT01894776)
Timeframe: 30 days

InterventionParticipants (Count of Participants)
Maraviroc72220849Maraviroc + Rifabutin72220849
Number of Participants with Adverse EventsNumber of Participants without Adverse Events
Maraviroc6
Maraviroc8
Maraviroc7

[back to top]

Pharmacokinetics of Maraviroc and Rifabutin AUC 0-12/24

Maraviroc pharmacokinetics: Maraviroc only AUC (h*μg/L), Maraviroc + Rifabutin AUC (h*μg/L), Rifabutin AUC (h*μg/L), 25-O-desacetyl rifabutin AUC (h*μg/L). (NCT01894776)
Timeframe: Maraviroc: 0, 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 10 and 12 hour. Rifabutin: 0, 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 10, 12 and 24 hour..

Interventionh*μg/L (Geometric Mean)
Maraviroc only AUC (h*μg/L)Maraviroc + Rifabutin AUC (h*μg/L)Rifabutin AUC (h*μg/L)25-O-desacetyl rifabutin AUC (h*μg/L)
Maraviroc1026.2847.04221.9331.9

[back to top]

Maraviroc and Rifabutin C12/C24/Cmax PK Concentrations in Plasma.

Maraviroc + Rifabutin pharmacokinetics: Maraviroc only Cmax (μg/L), Maraviroc only C12 (μg/L), Maraviroc + Rifabutin Cmax (μg/L), Maraviroc + Rifabutin C12 (μg/L), Rifabutin Cmax (μg/L), Rifabutin C24 (μg/L), 25-O-desacetyl rifabutin Cmax (μg/L), 25-O-desacetyl rifabutin C24 (μg/L). (NCT01894776)
Timeframe: Maraviroc: 0, 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 10 and 12 hour. Rifabutin: 0, 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 10, 12 and 24 hour..

Interventionμg/L (Geometric Mean)
Maraviroc only Cmax (μg/L)Maraviroc only C12 (μg/L)Maraviroc + Rifabutin Cmax (μg/L)Maraviroc + Rifabutin C12 (μg/L)Rifabutin Cmax (μg/L)Rifabutin C24 (μg/L)25-O-desacetyl rifabutin Cmax (μg/L)25-O-desacetyl rifabutin C24 (μg/L)
Maraviroc304.623.3239.816.3542.271.242.35.5

[back to top]

Number of Participants With Adverse Events

Number of participants with adverse events (NCT01896921)
Timeframe: 96 weeks

InterventionParticipants (Count of Participants)
Maraviroc + Raltegravir or Dolutegravir3

[back to top]

Number of Patients Virologically Suppressed (HIV RNA <50 Copies/ml) at 48 Weeks.

Number of patients virologically suppressed (HIV RNA <50 copies/ml) at 48 weeks. (NCT01896921)
Timeframe: 48 weeks

InterventionParticipants (Count of Participants)
Maraviroc + Raltegravir or Dolutegravir5

[back to top]

Number of Patients Who Are Virologically Suppressed (HIV RNA < 50 Copies/ml)

Number of patients who are virologically suppressed (HIV RNA < 50 copies/ml) (NCT01896921)
Timeframe: 96 weeks

InterventionParticipants (Count of Participants)
Maraviroc + Raltegravir or Dolutegravir4

[back to top]

Toxicities

Incidence of toxicities due to drug (NCT02167451)
Timeframe: Up to day +100

InterventionParticipants (Count of Participants)
Maraviroc0

[back to top]

Area Under The Concentration-Time Curve (AUC) of Maraviroc

pK target >100ng/ml at day 10 at the following time points : before the dose and 1, 2, 4, 6, 8, and 12 hours after maraviroc administration (NCT02167451)
Timeframe: Day 10

Interventionhour*ng/mL (Mean)
Maraviroc5917

[back to top]

Infectious Complications

Infections complications which include asymptomatic viremias for EBV, Adenovirus, CMV, and/or viral disease, bacterial and fungal infections as documented by blood cultures. (NCT02167451)
Timeframe: Up to day +100

Interventionpatients (Number)
bacterial infectionsFungal infectionsCMVEBVAdenovirusBk viremia
Maraviroc1125425

[back to top]

Area Under The Concentration-Time Curve (AUC) of Maraviroc

pK target >100ng/ml at day zero at the following time points : before the dose and 1, 2, 4, 6, 8, and 12 hours after maraviroc administration (NCT02167451)
Timeframe: Day 0

Interventionhour *ng/mL (Mean)
Maraviroc4805

[back to top]

Feasibility of Maraviroc

The ability to administer twice daily oral maraviroc in children and adults undergoing stem cell transplant in addition to routine standard graft versus host disease prophylaxis. (NCT02167451)
Timeframe: Up to day +100

InterventionParticipants (Count of Participants)
Maraviroc23

[back to top]

Graft Failure

Failure to engraft and loss of graft. (NCT02167451)
Timeframe: By day +100

InterventionParticipants (Count of Participants)
Maraviroc4

[back to top]

GVHD Incidence

Incidence of GVHD by day+100 (NCT02167451)
Timeframe: By day +100

InterventionParticipants (Count of Participants)
Maraviroc11

[back to top]

Incidence of Visceral GVHD

determine the number of patients who develop visceral GVHD by day+100 (NCT02167451)
Timeframe: day+100

InterventionParticipants (Count of Participants)
Maraviroc6

[back to top]

Overall Survival

Overall survival for patients who were enrolled and received maraviroc (NCT02167451)
Timeframe: By day +100

InterventionParticipants (Count of Participants)
Maraviroc27

[back to top]

Primary Disease Relapse

(NCT02167451)
Timeframe: By day +100

InterventionParticipants (Count of Participants)
Maraviroc0

[back to top]

Time to Neutrophil

Neutrophil engraftment is defined as the first of three consecutive measurements of ANC>500mcL over 3 or more days. (NCT02167451)
Timeframe: Up to day +100

Interventiondays (Median)
Maraviroc11

[back to top]

Time to Platelet Engraftment

Time to achieve platelets count of 20,000 without transfusions (NCT02167451)
Timeframe: days

Interventiondays (Median)
Maraviroc18

[back to top]

Percentage of Participants With Neutrophil Recovery

Neutrophil recovery is defined as achieving an absolute neutrophil count (ANC) ≥ 500/mm^3 for three consecutive measurements on three different days. (NCT02208037)
Timeframe: Days 28 and 100 Post-transplant

,,
Interventionpercentage of participants (Number)
Day 28Day 100
Tacrolimus/Methotrexate/Bortezomib9496
Tacrolimus/Methotrexate/Maraviroc9395
Tacrolimus/MMF/Cyclophosphamide9598

[back to top]

Percentage of Participants With Chronic GVHD

Chronic GVHD is classified per 2005 NIH Consensus Criteria (Filipovich et al. 2005) into categories of severity: none, mild, moderate, and severe. Occurrence of chronic GVHD is defined as the occurrence of mild, moderate, or severe chronic GVHD per this classification. (NCT02208037)
Timeframe: 1 Year Post-transplant

Interventionpercentage of participants (Number)
Tacrolimus/Methotrexate/Bortezomib39
Tacrolimus/Methotrexate/Maraviroc43
Tacrolimus/MMF/Cyclophosphamide28

[back to top]

