1
|
Riddler SA, Kelly CW, Hoesley CJ, Ho KS, Piper JM, Edick S, Heard F, Doncel GF, Johnson S, Anderson PL, Brand RM, Kunjara Na Ayudhya RP, Bauermeister JA, Hillier SL, Hendrix CW. A Phase 1 Clinical Trial to Assess the Safety and Pharmacokinetics of a Tenofovir Alafenamide/Elvitegravir Insert Administered Rectally for HIV Prevention. J Infect Dis 2024; 230:696-705. [PMID: 38655842 PMCID: PMC11420810 DOI: 10.1093/infdis/jiae211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/05/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND On-demand topical products could be an important tool for human immunodeficiency virus (HIV) prevention. We evaluated the safety, pharmacokinetics, and ex vivo pharmacodynamics of a tenofovir alafenamide/elvitegravir (TAF/EVG, 20 mg/16 mg) insert administered rectally. METHODS MTN-039 was a phase 1, open-label, single-arm, 2-dose study. Blood, rectal fluid, and rectal tissue were collected over 72 hours following rectal administration of 1 and 2 TAF/EVG inserts for each participant. RESULTS TAF/EVG inserts were safe and well tolerated. EVG and tenofovir (TFV) were detected in blood plasma at low concentrations: median peak concentrations after 2 inserts were EVG 2.4 ng/mL and TFV 4.4 ng/mL. Rectal tissue EVG peaked at 2 hours (median, 2 inserts = 9 ng/mg) but declined to below limit of quantification in the majority of samples at 24 hours, whereas tenofovir diphosphate (TFV-DP) remained high >2000 fmol/million cells for 72 hours with 2 inserts. Compared to baseline, median cumulative log10 HIV p24 antigen of ex vivo rectal tissue HIV infection was reduced at each time point for both 1 and 2 inserts (P < .065 and P < .039, respectively). DISCUSSION Rectal administration of TAF/EVG inserts achieved high rectal tissue concentrations of EVG and TFV-DP with low systemic drug exposure and demonstrable ex vivo inhibition of HIV infection for 72 hours. Clinical Trials Registration . NCT04047420.
Collapse
Affiliation(s)
- Sharon A Riddler
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Clifton W Kelly
- Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Craig J Hoesley
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medical Education, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ken S Ho
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jeanna M Piper
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Stacey Edick
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Faye Heard
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | | - Peter L Anderson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Rhonda M Brand
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | | | - José A Bauermeister
- Department of Family and Community Health, University of Pennsylvania School of Nursing, Philadelphia, Pennsylvania, USA
| | - Sharon L Hillier
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
- Department of Obstetrics and Gynecology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Craig W Hendrix
- Department of Medicine, Clinical Pharmacology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Chawki S, Goldwirt L, Mouhebb ME, Gabassi A, Taouk M, Bichard I, Loze B, Amara A, Brand R, Siegel A, McGowan I, Costagliola D, Assoumou L, Molina JM, Delaugerre C. Ex-vivo rectal tissue infection with HIV-1 to assess time to protection following oral preexposure prophylaxis with tenofovir disoproxil/emtricitabine. AIDS 2024; 38:455-464. [PMID: 37976073 PMCID: PMC10906210 DOI: 10.1097/qad.0000000000003789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/21/2023] [Accepted: 10/13/2023] [Indexed: 11/19/2023]
Abstract
OBJECTIVES We wished to assess time to protection from HIV-1 infection following oral tenofovir disoproxil and emtricitabine (TDF/FTC) as preexposure prophylaxis (PrEP), using ex-vivo rectal tissue infections and drug concentration measures in blood and rectal tissue. DESIGN/METHODS Participants from the ANRS PREVENIR study (NCT03113123) were offered this sub-study after a 14-day wash-out. We used an ex-vivo model to evaluate rectal tissue HIV-1 susceptibility before and after PrEP, 2 h after two pills or 7 days of a daily pill of TDF/FTC. PrEP efficacy was expressed by the difference (after-before) of 14-day cumulative p24 antigen levels. TFV-DP and FTC-TP levels were measured in rectal tissue and PBMCs and correlated with HIV-1 infection. RESULTS Twelve and 11 men were analyzed in the 2 h-double dose and 7 days-single dose groups, respectively. Cumulative p24 differences after-before PrEP were -144 pg/ml/mg (IQR[-259;-108]) for the 2 h-double dose group ( P = 0.0005) and -179 pg/ml/mg (IQR [-253;-86]) for the 7 days-single dose group ( P = 0.001), with no differences between groups ( P = 0.93). Rectal TFV-DP was below quantification after a double dose, but FTC-TP levels were similar to levels at 7 days. There was a significant correlation between rectal FTC-TP levels and p24 changes after a double dose ( R = -0.84; P = 0.0001). CONCLUSION Oral TDF/FTC provided similar protection against HIV-1 infection of rectal tissue 2 h after a double dose or 7 days of a daily dose. At 2 h, this protection seems driven by high FTC-TP concentrations in rectal tissue. This confirms the importance of combining TDF and FTC to achieve early protection.
