1
|
Sullivan BP, Craig CA, Bender AT, Blake E, Siriprakaisil O, Sukrakanchana PO, Cressey TR, Drain PK, Olanrewaju AO, Posner JD. Validation of the REverSe TRanscrIptase Chain Termination assay for measuring tenofovir diphosphate in dried blood spots from a clinical pharmacokinetic trial. J Antimicrob Chemother 2025; 80:1141-1147. [PMID: 39995274 PMCID: PMC11962380 DOI: 10.1093/jac/dkaf049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 01/29/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Tenofovir diphosphate concentration in red blood cells is an objective measure of long-term oral pre-exposure prophylaxis (PrEP) or antiretroviral therapy (ART) adherence. However, current methods for measuring tenofovir diphosphate are equipment and capital intensive, limiting widespread adoption. OBJECTIVES Low cost, rapid diagnostics for measuring tenofovir diphosphate may drive clinical adoption of routine drug level measurement as a tool for adherence monitoring of tenofovir disoproxil fumarate-based PrEP or ART. We validate a simple and accessible enzymatic assay [REverSe TRanscrIptase Chain Termination (RESTRICT)] for measuring tenofovir diphosphate in dried blood spots (DBS) obtained from a directly observed therapy study of individuals on PrEP. METHODS We performed RESTRICT measurements on 74 DBS samples from individuals on tenofovir disoproxil fumarate/emtricitabine regimens. We compared RESTRICT measurements with those from a gold-standard method of liquid chromatography tandem mass spectrometry (LC-MS/MS). The ability of RESTRICT to correctly classify DBS tenofovir diphosphate concentrations to established steady-state adherence benchmark concentrations was determined using area under receiver operating characteristic curves (AUCs). RESULTS The RESTRICT measurements of DBS samples were highly correlated with LC-MS/MS measurements of tenofovir diphosphate from DBS (r = -0.90; P < 0.0001). The RESTRICT assay correctly classified DBS samples as above or below established steady-state adherence benchmark concentrations corresponding to low (AUC = 0.974), moderate (AUC = 0.936) and high (AUC = 0.955) levels of adherence. CONCLUSIONS The enzymatic RESTRICT assay can accurately measure tenofovir diphosphate concentrations in DBS specimens using simple procedures and readily available laboratory equipment, offering accessible objective adherence monitoring for persons receiving tenofovir disoproxil fumarate-based PrEP or ART.
Collapse
Affiliation(s)
- Benjamin P Sullivan
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Cosette A Craig
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Andrew T Bender
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Emily Blake
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | | | - Pra-ornsuda Sukrakanchana
- AMS-PHPT Research Collaboration, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Tim R Cressey
- AMS-PHPT Research Collaboration, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Paul K Drain
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Ayokunle O Olanrewaju
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Jonathan D Posner
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
- Department of Family Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
2
|
Beesham I, Jaggernath M, Kriel Y, Hao J, Smith PM, Haberer JE, Hendrix CW, Psaros C, Bangsberg DR, Smit JA, Matthews LT. Longitudinal Changes in Tenofovir and Tenofovir Diphosphate Concentrations Among Pregnant Women Using Oral PrEP for HIV Prevention: Findings From Durban, South Africa. J Acquir Immune Defic Syndr 2025; 98:357-362. [PMID: 39705379 DOI: 10.1097/qai.0000000000003586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 09/24/2024] [Indexed: 12/22/2024]
Abstract
BACKGROUND Pregnant women are vulnerable to HIV acquisition. Oral HIV pre-exposure prophylaxis (PrEP) is safe and effective for use during pregnancy. We describe PrEP adherence among pregnant women using multiple measures. METHODS We conducted a secondary data analysis among women enrolled in a study evaluating an adherence intervention for PrEP among those planning for and with pregnancy in South Africa. Our analysis included women who used PrEP and became pregnant. Longitudinal PrEP use was assessed using concentrations of tenofovir (TFV) in plasma, tenofovir diphosphate (TFV-DP) in dried blood spots, and electronic pillcap data from quarterly visits. Plasma TFV ≤10 ng/mL and TFV-DP ≤16.6 fmol/punch were below quantifiable limits. Data were analyzed during prepregnancy (quarter before pregnancy) and pregnancy trimesters. RESULTS Among 35 women, 69% were 18-24 years old, 40% were nulliparous, and 94% did not know their partner's HIV serostatus. Median pillcap adherence was 55%-80% and was highest during prepregnancy (72%, interquartile range: 54%-86%) and third trimester (80%, interquartile range: 30%-94%). The proportion of women with quantifiable TFV was 47% (n = 8/17) prepregnancy and 33% (n = 9/27), 19% (n = 4/21), and 14% (n = 2/14) for trimesters 1-3, respectively. TFV-DP was detected in 75% of samples (n = 12/16) prepregnancy, and 50% (n = 13/26), 29% (n = 6/21), and 27% (n = 4/15) for trimesters 1-3, respectively. No women acquired HIV during pregnancy. CONCLUSIONS PrEP use declined during pregnancy by all measures. Discrepancies between pillcap measurements and drug concentrations could be due to physiologic changes during pregnancy or under- or overuse of the pillcaps. Determining what drug metabolite concentrations are needed to confer protection during pregnancy is important for optimizing counseling and prevention support.