Percentage of Participants With Chronic GVHD Requiring Immunosupressive Therapy

Chronic GVHD is classified per 2005 NIH Consensus Criteria (Filipovich et al. 2005) into categories of severity: none, mild, moderate, and severe. Occurrence of chronic GVHD is defined as the occurrence of mild, moderate, or severe chronic GVHD per this classification. This endpoint considers the occurrence of chronic GVHD that necessitated initiation of immunosuppressive therapy for treatment. (NCT02208037)
Timeframe: 1 Year Post-transplant

Interventionpercentage of participants (Number)
Tacrolimus/Methotrexate/Bortezomib29
Tacrolimus/Methotrexate/Maraviroc33
Tacrolimus/MMF/Cyclophosphamide22

[back to top]

Percentage of Participants With Disease Relapse or Progression

Relapse is defined by either morphological or cytogenetic evidence of acute leukemia or MDS consistent with pretransplant features, or radiologic evidence of lymphoma. Progression of disease applies to patients with lymphoproliferative diseases (lymphoma or chronic lymphocytic leukemia) not in remission prior to transplantation and is defined as increase in size of prior sites of disease or evidence of new sites of disease. (NCT02208037)
Timeframe: 1 Year Post-transplant

Interventionpercentage of participants (Number)
Tacrolimus/Methotrexate/Bortezomib24
Tacrolimus/Methotrexate/Maraviroc31
Tacrolimus/MMF/Cyclophosphamide28

[back to top]

Percentage of Participants With Grade III-IV Acute GVHD

"Acute GVHD is graded according to the scoring system proposed by Przepiorka et al.1995:~Skin stage:~0: No rash~Rash <25% of body surface area~Rash on 25-50% of body surface area~Rash on > 50% of body surface area~Generalized erythroderma with bullous formation~Liver stage (based on bilirubin level)*:~0: <2 mg/dL 1.2-3 mg/dL 2.3.01-6 mg/dL 3.6.01-15.0 mg/dL 4.>15 mg/dL~GI stage*:~0: No diarrhea or diarrhea <500 mL/day~Diarrhea 500-999 mL/day or persistent nausea with histologic evidence of GVHD~Diarrhea 1000-1499 mL/day~Diarrhea >1500 mL/day~Severe abdominal pain with or without ileus * If multiple etiologies are listed for liver or GI, the organ system is downstaged by 1.~GVHD grade:~0: All organ stages 0 or GVHD not listed as an etiology I: Skin stage 1-2 and liver and GI stage 0 II: Skin stage 3 or liver or GI stage 1 III: Liver stage 2-3 or GI stage 2-4 IV: Skin or liver stage 4" (NCT02208037)
Timeframe: Day 180 Post-transplant

Interventionpercentage of participants (Number)
Tacrolimus/Methotrexate/Bortezomib8
Tacrolimus/Methotrexate/Maraviroc9
Tacrolimus/MMF/Cyclophosphamide2

[back to top]

Percentage of Participants With GVHD-free Survival

GVHD-free survival is defined as being alive without previous onset of Grade III-IV acute GVHD or chronic GVHD requiring immunosuppressive therapy. (NCT02208037)
Timeframe: 1 Year Post-transplant

Interventionpercentage of participants (Number)
Tacrolimus/Methotrexate/Bortezomib43
Tacrolimus/Methotrexate/Maraviroc34
Tacrolimus/MMF/Cyclophosphamide53

[back to top]

Percentage of Participants With Grade II-IV Acute GVHD

"Acute GVHD is graded according to the scoring system proposed by Przepiorka et al.1995:~Skin stage:~0: No rash~Rash <25% of body surface area~Rash on 25-50% of body surface area~Rash on > 50% of body surface area~Generalized erythroderma with bullous formation~Liver stage (based on bilirubin level)*:~0: <2 mg/dL~2-3 mg/dL~3.01-6 mg/dL~6.01-15.0 mg/dL~>15 mg/dL~GI stage*:~0: No diarrhea or diarrhea <500 mL/day~Diarrhea 500-999 mL/day or persistent nausea with histologic evidence of GVHD~Diarrhea 1000-1499 mL/day~Diarrhea >1500 mL/day~Severe abdominal pain with or without ileus * If multiple etiologies are listed for liver or GI, the organ system is downstaged by 1.~GVHD grade:~0: All organ stages 0 or GVHD not listed as an etiology I: Skin stage 1-2 and liver and GI stage 0 II: Skin stage 3 or liver or GI stage 1 III: Liver stage 2-3 or GI stage 2-4 IV: Skin or liver stage 4" (NCT02208037)
Timeframe: Day 180 Post-transplant

Interventionpercentage of participants (Number)
Tacrolimus/Methotrexate/Bortezomib26
Tacrolimus/Methotrexate/Maraviroc32
Tacrolimus/MMF/Cyclophosphamide27

[back to top]

Percentage of Participants With GVHD/Relapse or Progression-free Survival (GRFS)

GRFS is defined as being free of grade III-IV acute GVHD onset, chronic GVHD onset requiring systemic immunosuppressive therapy, disease relapse or progression, and death from any cause. (NCT02208037)
Timeframe: 1 Year Post-transplant

Interventionpercentage of participants (Number)
Tacrolimus/Methotrexate/Bortezomib35.5
Tacrolimus/Methotrexate/Maraviroc27.2
Tacrolimus/MMF/Cyclophosphamide44.1

[back to top]

Percentage of Participants With Overall Survival

(NCT02208037)
Timeframe: 1 Year Post-transplant

Interventionpercentage of participants (Number)
Tacrolimus/Methotrexate/Bortezomib68
Tacrolimus/Methotrexate/Maraviroc66
Tacrolimus/MMF/Cyclophosphamide71

[back to top] [back to top]

Percentage of Participants With Platelet Recovery

Platelet recovery is defined as the first day of a sustained platelet count >20,000/mm^3 with no platelet transfusion in the preceding seven days. (NCT02208037)
Timeframe: Days 60 and 100 Post-transplant

,,
Interventionpercentage of participants (Number)
Day 60Day 100
Tacrolimus/Methotrexate/Bortezomib9191
Tacrolimus/Methotrexate/Maraviroc9292
Tacrolimus/MMF/Cyclophosphamide9096

[back to top]

Donor Cell Engraftment

Donor cell engraftment will be assessed with donor/recipient chimerism. Chimerism may be evaluated in bone marrow, whole blood, or CD3 fractions. Full donor chimerism is defined as the presence of ≥ 95% of donor cells as a proportion of total cells. Mixed chimerism is defined as the presence of donor cells, as a proportion of total cells, of < 95% but > 5% in the bone marrow or peripheral blood. Full and mixed chimerism will be evidence of donor cell engraftment. Donor cells of ≤ 5% will be considered as graft rejection. (NCT02208037)
Timeframe: Days 28 and 100 Post-transplant