Collapse
Affiliation(s)
- Sylvain Chawki
- Université de Paris Cité, INSERM U-944, Institut Recherche Saint Louis
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint Louis, Service de Maladies Infectieuses
| | - Lauriane Goldwirt
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint Louis, Laboratoire de Pharmacologie Biologique
| | - Mayssam El Mouhebb
- Sorbonne Université, INSERM, Institut Pierre Louis d’Epidémiologie et de Santé Publique
| | - Audrey Gabassi
- Université de Paris Cité, INSERM U-944, Institut Recherche Saint Louis
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint Louis, Service de Virologie
| | - Milad Taouk
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint Louis, Service de Gastro-entérologie, Paris, France
| | - Iris Bichard
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint Louis, Service de Maladies Infectieuses
| | - Bénédicte Loze
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint Louis, Service de Maladies Infectieuses
| | - Ali Amara
- Université de Paris Cité, INSERM U-944, Institut Recherche Saint Louis
| | - Rhonda Brand
- University of Pittsburgh, Magee-Women's Research Institute and Foundation, Pittsburgh, Pennsylvania, USA
| | - Aaron Siegel
- University of Pittsburgh, Magee-Women's Research Institute and Foundation, Pittsburgh, Pennsylvania, USA
| | - Ian McGowan
- University of Pittsburgh, Magee-Women's Research Institute and Foundation, Pittsburgh, Pennsylvania, USA
- Orion Biotechnology, Ottawa, Ontario, Canada
| | - Dominique Costagliola
- Sorbonne Université, INSERM, Institut Pierre Louis d’Epidémiologie et de Santé Publique
| | - Lambert Assoumou
- Sorbonne Université, INSERM, Institut Pierre Louis d’Epidémiologie et de Santé Publique
| | - Jean-Michel Molina
- Université de Paris Cité, INSERM U-944, Institut Recherche Saint Louis
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint Louis, Service de Maladies Infectieuses
| | - Constance Delaugerre
- Université de Paris Cité, INSERM U-944, Institut Recherche Saint Louis
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint Louis, Service de Virologie
| |
Collapse
|
3
|
Lantz AM, Nicol MR. Translational Models to Predict Target Concentrations for Pre-Exposure Prophylaxis in Women. AIDS Res Hum Retroviruses 2022; 38:909-923. [PMID: 36097755 PMCID: PMC9805887 DOI: 10.1089/aid.2022.0057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The HIV epidemic remains a significant public health burden. Women represent half of the global HIV epidemic, yet there is an urgent need for a variety of prevention options to meet the needs of more women. Pre-exposure prophylaxis (PrEP) is a valuable prevention tool that uses antiretrovirals before a potential HIV exposure to prevent virus transmission. Development of effective preventive drug regimens for women is dependent on convenient dosing schedules and routes of administration, and on identifying defined target concentrations in mucosal tissues that provide complete protection against HIV transmission. There is a critical need for a translational model that can accurately predict in vivo target concentrations that are completely protective against HIV infection. There is no gold-standard preclinical model to predict PrEP efficacy. In this study, we review the strengths and limitations of three different preclinical models and their utility in predicting target concentrations in the female genital tract: humanized mice, non-human primates, and the ex vivo tissue model.
Collapse
Affiliation(s)
- Alyssa M. Lantz
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Melanie R. Nicol
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
4
|
Berry N, Stein M, Ferguson D, Ham C, Hall J, Giles E, Kempster S, Adedeji Y, Almond N, Herrera C. Mucosal Responses to Zika Virus Infection in Cynomolgus Macaques. Pathogens 2022; 11:1033. [PMID: 36145466 PMCID: PMC9503824 DOI: 10.3390/pathogens11091033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 12/03/2022] Open
Abstract
Zika virus (ZIKV) cases continue to be reported, and no vaccine or specific antiviral agent has been approved for the prevention or treatment of infection. Though ZIKV is primarily transmitted by mosquitos, cases of sexual transmission and prolonged viral RNA presence in semen have been reported. In this observational study, we report the mucosal responses to sub-cutaneous and mucosal ZIKV exposure in cynomolgus macaques during acute and late chronic infection. Subcutaneous challenge induced a decrease in the growth factor VEGF in colorectal and cervicovaginal tissues 100 days post-challenge, in contrast to the observed increase in these tissues following vaginal infection. This different pattern was not observed in the uterus, where VEGF was upregulated independently of the challenge route. Vaginal challenge induced a pro-inflammatory profile in all mucosal tissues during late chronic infection. Similar responses were already observed during acute infection in a vaginal tissue explant model of ex vivo challenge. Non-productive and productive infection 100 days post-in vivo vaginal challenge induced distinct proteomic profiles which were characterized by further VEGF increase and IL-10 decrease in non-infected animals. Ex vivo challenge of mucosal explants revealed tissue-specific modulation of cytokine levels during the acute phase of infection. Mucosal cytokine profiles could represent biosignatures of persistent ZIKV infection.