Collapse
Affiliation(s)
- Ivana Beesham
- Division of Infectious Diseases, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL
- Africa Health Research Institute, Mtubatuba, KwaZulu-Natal, South Africa
| | - Manjeetha Jaggernath
- Wits Maternal Adolescent and Child Health Research Unit (WMRU), Department of Obstetrics and Gynaecology, Faculty of Health Sciences, University of the Witwatersrand, Durban, KwaZulu-Natal, South Africa
| | - Yolandie Kriel
- Wits Maternal Adolescent and Child Health Research Unit (WMRU), Department of Obstetrics and Gynaecology, Faculty of Health Sciences, University of the Witwatersrand, Durban, KwaZulu-Natal, South Africa
| | - Jiaying Hao
- Division of Infectious Diseases, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL
| | - Patricia M Smith
- Division of Infectious Diseases, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL
| | - Jessica E Haberer
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Craig W Hendrix
- Department of Medicine (Clinical Pharmacology), Johns Hopkins University, School of Medicine, Baltimore, MD
| | - Christina Psaros
- Behavioural Medicine Program, Department of Psychiatry, Massachusetts General Hospital, Boston, MA; and
| | | | - Jennifer A Smit
- Wits Maternal Adolescent and Child Health Research Unit (WMRU), Department of Obstetrics and Gynaecology, Faculty of Health Sciences, University of the Witwatersrand, Durban, KwaZulu-Natal, South Africa
| | - Lynn T Matthews
- Division of Infectious Diseases, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL
| |
Collapse
|
3
|
Mainella VA, Branchford B, Nemkov T, Hosford S, Coyle RP, Johnson B, Choi YJ, Williams M, Zheng JH, Bushman L, Kiser JJ, Anderson PL, Brooks KM. Cellular pharmacology of tenofovir alafenamide and emtricitabine in neutrophils and platelets in people with and without HIV. J Antimicrob Chemother 2025:dkaf052. [PMID: 40037647 DOI: 10.1093/jac/dkaf052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/29/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Previous studies have primarily focused on nucleos(t)ide reverse transcriptase inhibitor pharmacology in peripheral blood mononuclear cells (PBMCs) and erythrocytes via dried blood spots (DBS), but not other major blood cells. OBJECTIVES Our objectives were to describe and compare the concentrations of tenofovir-diphosphate (TFV-DP) and emtricitabine-triphosphate (FTC-TP) in DBS, PBMCs, neutrophils, and platelets in people with HIV (PWH) and people without HIV (PWOH). METHODS DBS, PBMCs, neutrophils, and platelets were isolated from whole blood drawn from PWH and PWOH receiving tenofovir alafenamide and emtricitabine. TFV-DP and FTC-TP concentrations were quantified using LC-MS/MS in each cell type. Linear regression models controlled for time on drug, adherence, and time since last dose, where applicable, to determine geometric mean percent differences (95% confidence interval) by HIV status and estimated half-lives. RESULTS Data were available in 13 PWH (96% male) and 30 PWOH (53% male). Compared with PWOH, TFV-DP in DBS was 48.9% (15.6%, 91.9%) higher and FTC-TP in platelets was 36.3% (4.5%, 77.7%) higher; TFV-DP in platelets also trended higher [43.5% (-3.24%, 113%)]. No other cell types significantly differed by HIV status. TFV-DP and FTC-TP demonstrated the longest half-lives in neutrophils, followed by PBMCs and then platelets. After normalizing to cell volume, both drugs accumulated from greatest to least in PBMCs, neutrophils, platelets, and erythrocytes across both PWH and PWOH. CONCLUSIONS Our findings highlight differential drug disposition across cell types that also vary by serostatus in DBS and platelets. The mechanisms and implications of these findings require additional research.