InterventionParticipants (Count of Participants)
Day 2872130179Day 2872130181Day 2872130180Day 10072130180Day 10072130181Day 10072130179
Full ChimerismMixed ChimerismGraft RejectionDeadNo Assay Performed
Tacrolimus/Methotrexate/Bortezomib58
Tacrolimus/MMF/Cyclophosphamide64
Tacrolimus/Methotrexate/Bortezomib11
Tacrolimus/MMF/Cyclophosphamide8
Tacrolimus/Methotrexate/Bortezomib1
Tacrolimus/Methotrexate/Maraviroc1
Tacrolimus/MMF/Cyclophosphamide2
Tacrolimus/Methotrexate/Bortezomib2
Tacrolimus/Methotrexate/Maraviroc4
Tacrolimus/MMF/Cyclophosphamide0
Tacrolimus/Methotrexate/Bortezomib17
Tacrolimus/Methotrexate/Maraviroc21
Tacrolimus/MMF/Cyclophosphamide18
Tacrolimus/Methotrexate/Bortezomib63
Tacrolimus/Methotrexate/Maraviroc56
Tacrolimus/MMF/Cyclophosphamide62
Tacrolimus/Methotrexate/Bortezomib14
Tacrolimus/Methotrexate/Maraviroc17
Tacrolimus/MMF/Cyclophosphamide13
Tacrolimus/Methotrexate/Bortezomib5
Tacrolimus/Methotrexate/Maraviroc3
Tacrolimus/MMF/Cyclophosphamide3
Tacrolimus/Methotrexate/Bortezomib4
Tacrolimus/Methotrexate/Maraviroc10
Tacrolimus/MMF/Cyclophosphamide5
Tacrolimus/Methotrexate/Bortezomib3
Tacrolimus/Methotrexate/Maraviroc6
Tacrolimus/MMF/Cyclophosphamide9

[back to top]

Percentage of Participants With Disease-free Survival

Disease-free survival is defined as being alive and free of disease relapse or progression. (NCT02208037)
Timeframe: 1 Year Post-transplant

Interventionpercentage of participants (Number)
Tacrolimus/Methotrexate/Bortezomib58
Tacrolimus/Methotrexate/Maraviroc56
Tacrolimus/MMF/Cyclophosphamide60

[back to top]

Count of Total Cells Obtained From Cervicovaginal Lavage Samples

The count of CCR5+CD4+ T cells in the genital tract, before participants began study treatment, after 7 days of treatment, and during the post-treatment drug elimination period. The precise role of CCR5+CD4+ T cells in the female genital tract is unknown, however, higher cell counts may suggest the potential for more HIV target cells in the genital tract. (NCT02333045)
Timeframe: Baseline, Day 7, Day 14, Day 21

,
Interventioncells (Mean)
BaselineDay 7Day 14Day 21
Maraviroc20,000,00018,700,00028,300,00021,800,000
Truvada42,000,00015,000,00020,000,00030,000,000

[back to top] [back to top]

CD8+ T-cell Counts

CD8+ T-cell counts were recorded at the given time point (NCT02519777)
Timeframe: Measured at Weeks 24, 48, and 96

,,
Interventioncells/mm^3 (Mean)
CD8 Count (Week 24)CD8 Count (Week 48)CD8 Count (Week 96)
Arm A: Placebo MVC and Placebo DTG757742747
Arm B: DTG and Placebo MVC752731773
Arm C: MVC and DTG862856879

[back to top]

Change in CD4+ T-cell Count

Changes in CD4+ T-cell count were calculated as the CD4+ T-cell count at a given time point minus the CD4+ T-cell count at Baseline. (NCT02519777)
Timeframe: Measured at Baseline and Weeks 24, 48, and 96

,,
Interventioncells/mm^3 (Mean)
Change from Baseline in CD4 Count (Week 24)Change from Baseline in CD4 Count (Week 48)Change from Baseline in CD4 Count (Week 96)
Arm A: Placebo MVC and Placebo DTG-13-43-10
Arm B: DTG and Placebo MVC-33-1910
Arm C: MVC and DTG422144

[back to top]

Change in CD8+ T-cell Count

Changes in CD8+ T-cell count were calculated as the CD8+ T-cell count at a given time point minus the CD8+ T-cell count at baseline. (NCT02519777)
Timeframe: Measured at Baseline and Weeks 24, 48, and 96

,,
Interventioncells/mm^3 (Mean)
Change from Baseline in CD8 Count (Week 24)Change from Baseline in CD8 Count (Week 48)Change from Baseline in CD8 Count (Week 96)
Arm A: Placebo MVC and Placebo DTG-29-45-43
Arm B: DTG and Placebo MVC-61-82-33
Arm C: MVC and DTG443544

[back to top]

Change in Functional Status Scores

Functional status scores were calculated based on the instrumental activities of daily living (IADLs) forms. IADL scores were calculated as the sum of the eight IADL tasks where the task scores were equal to 1 if a participant did not need assistance with the task and equal to 0 if a participant needed some level of help with the task. The maximum possible functional status score was 8, while the minimum possible functional status score was 0, with higher functional status scores indicating a higher level of functionality. (NCT02519777)
Timeframe: Measured at Baseline and Weeks 24, 48, 72, and 96

,,
Interventionunits on a scale (Mean)
Change from Baseline in IADL Score (Week 24)Change from Baseline in IADL Score (Week 48)Change from Baseline in IADL Score (Week 72)Change from Baseline in IADL Score (Week 96)
Arm A: Placebo MVC and Placebo DTG0.290.380.400.16
Arm B: DTG and Placebo MVC0.430.450.300.28
Arm C: MVC and DTG0.150.100.130.20

[back to top]

Change in Normalized Composite Neurocognitive Test Score at Weeks 24, 72, and 96 From Baseline

"The normalized composite neurocognitive test score (total z-score) was defined as the average of the z-scores from the following tests:~Domestic and International Participants:~Grooved pegboard dominant~Grooved pegboard non-dominant~HVLT-R Learning trials~HVLT-R Delayed recall~HVLT-R Delayed recognition~Semantic verbal fluency~Domestic only:~Stroop color naming~Stroop word reading~Stroop interference trial~Letter fluency~Trail Making A~Trail Making B~WAIS-III Symbol search~Digit Symbol~International only:~Timed Gait~Finger Tapping Dominant~Finger Tapping Non-dominant~Color Trail 1~Color Trail 2~Z-scores were calculated using a demographically appropriate norming process. Higher z-scores correspond with better neurocognitive performance. Change was calculated as the total z-score at the given time point minus the total z-score at Baseline." (NCT02519777)
Timeframe: Measured at Baseline and Weeks 24, 72, and 96

,,
Interventiontotal neurocognitive z-score (Mean)
Change of Normalized Composite Neurocognitive Test Score (Week 24)Change of Normalized Composite Neurocognitive Test Score (Week 72)Change of Normalized Composite Neurocognitive Test Score (Week 96)
Arm A: Placebo MVC and Placebo DTG0.140.290.37
Arm B: DTG and Placebo MVC0.180.320.34
Arm C: MVC and DTG0.200.370.38

[back to top]

Number of Participants With Plasma HIV-1 RNA Greater Than or Equal to 50 Copies/mL

The number of participants with Plasma HIV-1 RNA greater than or equal to 50 copies/mL was assessed at each given time point. (NCT02519777)
Timeframe: Measured at Weeks 24, 48, and 96

,,
InterventionParticipants (Count of Participants)
HIV RNA (Week 24)HIV RNA (Week 48)HIV RNA (Week 96)
Arm A: Placebo MVC and Placebo DTG331
Arm B: DTG and Placebo MVC106
Arm C: MVC and DTG112

[back to top]

CD4+ T-cell Counts

CD4+ T-cell counts were recorded at the given time point (NCT02519777)
Timeframe: Measured at Weeks 24, 48, and 96

,,
Interventioncells/mm^3 (Mean)
CD4 Count (Week 24)CD4 Count (Week 48)CD4 Count (Week 96)
Arm A: Placebo MVC and Placebo DTG660638674
Arm B: DTG and Placebo MVC669691720
Arm C: MVC and DTG773758788