Collapse
Affiliation(s)
- Neil Berry
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control (NIBSC), Potters Bar EN6 3QC, UK
| | - Monja Stein
- Department of Medicine, Imperial College London, London W2 1PG, UK
| | - Deborah Ferguson
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control (NIBSC), Potters Bar EN6 3QC, UK
| | - Claire Ham
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control (NIBSC), Potters Bar EN6 3QC, UK
| | - Jo Hall
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control (NIBSC), Potters Bar EN6 3QC, UK
| | - Elaine Giles
- Division of Analytical and Biological Sciences, NIBSC, Potters Bar EN6 3QC, UK
| | - Sarah Kempster
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control (NIBSC), Potters Bar EN6 3QC, UK
| | - Yemisi Adedeji
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control (NIBSC), Potters Bar EN6 3QC, UK
| | - Neil Almond
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control (NIBSC), Potters Bar EN6 3QC, UK
| | - Carolina Herrera
- Department of Medicine, Imperial College London, London W2 1PG, UK
| |
Collapse
|
5
|
Herrera C, Cottrell ML, Prybylski J, Kashuba ADM, Veazey RS, García-Pérez J, Olejniczak N, McCoy CF, Ziprin P, Richardson-Harman N, Alcami J, Malcolm KR, Shattock RJ. The ex vivo pharmacology of HIV-1 antiretrovirals differs between macaques and humans. iScience 2022; 25:104409. [PMID: 35663021 PMCID: PMC9157191 DOI: 10.1016/j.isci.2022.104409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 04/12/2022] [Accepted: 05/11/2022] [Indexed: 01/08/2023] Open
Abstract
Non-human primates (NHP) are widely used for the pre-clinical assessment of antiretrovirals (ARVs) for HIV treatment and prevention. However, the utility of these models is questionable given the differences in ARV pharmacology between humans and macaques. Here, we report a model based on ex vivo ARV exposure and the challenge of mucosal tissue explants to define pharmacological differences between NHPs and humans. For colorectal and cervicovaginal explants in both species, high concentrations of tenofovir (TFV) and maraviroc were predictive of anti-viral efficacy. However, their combinations resulted in increased inhibitory potency in NHP when compared to human explants. In NHPs, higher TFV concentrations were measured in colorectal versus cervicovaginal explants (p = 0.042). In humans, this relationship was inverted with lower levels in colorectal tissue (p = 0.027). TFV-resistance caused greater loss of viral fitness for HIV-1 than SIV. This, tissue explants provide an important bridge to refine and appropriately interpret NHP studies. Tenofovir-maraviroc combinations show greater potency in NHP than in human tissue Opposite drug distribution in mucosal tissues was observed between both species Greater loss of viral replication fitness with RT mutations for SIV than for HIV-1 Ex vivo tissue models are a bridge between NHP studies and human clinical trials
Collapse
Affiliation(s)
- Carolina Herrera
- Section of Virology, Faculty of Medicine, St. Mary's Campus, Imperial College London, UK
| | - Mackenzie L Cottrell
- University of North Carolina at Chapel Hill, UNC Eshelman School of Pharmacy, Division of Pharmacotherapy and Experimental Therapeutics, Chapel Hill, NC, USA
| | - John Prybylski
- University of North Carolina at Chapel Hill, UNC Eshelman School of Pharmacy, Division of Pharmacotherapy and Experimental Therapeutics, Chapel Hill, NC, USA
| | - Angela D M Kashuba
- University of North Carolina at Chapel Hill, UNC Eshelman School of Pharmacy, Division of Pharmacotherapy and Experimental Therapeutics, Chapel Hill, NC, USA
| | - Ronald S Veazey
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, USA
| | - Javier García-Pérez
- AIDS Immunopathology Unit. National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Natalia Olejniczak
- Section of Virology, Faculty of Medicine, St. Mary's Campus, Imperial College London, UK
| | - Clare F McCoy
- School of Pharmacy, Medical Biology Centre, Queen's University of Belfast, Belfast, UK
| | - Paul Ziprin
- Department of Surgery and Cancer, St Mary's Hospital, Imperial College London, UK
| | | | - José Alcami
- AIDS Immunopathology Unit. National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain.,HIV Unit, Hospital Clinic-IDIBAPS, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Karl R Malcolm
- School of Pharmacy, Medical Biology Centre, Queen's University of Belfast, Belfast, UK
| | - Robin J Shattock
- Section of Virology, Faculty of Medicine, St. Mary's Campus, Imperial College London, UK
| |
Collapse
|
6
|
Else L, Penchala SD, Pillay AD, Seiphetlo TB, Lebina L, Callebaut C, Minhas S, Morley R, Rashid T, Martinson N, Fox J, Khoo S, Herrera C. Pre-Clinical Evaluation of Tenofovir and Tenofovir Alafenamide for HIV-1 Pre-Exposure Prophylaxis in Foreskin Tissue. Pharmaceutics 2022; 14:pharmaceutics14061285. [PMID: 35745857 PMCID: PMC9227286 DOI: 10.3390/pharmaceutics14061285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/30/2022] [Accepted: 06/12/2022] [Indexed: 02/01/2023] Open
Abstract
Background: HIV-1 pre-exposure prophylaxis (PrEP) has focused predominantly on protective efficacy in receptive sex, with limited research on the dosing requirements for insertive sex. We pre-clinically assessed the ex vivo pharmacokinetic–pharmacodynamic (PK–PD) profile of tenofovir (TFV) and tenofovir alafenamide (TAF) in foreskin tissue. Methods: Inner and outer foreskin explants were exposed to serial dilutions of TFV or TAF prior to addition of HIV-1BaL at a high (HVT) or a low viral titer (LVT). Infection was assessed by measurement of p24 in foreskin culture supernatants. TFV, TAF and TFV–diphosphate (TFV–DP) concentrations were measured in tissues, culture supernatants and dosing and washing solutions. Results: Dose–response curves were obtained for both drugs, with greater potency observed against LVT. Inhibitory equivalency mimicking oral dosing was defined between 1 mg/mL of TFV and 15 µg/mL of TAF against HVT challenge. Concentrations of TFV–DP in foreskin explants were approximately six-fold higher after ex vivo dosing with TAF than with TFV. Statistically significant negative linear correlations were observed between explant levels of TFV or TFV–DP and p24 concentrations following HVT. Conclusions: Pre-clinical evaluation of TAF in foreskin explants revealed greater potency than TFV against penile HIV transmission. Clinical evaluation is underway to support this finding.