[back to top]

Change in Log10 IP-10 in CSF at Week 48 From Baseline

Changes in Log10 IP-10 in CSF were calculated as the Log10 IP-10 in CSF at Week 48 minus the Log10 IP-10 in CSF at Baseline. (NCT02519777)
Timeframe: Measured at Baseline and Week 48

InterventionLog10 pg/mL (Mean)
Arm A: Placebo MVC and Placebo DTG0.02
Arm B: DTG and Placebo MVC-0.11
Arm C: MVC and DTG-0.36

[back to top]

Change in Log10 MIP-1 Beta in Cerebrospinal Fluid (CSF) at Week 48 From Baseline

Changes in Log10 MIP-1 Beta in CSF were calculated as the Log10 MIP-1 Beta in CSF at Week 48 minus the Log10 MIP-1 Beta in CSF at Baseline. (NCT02519777)
Timeframe: Measured at Baseline and Week 48

InterventionLog10 pg/mL (Mean)
Arm A: Placebo MVC and Placebo DTG-0.24
Arm B: DTG and Placebo MVC-0.32
Arm C: MVC and DTG0.17

[back to top]

Change in Log10 MIP-1 Beta in Plasma at Week 48 From Baseline

Changes in Log10 MIP-1 Beta in Plasma were calculated as the Log10 MIP-1 Beta in Plasma at Week 48 minus the Log10 MIP-1 Beta in Plasma at Baseline. Results below the lower limit of quantification were set to the lower limit value of 11. (NCT02519777)
Timeframe: Measured at Baseline and Week 48

InterventionLog10 pg/mL (Mean)
Arm A: Placebo MVC and Placebo DTG0.05
Arm B: DTG and Placebo MVC-0.02
Arm C: MVC and DTG0.30

[back to top]

Change in Log10 Neopterin in CSF at Week 48 From Baseline

Changes in Log10 Neopterin in CSF were calculated as the Log10 Neopterin in CSF at Week 48 minus the Log10 Neopterin in CSF at Baseline. (NCT02519777)
Timeframe: Measured at Baseline and Week 48

InterventionLog10 nmol/L (Mean)
Arm A: Placebo MVC and Placebo DTG0.01
Arm B: DTG and Placebo MVC-0.06
Arm C: MVC and DTG-0.17

[back to top]

Change in Log10 NFL in CSF at Week 48 From Baseline

Changes in Log10 NFL in CSF were calculated as the Log10 NFL in CSF at Week 48 minus the Log10 NFL in CSF at Baseline. (NCT02519777)
Timeframe: Measured at Baseline and Week 48

InterventionLog10 pg/mL (Mean)
Arm A: Placebo MVC and Placebo DTG-0.02
Arm B: DTG and Placebo MVC-0.05
Arm C: MVC and DTG0.00

[back to top]

Change in Log10 sCD14 in Plasma at Week 48 From Baseline

Changes in Log10 sCD14 in Plasma were calculated as the Log10 sCD14 in Plasma at Week 48 minus the Log10 sCD14 in Plasma at Baseline. (NCT02519777)
Timeframe: Measured at Baseline and Week 48

InterventionLog10 ng/mL (Mean)
Arm A: Placebo MVC and Placebo DTG0.04
Arm B: DTG and Placebo MVC0.03
Arm C: MVC and DTG0.01

[back to top]

Change in Log10 sTNFr-II in Plasma at Week 48 From Baseline

Changes in Log10 sTNFr-II in Plasma were calculated as the Log10 sTNFr-II in Plasma at Week 48 minus the Log10 sTNFr-II in Plasma at Baseline. (NCT02519777)
Timeframe: Measured at Baseline and Week 48

InterventionLog10 pg/mL (Mean)
Arm A: Placebo MVC and Placebo DTG0.02
Arm B: DTG and Placebo MVC0.01
Arm C: MVC and DTG0.00

[back to top]

Change in Log10 VCAM in Plasma at Week 48 From Baseline

Changes in Log10 VCAM in Plasma were calculated as the Log10 VCAM in Plasma at Week 48 minus the Log10 VCAM in Plasma at Baseline. (NCT02519777)
Timeframe: Measured at Baseline and Week 48

InterventionLog10 pg/mL (Mean)
Arm A: Placebo MVC and Placebo DTG0.01
Arm B: DTG and Placebo MVC0.00
Arm C: MVC and DTG0.00

[back to top]

Change in Normalized Composite Neurocognitive Test Score at Week 48 From Baseline

"The normalized composite neurocognitive test score (total z-score) was defined as the average of the z-scores from the following tests:~Domestic (US-based) and International Participants:~Grooved pegboard dominant~Grooved pegboard non-dominant~Hopkins Verbal Learning Test (HVLT-R) Learning trials~HVLT-R Delayed recall~HVLT-R Delayed recognition~Semantic verbal fluency~Domestic only:~Stroop color naming~Stroop word reading~Stroop interference trial~Letter fluency~Trail Making A~Trail Making B~WAIS-III Symbol search~Digit Symbol~International only:~Timed Gait~Finger Tapping Dominant~Finger Tapping Non-dominant~Color Trail 1~Z-scores were calculated using a demographically appropriate norming process. Higher z-scores correspond with better neurocognitive performance. Change was calculated as the total z-score at Week 48 minus the total z-score at Baseline." (NCT02519777)
Timeframe: Measured at Baseline and Week 48

Interventiontotal neurocognitive z-score (Mean)
Arm A: Placebo MVC and Placebo DTG0.20
Arm B: DTG and Placebo MVC0.26
Arm C: MVC and DTG0.31

[back to top]

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

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

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

[back to top]

Part 2: Number of Participants With Laboratory Abnormalities

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

Part 2: Average Plasma Concentration (Cavg) of Maraviroc

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

Part 1: Average Plasma Concentration (Cavg) of Maraviroc

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

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

[back to top]

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

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

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

[back to top]

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

An AE was any untoward medical occurrence in a participant who received study drug without regard to possibility of causal relationship. SAE was an AE resulting in any of the following outcomes or deemed significant for any other reason: death; initial or prolonged inpatient hospitalization; life-threatening experience (immediate risk of dying); persistent or significant disability/incapacity; congenital anomaly. Treatment-emergent are events between first dose of study drug and up to 11 days that were absent before treatment or that worsened relative to pretreatment state. (NCT02625207)
Timeframe: Baseline up to end of study (up to 11 days)

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

[back to top]

Part 1: Number of Participants With Laboratory Abnormalities

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

An AE was any untoward medical occurrence in a participant who received study drug without regard to possibility of causal relationship. SAE was an AE resulting in any of the following outcomes or deemed significant for any other reason: death; initial or prolonged inpatient hospitalization; life-threatening experience (immediate risk of dying); persistent or significant disability/incapacity; congenital anomaly. Treatment-emergent are events between first dose of study drug and up to 6 days that were absent before treatment or that worsened relative to pre-treatment state. (NCT02625207)
Timeframe: Baseline up to end of study (up to 6 days)

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

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

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

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

[back to top]

Tissue Common Rejection Module (tCRM) Score Using the 11-gene tCRM Module on FFPE Biopsy Shaves