Collapse
Affiliation(s)
- Laura Else
- Bioanalytical Facility, Molecular and Clinical Pharmacology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7BE, UK; (L.E.); (S.D.P.); (S.K.)
| | - Sujan D. Penchala
- Bioanalytical Facility, Molecular and Clinical Pharmacology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7BE, UK; (L.E.); (S.D.P.); (S.K.)
| | - Azure-Dee Pillay
- Division of Immunology, University of Cape Town, Cape Town 7935, South Africa; (A.-D.P.); (T.B.S.)
| | - Thabiso B. Seiphetlo
- Division of Immunology, University of Cape Town, Cape Town 7935, South Africa; (A.-D.P.); (T.B.S.)
| | - Limakatso Lebina
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa; (L.L.); (N.M.)
| | | | - Suks Minhas
- Imperial College Healthcare NHS Trust, Charing Cross Hospital, London W6 8RF, UK; (S.M.); (R.M.); (T.R.)
| | - Roland Morley
- Imperial College Healthcare NHS Trust, Charing Cross Hospital, London W6 8RF, UK; (S.M.); (R.M.); (T.R.)
| | - Tina Rashid
- Imperial College Healthcare NHS Trust, Charing Cross Hospital, London W6 8RF, UK; (S.M.); (R.M.); (T.R.)
| | - Neil Martinson
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa; (L.L.); (N.M.)
| | - Julie Fox
- Guys and St. Thomas’ NHS Foundation Trust and King’s College London, London SE1 9RT, UK;
| | - Saye Khoo
- Bioanalytical Facility, Molecular and Clinical Pharmacology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7BE, UK; (L.E.); (S.D.P.); (S.K.)
| | - Carolina Herrera
- Department of Infectious Diseases, Faculty of Medicine, Imperial College, London W2 1PG, UK
- Correspondence: ; Tel.: +44-207-594-2545
| |
Collapse
|
7
|
McGowan IM, Kunjara Na Ayudhya RP, Brand RM, Marzinke MA, Hendrix CW, Johnson S, Piper J, Holtz TH, Curlin ME, Chitwarakorn A, Raengsakulrach B, Doncel G, Schwartz JL, Rooney JF, Cranston RD. An Open-Label Pharmacokinetic and Pharmacodynamic Assessment of Tenofovir Gel and Oral Emtricitabine/Tenofovir Disoproxil Fumarate. AIDS Res Hum Retroviruses 2022; 38:279-287. [PMID: 34541872 DOI: 10.1089/aid.2021.0115] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The Microbicide Trials Network-017 study was undertaken to characterize the safety, acceptability, pharmacokinetic (PK), and pharmacodynamic profile of the reduced-glycerin (RG) 1% tenofovir (RG-TFV) gel compared to oral emtricitabine/tenofovir disoproxil fumarate (FTC/TDF). The study was a Phase 2, three-period, randomized sequence, open-label, expanded safety and acceptability crossover study. In each 8-week study period, HIV-1-uninfected participants were randomized to RG-TFV rectal gel daily or RG-TFV rectal gel before and after receptive anal intercourse (RAI) (or at least twice weekly in the event of no RAI), or daily oral FTC/TDF. A mucosal substudy was conducted at sites in the United States and Thailand. Samples were collected to evaluate PK and ex vivo biopsy challenge with HIV-1. A total of 195 men who have sex with men and transgender women were enrolled in the parent study and 37 in the mucosal substudy. As previously reported, both products were found to be safe and acceptable. Systemic TFV concentrations were significantly higher following oral exposure and daily rectal administration compared to RAI-associated product use (p < .001). All three routes of pre-exposure prophylaxis (PrEP) administration resulted in the inhibition of explant infection (p < .05), and there was a significant inverse correlation between explant HIV-1 p24 and tissue concentrations of TFV and FTC (p < .0001). Despite significant differences in systemic and mucosal drug concentrations, all three PrEP regimens were able to protect rectal explants from ex vivo HIV infection. These data suggest that there is a rationale for co-development of oral and topical antiretroviral PrEP for HIV prevention. Clinical Trial Registration number: NCT01687218.