Tissue Common Rejection Module (tCRM) score using the 11-gene tCRM module on FFPE biopsy shaves at 26 weeks. The score measures the average (geometric mean) gene expression level of CRM genes (BASP1, CD6, CXCL10, CXCL9, INPP5D, ISG20, LCK, NKG7, PSMB9, RUNX3, TAP1) in kidney tissue. Possible range (min-max) for the tCRM score is 0.01 - 15.0, with higher values representing worse outcomes. (NCT02741323)
Timeframe: Measured at Week 26 Post-transplant

Interventionscore on a scale (Median)
Arm 1: Maraviroc (MVC)1.83
Arm 2: Placebo1.67

[back to top]

Trough Levels of CCR5 Blockade (Maraviroc)

Trough levels of CCR5 blockade (maraviroc) from 0-12 hours post-dose testing at month 3 post-transplant in a subset of participants enrolled at UCSF (NCT02741323)
Timeframe: Month 3 Post-transplant (0-12 hours post-dose)

InterventionnM (Median)
Arm 1: Maraviroc (MVC)90.95

[back to top]

Urine Common Rejection Module (uCRM) Score Using the 11-gene uCRM Module on Urine Cell Pellets

Urine Common Rejection Module (uCRM) score using the 11-gene uCRM module on urine cell pellets at week 26. The score measures the average (geometric mean) gene expression level of CRM genes (BASP1, CD6, CXCL10, CXCL9, INPP5D, ISG20, LCK, NKG7, PSMB9, RUNX3, TAP1) in urine sediment. Possible range (min-max) for the uCRM score is 0.01 - 15.0, with higher values representing worse outcomes. (NCT02741323)
Timeframe: Measured at Week 26 Post-transplant

Interventionscore on a scale (Median)
Arm 1: Maraviroc (MVC)0.48
Arm 2: Placebo0.42

[back to top]

Urine Common Rejection Module (uCRM) Score Using the 11-gene uCRM Module on Urine Cell Pellets

Urine Common Rejection Module (uCRM) score using the 11-gene uCRM module on urine cell pellets at week 52. The score measures the average (geometric mean) gene expression level of CRM genes (BASP1, CD6, CXCL10, CXCL9, INPP5D, ISG20, LCK, NKG7, PSMB9, RUNX3, TAP1) in urine sediment. Possible range (min-max) for the uCRM score is 0.01 - 15.0, with higher values representing worse outcomes. (NCT02741323)
Timeframe: Measured at Week 52 Post-transplant

Interventionscore on a scale (Median)
Arm 1: Maraviroc (MVC)0.40
Arm 2: Placebo0.25

[back to top]

AUC of CCR5 Blockade (Maraviroc)

AUC of CCR5 blockade (maraviroc), 0-12 hours post-dose at month 3 post-transplant in a subset of participants enrolled at UCSF (NCT02741323)
Timeframe: Month 3 Post-transplant (0-12 hours post-dose)

Interventionnmol-h/L (Median)
Arm 1: Maraviroc (MVC)3101.82

[back to top]

Incidence/Proportion of Participants With HIV Infection in the Renal Allograft

Histology/in situ hybridization was used to assess HIV infection in the renal allograft and calculate the proportion of participants with HIV infection (NCT02741323)
Timeframe: Month 6 Post-transplant

InterventionProportion of participants (Number)
Arm 1: Maraviroc (MVC)0
Arm 2: Placebo0

[back to top]

Calcineurin Inhibitor (Tacrolimus) AUC for Participants on Maraviroc Versus Placebo

Calcineurin inhibitor (tacrolimus) AUC, 0-12 hours post-dose at month 3 post-transplant for a subset of participants enrolled at UCSF (NCT02741323)
Timeframe: Month 3 Post-transplant (0-12 hours post-dose)

Interventionnmol-h/L (Median)
Arm 1: Maraviroc (MVC)191.17
Arm 2: Placebo193.22

[back to top]

Calcineurin Inhibitor (Tacrolimus) Trough Levels for Participants on Maraviroc Versus Placebo

Calcineurin inhibitor (tacrolimus) trough levels, from 0-12 hours post-dose at month 3 post-transplant for a subset of participants enrolled at UCSF (NCT02741323)
Timeframe: Month 3 Post-transplant (0-12 hours post-dose)

InterventionnM (Median)
Arm 1: Maraviroc (MVC)9.70
Arm 2: Placebo12.10

[back to top]

Cumulative Incidence of Graft Loss, Toxicities ≥ Grade 3 Per the DAIDS Toxicity Table and/or Permanent Treatment Discontinuation

The primary safety endpoint will be the incidence of graft loss and toxicities ≥ Grade 3 and/or permanent treatment discontinuation within the first 52 weeks post-transplant (NCT02741323)
Timeframe: Measured through Week 52 Post-transplant

InterventionProportion (KM estimate for incidence) (Number)
Arm 1: Maraviroc (MVC)0.84
Arm 2: Placebo0.85

[back to top]

Cumulative Incidence/Proportion of Acute Cellular Rejection Grade Equal to or Greater Than 1A During the Entire Study Follow-up

Measured by the Banff 2007 criteria as identified on central read of biopsy slides; for site biopsy results, grading was not available, all results except for borderline results were assumed to be grade 1A or greater. If available, central read result was used; if not, site biopsy result was used. (NCT02741323)
Timeframe: Within 3 years post-transplant

InterventionProportion of participants (Number)
Arm 1: Maraviroc (MVC)0.1224
Arm 2: Placebo0.1429

[back to top]

Cumulative Incidence/Proportion of Biopsy Proven Acute Rejection Within 1 Year Post Transplant

Defined by histologic evidence of rejection and graft dysfunction as identified on central read of biopsy slides as well as on site biopsies. When a central read of biopsy slide is available, those results will be used; in its absence, site biopsy result will be used. Both acute cellular and humoral rejections were considered for this outcome measure, but borderline results were excluded. (NCT02741323)
Timeframe: Within Year 1 Post-transplant

InterventionProportion of participants (Number)
Arm 1: Maraviroc (MVC)0.0816
Arm 2: Placebo0.1250

[back to top]

HIV DNA in Peripheral Blood CD4+ T Cells at Week 52

HIV DNA in peripheral blood CD4+ T cells at Week 52. The standard ACTG type extraction protocol was used to extract DNA from PBMC. The readout was copies of cellular HIV-1 DNA per million PBMC. Subsequently, the outcome measure/value was obtained by multiplying the readout by the participant's CD4 percentage level at that time point, to obtain the measure of copies of HIV-1 DNA per million peripheral blood CD4+ T cells (NCT02741323)
Timeframe: At week 52 post-transplant

Interventioncopies of HIV-1 DNA/million CD4+ T cells (Median)
Arm 1: Maraviroc (MVC)22.2
Arm 2: Placebo43.9

[back to top]

HIV RNA in Peripheral Blood CD4+ T Cells at Week 52.

HIV RNA in peripheral blood CD4+ T cells at Week 52. The standard ACTG type extraction protocol was used to extract RNA from PBMC. The readout was copies of cellular HIV-1 RNA per million PBMC. Subsequently, the outcome measure/value was obtained by multiplying the readout by the participant's CD4 percentage level at that time point, to obtain the measure of copies of HIV-1 RNA per million peripheral blood CD4+ T cells (NCT02741323)
Timeframe: Week 52 Post Transplant

Interventioncopies of HIV-1 RNA/million CD4+ T cells (Median)
Arm 1: Maraviroc (MVC)402.1
Arm 2: Placebo902.6

[back to top]

Incidence of All Adverse Events (AEs) Greater Than or Equal to Grade 3 at Year 1

Incidence of all adverse events (AEs) greater than or equal to Grade 3 within 1 year post-transplant (NCT02741323)
Timeframe: Within Year 1 Post-transplant

InterventionPercent (KM estimate for incidence) (Number)
Arm 1: Maraviroc (MVC)81.6
Arm 2: Placebo77.6

[back to top]

Incidence of Death at Year 1

Incidence of death within Year 1 post-transplant (NCT02741323)
Timeframe: Within Year 1 Post-transplant

InterventionPercent (KM estimate for incidence) (Number)
Arm 1: Maraviroc (MVC)4.3
Arm 2: Placebo2.2

[back to top]

Incidence of Graft Loss in Year 1

Incidence of graft loss within Year 1 post-transplant. (NCT02741323)
Timeframe: Within Year 1 Post-transplant

InterventionPercent (KM estimate for incidence) (Number)
Arm 1: Maraviroc (MVC)6.3
Arm 2: Placebo6.6

[back to top]

Incidence of Non-opportunistic Infections Requiring Hospitalization Within Year 1

Incidence of non-opportunistic infections requiring hospitalization within 1 year post-transplant (NCT02741323)
Timeframe: Within Year 1 Post-transplant

InterventionPercent (KM estimate for incidence) (Number)
Arm 1: Maraviroc (MVC)21.3
Arm 2: Placebo28.6

[back to top]

Incidence of Opportunistic Infections or Neoplasms Within Year 1

Incidence of opportunistic infections or neoplasms within 1 year post-transplant (NCT02741323)
Timeframe: Within Year 1 Post-transplant

InterventionPercent (KM estimate for incidence) (Number)
Arm 1: Maraviroc (MVC)19.1
Arm 2: Placebo6.7

[back to top]

Incidence of Serious Adverse Events (SAEs) Greater Than or Equal to Grade 3 Within Year 1

Incidence of serious adverse events (SAEs) greater than or equal to Grade 3 within 1 year post-transplant (NCT02741323)
Timeframe: Within Year 1 Post-transplant

InterventionPercent (KM estimate for incidence) (Number)
Arm 1: Maraviroc (MVC)73.5
Arm 2: Placebo53.2

[back to top]

Incidence/Proportion of Antibody Mediated Rejection

Incidence of humoral/antibody mediated rejection within 52 weeks of the transplant. Both central reads and site biopsy results were included, and central read result was used if one was available, and if not, the site biopsy result was used. (NCT02741323)
Timeframe: Within 52 weeks post transplant

InterventionProportion of participants (Number)
Arm 1: Maraviroc (MVC)0.0000
Arm 2: Placebo0.0208

[back to top]

Mean CD45 Gene Expression Count (PTPRC)

"Based on the formalin-fixed paraffin-embedded (FFPE) kidney biopsy sample. CD45 RNA In Situ Hybridization was performed, and the CD45 gene expression count is calculated by counting the spots (the RNA signal) in QuPath and then dividing the number of spots by biopsy tissue area in mm²." (NCT02741323)
Timeframe: Measured at Week 26 Post-transplant

Interventionspots/mm² (Median)
Arm 1: Maraviroc (MVC)49.4
Arm 2: Placebo50.9

[back to top]

Mean CD45 Quantitative Immunohistochemistry (IHC)

Mean CD45 quantitative immunohistochemistry (IHC) based on FFPE sample (NCT02741323)
Timeframe: Measured at Week 26 Post-transplant

Interventioncells/mm² (Median)
Arm 1: Maraviroc (MVC)144.7
Arm 2: Placebo77.5

[back to top]

Mean eGFR at Week 52 Based on CKD-EPI Creatinine Equation

Mean eGFR at Week 52 calculated by CKD-EPI creatinine equation (NCT02741323)
Timeframe: Measured at Week 52 Post-transplant

InterventionmL/min/1.73 m² (Mean)
Arm 1: Maraviroc (MVC)59.2
Arm 2: Placebo49.3

[back to top]

Mean Glomerular Filtration Rate by Iohexol Clearance at Week 52

The primary efficacy endpoint is the 52-week GFR as measured by iohexol clearance (NCT02741323)
Timeframe: Measured at Week 52 Post-transplant

InterventionmL/min/1.73 m² (Mean)
Arm 1: Maraviroc (MVC)37.0
Arm 2: Placebo40.1

[back to top]

Plasma HIV RNA Levels (Single Copy Assay) at Week 52

Plasma HIV RNA levels (single copy assay) at Week 52 Post-transplant (NCT02741323)
Timeframe: Week 52 Post-transplant

Interventioncopies of HIV-1 RNA/mL (Median)
Arm 1: Maraviroc (MVC)0
Arm 2: Placebo0.3

[back to top]

Proportion of Participants With de Novo Anti-donor Human Leukocyte Antigen (HLA) Antibodies

Proportion of participants with de novo anti-donor human leukocyte antigen (HLA) antibodies at Week 52 (NCT02741323)
Timeframe: Measured at Week 52

InterventionProportion of participants (Number)
Arm 1: Maraviroc (MVC)0.1600
Arm 2: Placebo0.0870

[back to top]

Proportion of Participants With Defined CKD Stage 4 or 5 at Year 1

Proportion of participants with defined CKD stage 4 or 5 at week 52 post-transplant. CKD Stage 4 or 5 is defined as a glomerular filtration rate (GFR) of <30 mL/min. (NCT02741323)
Timeframe: Year 1 time point

InterventionProportion of participants (Number)
Arm 1: Maraviroc (MVC)0.07
Arm 2: Placebo0.14

[back to top]

Proportion of Participants With Estimated Glomerular Filtration Rate (eGFR) Less Than 60 mL/Min/1.73 m² at Week 52

Measured by Chronic Kidney Disease Epidemiology collaboration equation (CKD-EPI) Creatinine equation (NCT02741323)
Timeframe: Measured at Week 52 Post-transplant

InterventionProportion (Number)
Arm 1: Maraviroc (MVC)0.45
Arm 2: Placebo0.73

[back to top]

The Slope of eGFR Over Time in Year 1

The slope of eGFR over time in Year 1, calculated by CKD-EPI Creatinine equation. Slope is computed via the repeated measures analysis, covering the study time points of weeks 13, 26, 39 and 52. The estimated average slope (and corresponding 95% confidence interval) is provided for incremental progression from one time point to the next (i.e., the displayed slope shows the extent of increase (positive) or decrease (negative) in eGFR level per every time point (13 weeks) elapsed. (NCT02741323)
Timeframe: Time points in Year 1 (four time points: weeks 13, 26, 39, 52)

InterventionmL/min/1.73 m² per 13 weeks (Mean)
Arm 1: Maraviroc (MVC)-0.1
Arm 2: Placebo-0.5

[back to top]

Number of Participants Failing to Meet PK Target

"Number of failures. The pharmacokinetic (PK) target is Average Concentration (Cavg) greater than or equal to 75 ng/mL (based on a dose interval of every 12 hours). Failure is defined as Cavg <75 ng/mL at each intensive PK visit.~For Cohort 1:~For the Entry Visit, PK samples were drawn at: pre-dose and at 1-2, 4-8, 11-13, 20-24, and 48-72 hours post-dose. For the Week 1 Visit, PK samples were drawn at: pre-dose and 1-2 and 22-26 hours post-dose.~For Cohort 2:~For both intensive PK visits, PK samples were drawn at: pre-dose and 1-2, 3-5, 6-8, and 11-13 hours post-dose." (NCT02778204)
Timeframe: Cohort 1: Measured at Entry (First visit) and Week 1 Visit (Second visit). Cohort 2: Measured at Week 1 (First visit) and Week 4 Visit (Second visit).