Collapse
Affiliation(s)
- Ian M. McGowan
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Orion Biotechnology, Ottawa, Ontario, Canada
| | | | - Rhonda M. Brand
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mark A. Marzinke
- Department of Medicine, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Craig W. Hendrix
- Department of Medicine, The Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Jeanna Piper
- National Institute of Allergy and Infectious Disease, Bethesda, Maryland, USA
| | - Timothy H. Holtz
- Division of HIV/AIDS Prevention, United States Centers for Disease Control and Prevention, Atlanta, Georgia, USA
- Thailand Ministry of Public Health, United States Centers for Disease Control and Prevention Collaboration, Bangkok, Thailand
- Office of AIDS Research, NIH, Bethesda, Maryland, USA
| | - Marcel E. Curlin
- Division of HIV/AIDS Prevention, United States Centers for Disease Control and Prevention, Atlanta, Georgia, USA
- Thailand Ministry of Public Health, United States Centers for Disease Control and Prevention Collaboration, Bangkok, Thailand
| | - Anupong Chitwarakorn
- Thailand Ministry of Public Health, United States Centers for Disease Control and Prevention Collaboration, Bangkok, Thailand
| | - Boonyos Raengsakulrach
- Thailand Ministry of Public Health, United States Centers for Disease Control and Prevention Collaboration, Bangkok, Thailand
| | - Gustavo Doncel
- CONRAD/Eastern Virginia Medical School, Arlington, Virginia, USA
| | - Jill L. Schwartz
- CONRAD/Eastern Virginia Medical School, Arlington, Virginia, USA
| | | | - Ross D. Cranston
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | | |
Collapse
|
8
|
McGowan IM, Chawki S, Hendrix CW, Anton PA, Marzinke MA, Brand RM, Engstrom JC, Rohan LC, Abebe KZ, Richardson-Harman N, Siegel A, Reinhart A, Steytler J, Stall R, Spiegel H, Chen B, Achilles SL, Jacobson CE, Khanukova E, Cranston RD. A Randomized, Open-Label, Crossover Phase 1 Safety and Pharmacokinetic Study of Oral Maraviroc and Maraviroc 1% Gel (the CHARM-03 Study). AIDS Res Hum Retroviruses 2022; 38:269-278. [PMID: 34384282 PMCID: PMC9048171 DOI: 10.1089/aid.2021.0096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
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. 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. Male participants received oral and rectal dosing and female participants received oral, rectal, and vaginal dosing. Assessments were undertaken at baseline and following each 8-day period and included collection of plasma, rectal/cervical tissue (CT), and rectal/endocervical/vaginal fluids. Eleven men and nine women were enrolled. Two participants withdrew from the study before receiving study product. There were 25 adverse events, of which 24 were Grade 1 (G1) and one was G2 (unrelated). After eight doses, MVC was quantifiable in all samples following oral, rectal, or vaginal product administration. The highest drug concentrations in plasma, rectal tissue (RT), and CT were associated with oral, rectal, and vaginal drug delivery, respectively. There were significant reductions in tissue drug concentrations when rectal and cervical biopsies were incubated in media before tissue processing for PK (p < .0001). Only oral MVC was associated with limited protection in the rectal explant HIV challenge model (p < .05). There were no immunological changes in RT, and all products were acceptable to participants. In conclusion, all products were found to be safe and acceptable and did not induce local inflammation. The lack of ex vivo efficacy demonstrated in study samples may be due to rapid disassociation of MVC from the explant tissue. ClinicalTrials.gov Identifier: NCT02346084.
Collapse
Affiliation(s)
- Ian M. McGowan
- Orion Biotechnology, Ottawa, Canada
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Sylvain Chawki
- INSERM, Paris, France
- Department of Virology, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, France
| | - Craig W. Hendrix
- Division of Clinical Pharmacology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Peter A. Anton
- Department of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Mark A. Marzinke
- Division of Clinical Pharmacology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Rhonda M. Brand
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | | | - Lisa C. Rohan
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Kaleab Z. Abebe
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Aaron Siegel
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | | | - John Steytler
- International Partnership for Microbicides, Silver Spring, Maryland, USA
| | - Ronald Stall
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hans Spiegel
- Kelly Government Solutions, Department of Health and Human Services, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Beatrice Chen
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Sharon L. Achilles
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | | | - Elena Khanukova
- Division of Clinical Pharmacology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ross D. Cranston
- Department of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
9
|
Cranston RD, Brown E, Bauermeister J, Dunne EF, Hoesley C, Ho K, Johnson S, Lucas J, Dominguez-Islas C, Gundacker H, Peda M, Jacobson CE, Kramzer L, Singh D, Dezzutti CS, Kunjara Na Ayudhya RP, Brand RM, Wang L, Marzinke MA, Piper J, Devlin B, Nuttall J, McGowan I, Hendrix CW. A Randomized, Double Blind, Placebo-Controlled, Phase 1 Safety, and Pharmacokinetic Study of Dapivirine Gel (0.05%) Administered Rectally to HIV-1 Seronegative Adults (MTN-026). AIDS Res Hum Retroviruses 2022; 38:257-268. [PMID: 34498980 DOI: 10.1089/aid.2021.0071] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Dapivirine (DPV), formulated as vaginal ring, demonstrated HIV risk reduction. MTN-026 explored DPV, formulated as rectal gel, for safety, pharmacokinetics (PK), and acceptability. HIV-uninfected men and women aged 18-45 years were enrolled at United States and Thailand sites and randomized 2:1 to receive DPV 0.05% or placebo gel via rectal applicator. A single-dose phase was followed by seven observed daily doses. Plasma and fluid and tissue from both rectum and cervix were collected at baseline and after the final dose over 72 h for PK, ex-vivo HIV-1 biopsy challenge, histology, and flow cytometry. Twenty-eight participants were randomized; 2 terminated early; 9 were female and 19 male; 12 were white, 11 Asian, 4 black, and 1 other race/ethnicity. Mean age was 28.5 and 34.2 years in the DPV and placebo arms, respectively. Thirty adverse events occurred (all Grade 1 or 2, except one unrelated Grade 3) without study arm differences. DPV rectal tissue concentrations [median (interquartile range)] 0.5-1 and 2 h after a single dose were 256 ng/g [below the lower limit of quantification (BLQ)-666] and BLQ (BLQ-600), respectively, then BLQ (BLQ-BLQ) from 24 to 72 h; concentrations following multiple doses were similar. The largest median DPV plasma concentrations were 0.33 ng/mL (0.15-0.48) after one dose and 0.40 (0.33-0.49) after seven doses. The DPV rectal gel was acceptable and without safety concerns. While DPV plasma concentrations were similar to the vaginal ring, rectal tissue concentrations were well below vaginal ring tissue concentrations, suggesting need for reformulation. Clinical trial number: NCT03239483.