,
Interventionparticipants (Number)
First VisitSecond Visit
Cohort 2 Stratum 2A34
Cohort 2 Stratum 2B45

[back to top]

Pharmacokinetic (PK) Parameter: Area-under-the-curve (AUC)

"Pharmacokinetic parameters were determined from plasma concentration-time profiles using non-compartmental methods (Phoenix 64, WinNonlin, Pharsight Corp., Mountain View, CA). For Cohort 1 (single doses), area-under-the-curve (AUC) was determined from time zero to infinity. For Cohort 2 (at steady-state), area-under-the-curve (AUC) was determined from time pre-dose to tau (12 hours).~For Cohort 1:~For the Entry Visit, PK samples were drawn at: pre-dose and at 1-2, 4-8, 11-13, 20-24, and 48-72 hours post-dose. For the Week 1 Visit, PK samples were drawn at: pre-dose and 1-2 and 22-26 hours post-dose.~For Cohort 2:~For both intensive PK visits, PK samples were drawn at: pre-dose and 1-2, 3-5, 6-8, and 11-13 hours post-dose." (NCT02778204)
Timeframe: Cohort 1: Measured at Entry (First visit) and Week 1 Visit (Second visit). Cohort 2: Measured at Week 1 (First visit) and Week 4 Visit (Second visit).

,
Interventionng*hr/mL (Median)
First Visit
Cohort 1 Stratum 1A2285.68
Cohort 1 Stratum 1B4506.17

[back to top] [back to top]

Pharmacokinetic (PK) Parameter: Area-under-the-curve (AUC)

"Pharmacokinetic parameters were determined from plasma concentration-time profiles using non-compartmental methods (Phoenix 64, WinNonlin, Pharsight Corp., Mountain View, CA). For Cohort 1 (single doses), area-under-the-curve (AUC) was determined from time zero to infinity. For Cohort 2 (at steady-state), area-under-the-curve (AUC) was determined from time pre-dose to tau (12 hours).~For Cohort 1:~For the Entry Visit, PK samples were drawn at: pre-dose and at 1-2, 4-8, 11-13, 20-24, and 48-72 hours post-dose. For the Week 1 Visit, PK samples were drawn at: pre-dose and 1-2 and 22-26 hours post-dose.~For Cohort 2:~For both intensive PK visits, PK samples were drawn at: pre-dose and 1-2, 3-5, 6-8, and 11-13 hours post-dose." (NCT02778204)
Timeframe: Cohort 1: Measured at Entry (First visit) and Week 1 Visit (Second visit). Cohort 2: Measured at Week 1 (First visit) and Week 4 Visit (Second visit).

,
Interventionng*hr/mL (Median)
First VisitSecond Visit
Cohort 2 Stratum 2A1826.871122.99
Cohort 2 Stratum 2B1496.051216.62

[back to top]

Pharmacokinetic (PK) Parameter: Average Concentration (Cavg)

"Pharmacokinetic parameters were determined from plasma concentration-time profiles using non-compartmental methods (Phoenix 64, WinNonlin, Pharsight Corp., Mountain View, CA). Cavg was determined as the area-under-the-curve (AUC) divided by the dose interval, tau (τ) of every 12 hours.~For Cohort 1:~For the Entry Visit, PK samples were drawn at: pre-dose and at 1-2, 4-8, 11-13, 20-24, and 48-72 hours post-dose. For the Week 1 Visit, PK samples were drawn at: pre-dose and 1-2 and 22-26 hours post-dose.~For Cohort 2:~For both intensive PK visits, PK samples were drawn at: pre-dose and 1-2, 3-5, 6-8, and 11-13 hours post-dose." (NCT02778204)
Timeframe: Cohort 1: Measured at Entry and Week 1 Visit. Cohort 2: Measured at Week 1 and Week 4 Visit

,
Interventionng/mL (Median)
First Visit
Cohort 1 Stratum 1A190.47
Cohort 1 Stratum 1B375.47

[back to top]

Pharmacokinetic (PK) Parameter: Average Concentration (Cavg)

"Pharmacokinetic parameters were determined from plasma concentration-time profiles using non-compartmental methods (Phoenix 64, WinNonlin, Pharsight Corp., Mountain View, CA). Cavg was determined as the area-under-the-curve (AUC) divided by the dose interval, tau (τ) of every 12 hours.~For Cohort 1:~For the Entry Visit, PK samples were drawn at: pre-dose and at 1-2, 4-8, 11-13, 20-24, and 48-72 hours post-dose. For the Week 1 Visit, PK samples were drawn at: pre-dose and 1-2 and 22-26 hours post-dose.~For Cohort 2:~For both intensive PK visits, PK samples were drawn at: pre-dose and 1-2, 3-5, 6-8, and 11-13 hours post-dose." (NCT02778204)
Timeframe: Cohort 1: Measured at Entry and Week 1 Visit. Cohort 2: Measured at Week 1 and Week 4 Visit

,
Interventionng/mL (Median)
First VisitSecond Visit
Cohort 2 Stratum 2A152.2493.58
Cohort 2 Stratum 2B124.67101.39

[back to top]

Number of Participants Failing to Meet PK Target

"Number of failures. The pharmacokinetic (PK) target is Average Concentration (Cavg) greater than or equal to 75 ng/mL (based on a dose interval of every 12 hours). Failure is defined as Cavg <75 ng/mL at each intensive PK visit.~For Cohort 1:~For the Entry Visit, PK samples were drawn at: pre-dose and at 1-2, 4-8, 11-13, 20-24, and 48-72 hours post-dose. For the Week 1 Visit, PK samples were drawn at: pre-dose and 1-2 and 22-26 hours post-dose.~For Cohort 2:~For both intensive PK visits, PK samples were drawn at: pre-dose and 1-2, 3-5, 6-8, and 11-13 hours post-dose." (NCT02778204)
Timeframe: Cohort 1: Measured at Entry (First visit) and Week 1 Visit (Second visit). Cohort 2: Measured at Week 1 (First visit) and Week 4 Visit (Second visit).

,
Interventionparticipants (Number)
First Visit
Cohort 1 Stratum 1A0
Cohort 1 Stratum 1B0

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

Pharmacokinetic (PK) Parameter: Maximum Concentration (Cmax)

"Pharmacokinetic parameters were determined from plasma concentration-time profiles. Cmax was the observed highest concentration.~For Cohort 1:~For the Entry Visit, PK samples were drawn at: pre-dose and at 1-2, 4-8, 11-13, 20-24, and 48-72 hours post-dose. For the Week 1 Visit, PK samples were drawn at: pre-dose and 1-2 and 22-26 hours post-dose.~For Cohort 2:~For both intensive PK visits, PK samples were drawn at: pre-dose and 1-2, 3-5, 6-8, and 11-13 hours post-dose." (NCT02778204)
Timeframe: Cohort 1: Measured at Entry (First visit) and Week 1 Visit (Second visit). Cohort 2: Measured at Week 1 (First visit) and Week 4 Visit (Second visit).