Collapse
Affiliation(s)
- Ross D. Cranston
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Elizabeth Brown
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - José Bauermeister
- Department of Family and Community Health, University of Pennsylvania, Pennsylvania, Philadelphia, USA
| | - Eileen F. Dunne
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
- Thailand Ministry of Public Health-US CDC Collaboration, Bangkok, Thailand
| | - Craig Hoesley
- Department of Medical Education, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ken Ho
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | - Clara Dominguez-Islas
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Holly Gundacker
- Statistical Center for HIV/AIDS Research & Prevention, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Melissa Peda
- Statistical Center for HIV/AIDS Research & Prevention, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | | | - Devika Singh
- Microbicide Trials Network, Pittsburgh, Pennsylvania, USA
| | - Charlene S. Dezzutti
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Microbicide Trials Network, Pittsburgh, Pennsylvania, USA
| | | | - Rhonda M. Brand
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lin Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mark A. Marzinke
- Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeanna Piper
- Division of AIDS/NIAID/NIH, Bethesda, Maryland, USA
| | - Bríd Devlin
- International Partnership for Microbicides, Silver Spring, Maryland, USA
| | - Jeremy Nuttall
- International Partnership for Microbicides, Silver Spring, Maryland, USA
| | - Ian McGowan
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Craig W. Hendrix
- Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | |
Collapse
|
10
|
Herrera C, Lwanga J, Lee M, Mantori S, Amara A, Else L, Penchala SD, Egan D, Challenger E, Dickinson L, Boffito M, Shattock R, Khoo S, Fox J. Pharmacokinetic/pharmacodynamic investigation of raltegravir with or without lamivudine in the context of HIV-1 pre-exposure prophylaxis (PrEP). J Antimicrob Chemother 2021; 76:2129-2136. [PMID: 33993302 PMCID: PMC8325523 DOI: 10.1093/jac/dkab136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/05/2021] [Indexed: 02/01/2023] Open
Abstract
Background To characterize their potential use in pre-exposure prophylaxis (PrEP) we compared the pharmacokinetics of raltegravir and lamivudine in genital tissue against ex vivo tissue infection with HIV-1. Methods Open-label trial of 36 HIV-negative females and males randomized to 7 days raltegravir 400 mg twice daily and 7 days raltegravir 400 mg+lamivudine 150 mg twice daily (after washout), or vice versa. Blood, saliva, rectal fluid, rectal tissue, vaginal fluid and vaginal tissue were sampled at baseline and on and off PrEP during a total of 12 days, for pharmacokinetics and antiviral activity via ex vivo HIV-1BaL challenge. Ex vivo infectivity was compared with baseline. The trial has been registered in https://clinicaltrials.gov/ with the identifier NCT03205566. Results Steady state for both drugs was reached by day 4. Dosing with raltegravir alone provided modest ex vivo HIV protection with higher drug levels in rectal tissue and vaginal tissue than in plasma on and off PrEP. Off PrEP, plasma and vaginal concentrations declined rapidly, while persisting in the rectum. On PrEP, the highest lamivudine concentrations were in the rectum, followed by vaginal tissue then plasma. Lamivudine washout was rapid in plasma, while persisting in the rectum and vagina. Raltegravir/lamivudine increased ex vivo protection on and off PrEP compared with raltegravir alone, reaching maximum protection at day 2 in rectal tissue and at day 8 in vaginal tissue. Conclusions Raltegravir 400 mg+lamivudine 150 mg showed high levels of ex vivo HIV protection, associated with high drug concentrations persisting after discontinuation in vaginal and rectal compartments, supporting further investigation of these agents for PrEP.
Collapse
Affiliation(s)
| | - Julianne Lwanga
- Guys and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Ming Lee
- Guys and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Suna Mantori
- Guys and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Alieu Amara
- Department of Pharmacology, University of Liverpool, Liverpool, UK
| | - Laura Else
- Department of Pharmacology, University of Liverpool, Liverpool, UK
| | | | - Deirdre Egan
- Department of Pharmacology, University of Liverpool, Liverpool, UK
| | | | - Laura Dickinson
- Department of Pharmacology, University of Liverpool, Liverpool, UK
| | - Marta Boffito
- Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Robin Shattock
- Department of Medicine, Imperial College London, London, UK
| | - Saye Khoo
- Department of Pharmacology, University of Liverpool, Liverpool, UK
| | - Julie Fox
- Guys and St Thomas' NHS Foundation Trust and King's College London, London, UK
| |
Collapse
|
11
|
Marchetti G, Asmuth D. Women are from venus: implications for diversified sex-based preexposure prophylaxis approaches. AIDS 2021; 35:1691-1693. [PMID: 34270492 DOI: 10.1097/qad.0000000000002995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Giulia Marchetti
- Department of Health Sciences, Clinic of infectious Diseases and Tropical Medicine, University of Milan, ASST Santi Paolo e Carlo, Milan, Italy
| | - David Asmuth
- Division of Infectious Diseases, University of California Davis Medical Center, Sacramento, CA, USA
| |
Collapse
|
12
|
Sekabira R, McGowan I, Yuhas K, Brand RM, Marzinke MA, Manabe YC, Frank I, Eron J, Landovitz RJ, Anton P, Cranston RD, Anderson P, Mayer KH, Amico KR, Wilkin TJ, Chege W, Kekitiinwa AR, McCauley M, Gulick RM, Hendrix CW. Higher colorectal tissue HIV infectivity in cisgender women compared with MSM before and during oral preexposure prophylaxis. AIDS 2021; 35:1585-1595. [PMID: 33831911 PMCID: PMC8483241 DOI: 10.1097/qad.0000000000002907] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The objective of this study was to compare HIV-negative cisgender women (CGW) with MSM for mucosal tissue differences in pharmacokinetics, HIV infectivity and cell phenotype. DESIGN A substudy of HPTN 069/ACTG A5305, 48-week study of three oral candidate preexposure prophylaxis regimens: maraviroc, maraviroc/emtricitabine and maraviroc/tenofovir disoproxil fumarate (TDF) compared with a TDF/emtricitabine control group. METHODS Plasma, peripheral blood mononuclear cells and cervical and colorectal tissue biopsies were collected at Baseline (no drug), Week 24 and 48 (on drug), and Week 49 (1-week postdrug). Drug concentrations were assessed in all matrices. HIV infectivity was assessed using tissue biopsy 'explants' challenged with HIV ex vivo followed by HIV p24 measurement. Flow cytometry evaluated colorectal cell phenotype. RESULTS Thirty-seven CGW and 54 MSM participated. CGW's colorectal explant p24 was higher than MSM before (0.31 log10, P = 0.046), during (1.01-1.19 log10, P = 0.016) and one week after (0.61 log10, P = 0.011) study drug dosing. Pooling regimens, cervical explant p24 did not differ among visits. CGW had higher plasma maraviroc and colorectal tissue tenofovir diphosphate and lower colorectal tissue emtricitabine (all P < 0.005) compared with MSM. Each study drug's cervical tissue concentrations were more than 10-fold below paired colorectal concentrations (P < 0.001). Cell phenotype sex differences included 4% higher CD38+/CD8+ cells at baseline and 3-7% higher CD69+/CD8+ cells throughout Weeks 24-49 in CGW compared with MSM (P < 0.05). CONCLUSION Colorectal explants in CGW demonstrated greater HIV infectivity than MSM with and without study drugs. Small differences in adherence, drug concentration and colorectal tissue flow cytometry cannot fully explain this difference.
Collapse
Affiliation(s)
- Rogers Sekabira
- Baylor College of Medicine Children's Foundation Uganda, Kampala, Uganda
| | - Ian McGowan
- University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA
- Orion Biotechnology, Ottawa, Ontario, Canada
| | - Krista Yuhas
- Fred Hutchinson Cancer Research Center, Medical School, Pittsburgh, Pennsylvania
| | - Rhonda M Brand
- University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA
| | - Mark A Marzinke
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yukari C Manabe
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ian Frank
- University of Pennsylvania, Infectious Disease Division, Philadelphia, Pennsylvania
| | - Joseph Eron
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Raphael J Landovitz
- University of California Los Angeles, Center for Clinical AIDS Research and Education, Los Angeles, California, USA
| | - Peter Anton
- University of California Los Angeles, Center for Clinical AIDS Research and Education, Los Angeles, California, USA
| | | | | | | | - K Rivet Amico
- University of Michigan, School of Public Health, Ann Arbor, Michigan
| | | | - Wairimu Chege
- Prevention Sciences Program, Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | | | | | | | - Craig W Hendrix
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
13
|
McGowan IM, Tzakis N, Kosak B, Korczak B, Engstrom J, Tomaszewska-Kiecana M, Hartley O. Evaluation of the Safety, Acceptability, and Pharmacokinetic Profile of a Gel Formulation of OB-002 in Healthy Volunteers. AIDS Res Hum Retroviruses 2021; 37:453-460. [PMID: 33749321 DOI: 10.1089/aid.2021.0010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OB-002 is an extremely potent CCR5 antagonist that has previously been shown to completely block transmission in a nonhuman primate model of HIV infection. The purpose of this study was to characterize the safety, acceptability, and pharmacokinetic profile of a gel formulation of OB-002 (OB-002H). The trial had two phases, an open label single dose exposure (vaginal and rectal) and a randomized placebo controlled multiple dose phase during which study participants received five vaginal daily doses of OB-002H gel or matched placebo in a 2:1 ratio. Serum OB-002 levels were quantified at multiple time points up to 24 h after the first dose. A total of thirty female and male participants were enrolled in the study (12 in the single dose phase and 18 in the multiple dose phase). All adverse events were Grade 1 or 2, and the majority was unrelated to study product. Only two product-related transient Grade 2 events (both vulval dryness) occurred in the study, both in the OB-002H gel randomized multiple dose arm. All colposcopic and anoscopic assessments following product exposure were normal. There was no evidence of systemic absorption of OB-002. Overall, the product had a positive acceptability profile, and most study participants would consider using the product for protection against HIV or pregnancy. Future studies are needed to assess the extended safety and acceptability of OB-002H gel in sexually active participants. Clinical Trial Registration Number: NCT04791007.