,,,
Interventionng/mL (Median)
First VisitSecond Visit
Cohort 1 Stratum 1A227.3128.9
Cohort 1 Stratum 1B550.5163.4
Cohort 2 Stratum 2A256.9416.5
Cohort 2 Stratum 2B308.8221.8

[back to top]

Pharmacokinetic (PK) Parameter: Time of Maximum Concentration (Tmax)

"Pharmacokinetic parameters were determined from plasma concentration-time profiles. Tmax was the time at which Cmax, the observed highest concentration, occurred.~For Cohort 1:~For the Entry Visit, PK samples were drawn at: pre-dose and at 1-2, 4-8, 11-13, 20-24, and 48-72 hours post-dose. For the Week 1 Visit, PK samples were drawn at: pre-dose and 1-2 and 22-26 hours post-dose.~For Cohort 2:~For both intensive PK visits, PK samples were drawn at: pre-dose and 1-2, 3-5, 6-8, and 11-13 hours post-dose." (NCT02778204)
Timeframe: Cohort 1: Measured at Entry (First visit) and Week 1 Visit (Second visit). Cohort 2: Measured at Week 1 (First visit) and Week 4 Visit (Second visit).

,,,
Interventionhours (Median)
First VisitSecond Visit
Cohort 1 Stratum 1A4.681.18
Cohort 1 Stratum 1B1.521.08
Cohort 2 Stratum 2A1.501.50
Cohort 2 Stratum 2B3.002.19

[back to top]

Pharmacokinetic (PK) Parameter: Trough Concentration (Ctau)

"Pharmacokinetic parameters were determined from plasma concentration-time profiles. Ctau was the observed concentration at the trough time of 12 hours post-dose with steady-state dosing.~For Cohort 2:~For both intensive PK visits, PK samples were drawn at: pre-dose and 1-2, 3-5, 6-8, and 11-13 hours post-dose." (NCT02778204)
Timeframe: Measured at Week 1 and Week 4 Visit

,
Interventionng/mL (Median)
Week 1 VisitWeek 4 Visit
Cohort 2 Stratum 2A27.934.4
Cohort 2 Stratum 2B23.454.9

[back to top] [back to top]

Hair Antiretroviral Imaging

Signal Strength concentrations will be measured in hair for all study drugs, inclusive of FTC (emtricitabine), tenofovir (TFV), maraviroc (MRV), and dolutegravir (DTG) using Matrix-assisted Laser Desorption Electrospray Ionization (IR-MALDESI) to be reported by signal abundance (au). Signal abundance is the industry standard unit, and higher values represent greater signal with capability to relate to comparative concentrations. (NCT03218592)
Timeframe: Up to 28 days post dose

,,
InterventionSignal Abundance (au) (Median)
Phase 3: Zero Doses Per WeekPhase 3: 1 Dose Per weekPhase 3: 3 Doses Per WeekPhase 2: Daily DosingPhase 1: Single Dose
Dolutegravir1092.1308682377.8682265504.88316914251.46653NA
Maraviroc156.48266563942.69239627814.9882346013.29104NA
Truvada0.501209156.1127125466.7986895824.721385NA

[back to top]

Plasma Antiretroviral Concentrations

Concentrations will be measured in hair of all study drugs, inclusive of FTC (emtricitabine), TFV (tenofovir), MRV (Maraviroc), and DTG (dolutegravir) (NCT03218592)
Timeframe: Up to 28 days post-dose

,,,
Interventionng/mL (Median)
Phase 1: Single dosePhase2: 7 Doses per WeekPhase 3: 3 Doses Per WeekPhase 3: 1 Dose Per WeekPhase 3: 0 Doses per week
Dolutegravir1124069.33.0951
Emtricitabine0.572.317.21.20.5
Maraviroc19.7151.7111
Tenofovir0.561.99.40.50.5

[back to top]

Whole Blood Antiretroviral Concentrations

Concentrations will be measured in dried blood spots of all study drugs, inclusive of FTC (emtricitabine), TFV (tenofovir), MRV (Maraviroc), and DTG (dolutegravir). Truvada is a combination pill, so results for both FTC and TFV are reported in separate columns. (NCT03218592)
Timeframe: Up to 28 days post-dose

,,,
Interventionfmol / 3mm punch (Median)
Phase 1: Single DosePhase 2: 7 Doses per WeekPhase 3: 3 Doses Per WeekPhase 3: 1 Dose Per WeekPhase 3: 0 Doses per week
Dolutegravir10718.560.951010
Emtricitabine50280150.55050
Maraviroc39.185333
Tenofovir50809.5811644.5621

[back to top]

Peripheral Blood Mononuclear Cells (PBMC) Antiretroviral Concentrations

Concentrations of emtricitabine-triphosphate, tenofovir-diphosphate will be measured in peripheral blood mononuclear cells in the Truvada arm only. Truvada is a combination pill, so results for both FTC and TFV are reported in separate columns. (NCT03218592)
Timeframe: Up to 28 days post-dose

,
Interventionfmol/10^6 cells (Median)
Phase 1 Single DosePhase 2: 7 Doses per WeekPhase 3: 3 Doses Per WeekPhase 3: 1 Dose Per WeekPhase 3: Zero Doses per week
Emtricitabine (Truvada)27.0944602388.69267.8227.48
Tenfovir (Truvada)2.1127.16015.710.4

[back to top]

Efficacy Endpoint: Disease Control Rate

Determine DCR defined as percentage of patients displaying CR/PR or SD as best response according to the RECIST criteria version 1.1. (NCT03274804)
Timeframe: through study completion (20 months)

InterventionParticipants (Count of Participants)
Study Treatment Arm1

[back to top]

Efficacy Endpoint: Objective Response Rate

ORR and immune related (ir) ORR (irORR) will be analyzed. (NCT03274804)
Timeframe: through study completion (20 months)

InterventionParticipants (Count of Participants)
Single Arm, Prospective, Open-label Trial1

[back to top]

Efficacy Endpoint: Progression-free Survival

Individual PFS will be analyzed. (NCT03274804)
Timeframe: through study completion (20 months)

Interventionweeks (Median)
Study Treatment Arm9

[back to top]

Feasibility Rate of a Combined Therapy

"Defined as the rate of patients receiving the protocol treatment according to the planned schedule without occurrence of at least one of the following events:~Study treatment-related Grade ≥ 3 immune-related abnormalities; Study treatment-related Grade ≥ 4 AEs of any aetiology; Any toxic event leading to the premature withdrawal of protocol treatment" (NCT03274804)
Timeframe: After core treatment period of 8 cycles (each cycle is 21 days)

InterventionParticipants (Count of Participants)
Single Arm, Prospective, Open-label Trial18

[back to top]

Overall Survival

Individual OS will be analyzed. (NCT03274804)
Timeframe: through study completion (20 months)

InterventionTime until event, month (Median)
Study Treatment Arm9

[back to top]

Safety and Toxicity of a Combined Therapy Based on Subjects Who Experienced Toxicities

The primary safety analysis will be based on subjects who experienced toxicities as defined by the current National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE, v4.0; Section 11.2). The attribution to drug, time-of-onset, duration of the event, its resolution, and any concomitant medications administered will be recorded. (NCT03274804)
Timeframe: After core treatment period of 8 cycles (each cycle is 21 days)

InterventionParticipants (Count of Participants)
Study Treatment Arm20

[back to top]