Collapse
Affiliation(s)
- Ian Michael McGowan
- Orion Biotechnology Polska, Krakow, Poland
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | - Oliver Hartley
- Orion Biotechnology, Ottawa, Canada
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
14
|
Translational Approach to Predicting the Efficacy of Maraviroc-Based Regimens as HIV Preexposure Prophylaxis. Antimicrob Agents Chemother 2020; 64:AAC.01729-19. [PMID: 31740561 DOI: 10.1128/aac.01729-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023] Open
Abstract
Maraviroc-based regimens have been explored as preexposure prophylaxis (PrEP) against human immunodeficiency virus (HIV). In this study, we utilized mucosal tissue drug exposure data, combined with target concentrations generated in vitro, in a pharmacokinetic-pharmacodynamic analysis to predict the effects of drug combinations and adherence on PrEP efficacy. Mucosal tissue concentrations of maraviroc were measured in 24 healthy women. The 90% effective concentrations (EC90) of maraviroc (alone and combined with tenofovir and emtricitabine) for protection against HIV were identified in CD4+ T cells. Monte Carlo simulations were performed to identify dosing strategies to protect colorectal and female genital tract (FGT) tissues from HIV infection. Colorectal maraviroc concentrations were 350-fold higher than in the FGT. Under steady-state conditions, our model predicted that one 300-mg dose/week was sufficient to protect colorectal tissue from HIV in 99% of the population, while 300 mg daily would protect the FGT in only 63% of the population. FGT protection increased to >90% when maraviroc was used in combination with tenofovir (5 doses/week) or emtricitabine (3 doses/week). Poor adherence resulted in a drastic decrease in efficacy in the FGT but not colorectal tissue. However, greater forgiveness was seen when maraviroc was combined with tenofovir or emtricitabine, suggesting that maraviroc should not be used alone as PrEP.
Collapse
|
15
|
Abstract
HIV topical microbicides are products with anti-HIV activity, generally incorporating a direct-acting antiretroviral agent, that when applied to the vagina or rectum have the potential to prevent the sexual acquisition of HIV in women and men. Topical microbicides may meet the prevention needs of individuals and groups for whom oral daily forms of pre-exposure prophylaxis (PrEP) have not been acceptable. Microbicides can provide personal control over HIV prevention and offer the possibility of discreet use, qualities that may be particularly important for receptive partners in sexual relationships such as women and transgender women and men, who together account for the clear majority of new HIV infections worldwide. Although the promise of such a product emerged nearly three decades ago, proof of concept has been demonstrated only within the last decade. A robust pipeline of microbicidal gels, films, inserts, and rings has been evaluated in multiple studies among at-risk women and men, and refinement of products for ease of use, reversibility, and high safety is the priority for the field.
Collapse
Affiliation(s)
- Jared M Baeten
- Departments of Global Health, Medicine, and Epidemiology, University of Washington, Seattle, Washington 98104, USA;
| | - Craig W Hendrix
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, USA;
| | - Sharon L Hillier
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Department of Microbiology and Molecular Genetics, University of Pittsburgh, Magee-Womens Research Institute, Pittsburgh, Pennsylvania 15213, USA;
| |
Collapse
|
16
|
Miao M, De Clercq E, Li G. Clinical significance of chemokine receptor antagonists. Expert Opin Drug Metab Toxicol 2020; 16:11-30. [PMID: 31903790 DOI: 10.1080/17425255.2020.1711884] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction: Chemokine receptors are important therapeutic targets for the treatment of many human diseases. This study will provide an overview of approved chemokine receptor antagonists and promising candidates in advanced clinical trials.Areas covered: We will describe clinical aspects of chemokine receptor antagonists regarding their clinical efficacy, mechanisms of action, and re-purposed applications.Expert opinion: Three chemokine antagonists have been approved: (i) plerixafor is a small-molecule CXCR4 antagonist that mobilizes hematopoietic stem cells; (ii) maraviroc is a small-molecule CCR5 antagonist for anti-HIV treatment; and (iii) mogamulizumab is a monoclonal-antibody CCR4 antagonist for the treatment of mycosis fungoides or Sézary syndrome. Moreover, phase 3 trials are ongoing to evaluate many potent candidates, including CCR5 antagonists (e.g. leronlimab), dual CCR2/CCR5 antagonists (e.g. cenicriviroc), and CXCR4 antagonists (e.g. balixafortide, mavorixafor, motixafortide). The success of chemokine receptor antagonists depends on the selective blockage of disease-relevant chemokine receptors which are indispensable for disease progression. Although clinical translation has been slow, antagonists targeting chemokine receptors with multifaced functions offer the potential to treat a broad spectrum of human diseases.
Collapse
Affiliation(s)
- Miao Miao
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Hunan, China
| | - Erik De Clercq
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Leuven, Belgium
| | - Guangdi Li
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Hunan, China
| |
Collapse
|
17
|
Herrera C. The Pre-clinical Toolbox of Pharmacokinetics and Pharmacodynamics: in vitro and ex vivo Models. Front Pharmacol 2019; 10:578. [PMID: 31178736 PMCID: PMC6543330 DOI: 10.3389/fphar.2019.00578] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/06/2019] [Indexed: 01/05/2023] Open
Abstract
Prevention strategies against sexual transmission of human immunodeficiency virus (HIV) are essential to curb the rate of new infections. In the absence of a correlate of protection against HIV infection, pre-clinical evaluation is fundamental to facilitate and accelerate prioritization of prevention candidates and their formulations in a rapidly evolving clinical landscape. Characterization of pharmacokinetic (PK) and pharmacodynamic (PD) properties for candidate inhibitors is the main objective of pre-clinical evaluation. in vitro and ex vivo systems for pharmacological assessment allow experimental flexibility and adaptability at a relatively low cost without raising as significant ethical concerns as in vivo models. Applications and limitations of pre-clinical PK/PD models and future alternatives are reviewed in the context of HIV prevention.
Collapse
Affiliation(s)
- Carolina Herrera
- Section of Virology, Division of Infectious Diseases, Department of Medicine, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|