Collapse
Affiliation(s)
- Vincent A Mainella
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E Montview Blvd, Mail Stop C238, Aurora, CO 80045, USA
| | - Brian Branchford
- Versiti Medical Sciences Institute and Blood Research Institute, Medical College of Wisconsin and Children's Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Travis Nemkov
- Department of Biochemistry & Molecular Genetics, School of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Seth Hosford
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E Montview Blvd, Mail Stop C238, Aurora, CO 80045, USA
- Division of Infectious Diseases, School of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ryan P Coyle
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E Montview Blvd, Mail Stop C238, Aurora, CO 80045, USA
| | - Bethany Johnson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E Montview Blvd, Mail Stop C238, Aurora, CO 80045, USA
| | - Ye Ji Choi
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E Montview Blvd, Mail Stop C238, Aurora, CO 80045, USA
| | - Martin Williams
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E Montview Blvd, Mail Stop C238, Aurora, CO 80045, USA
| | - Jia-Hua Zheng
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E Montview Blvd, Mail Stop C238, Aurora, CO 80045, USA
| | - Lane Bushman
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E Montview Blvd, Mail Stop C238, Aurora, CO 80045, USA
| | - Jennifer J Kiser
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E Montview Blvd, Mail Stop C238, Aurora, CO 80045, USA
| | - Peter L Anderson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E Montview Blvd, Mail Stop C238, Aurora, CO 80045, USA
| | - Kristina M Brooks
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E Montview Blvd, Mail Stop C238, Aurora, CO 80045, USA
| |
Collapse
|
4
|
Devanathan AS, Poliseno AJ, White NR, Schauer AP, Sykes C, Weideman AMK, Kilpatrick KW, Hudgens MG, Gay CL, Rosen EP, Dumond JB, Kashuba ADM, Cottrell ML. A Cross-Biomeasure Study to Optimize Antiretroviral Adherence Estimation. J Acquir Immune Defic Syndr 2025; 98:291-299. [PMID: 39813294 DOI: 10.1097/qai.0000000000003570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/21/2024] [Indexed: 01/18/2025]
Abstract
BACKGROUND Incomplete adherence to daily tenofovir disoproxil fumarate/emtricitabine (TDF/FTC) reduces effectiveness. Adherence biomeasures (ie, drug concentrations in biological specimen) are more accurate than self-report. TDF/FTC's intracellular active metabolites (tenofovir-diphosphate; TFVdp and FTC-triphosphate; FTCtp) can be quantified in different types of blood samples to estimate adherence. To optimize adherence estimation, we investigated approaches to measure TFVdp and FTCtp in 4 blood matrices. METHODS Twelve HIV-negative, healthy volunteers were enrolled in a single-center, open-label, 3-phase, directly observed therapy study. LC-MS/MS methods quantified TFVdp/FTCtp in dried blood spots, volumetrically accurate microsampling, upper layer packed cells, and peripheral blood mononuclear cells (PBMCs). Noncompartmental analysis estimated half-lives and accumulation ratios. Correlations characterized relationships between clinical variables and exposure. Regression models were fit to determine concentrations associated with <4 and ≥4 doses/week; correct classification percentages were determined. RESULTS Terminal half-life estimates of 3-4 vs 15-22 days distinguished between moderate-term (FTCtp in all samples; TFVdp in PBMCs) versus long-term (TFVdp in red blood cell-containing matrices) measures. Model-derived thresholds accurately categorized <4 and ≥4 doses/week when including both metabolites for 14- and 28-day dosing periods (81%-91% and 82%-85%, respectively). Within each classification and regression trees analyses containing both moderate- and long-term measures, dried blood spots exhibited highest accuracy to predict stable (74%-94%) and changing (42%-47%) adherence patterns. CONCLUSIONS We demonstrate higher accuracy of moderate-term biomeasures to classify adherence over a 14-day period compared with long-term biomeasures to classify adherence over a 28-day period. Combined moderate- and long-term biomeasures predicted stable and changing adherence patterns, with dried blood spots exhibiting highest accuracy.
Collapse
Affiliation(s)
| | - Amanda J Poliseno
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC
| | - Nicole R White
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC
| | - Amanda P Schauer
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC
| | - Craig Sykes
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC
| | - Ann Marie K Weideman
- Biostatistics Core, Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | - Michael G Hudgens
- Biostatistics Core, Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Cynthia L Gay
- University of North Carolina School of Medicine, Chapel Hill, NC; and
| | - Elias P Rosen
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC
| | - Julie B Dumond
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC
| | - Angela D M Kashuba
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC
- University of North Carolina School of Medicine, Chapel Hill, NC; and
| | | |
Collapse
|
5
|
Irie WC, Nicol MR, Clement M, Bukusi EA, Bekker LG, Molina JM, Stewart J. Event-driven PrEP beyond cisgender men who have sex with men. Lancet HIV 2025; 12:e143-e153. [PMID: 39788131 DOI: 10.1016/s2352-3018(24)00300-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 01/12/2025]
Abstract
Despite advancements in existing antiretroviral-based prevention strategies, including daily oral, locally acting, and injectable options, there is a pressing need for more inclusive and flexible event-driven pre-exposure prophylaxis (PrEP) strategies for all. Event-driven or intermittent dosing of PrEP in populations beyond cisgender men who have sex with men would offer a promising alternative by fitting prevention into the diverse lifestyles of affected populations and thereby advancing health equity. Evidence from PrEP clinical trials, pharmacokinetic studies, modelling studies, and real-world observational research suggests that event-driven PrEP could be a flexible and inclusive option, yet optimal dosing has not been established across sex and gender spectrums. To advance PrEP equity through inclusivity, studies on event-driven PrEP should include people across the gender spectrum. Real-world demonstration studies and simulation studies of optimal dosing strategies are needed. While awaiting further evidence, clinical providers can offer shared decision making and counselling on available data to include event-driven dosing as an option, especially when daily oral, locally acting, or injectable PrEP are not acceptable or preferred methods. Wider access to diverse PrEP options for all populations fosters a more inclusive and effective global HIV prevention strategy.
Collapse
Affiliation(s)
- Whitney C Irie
- Boston College and Harvard Medical School, Chestnut Hill, MA, USA
| | - Melanie R Nicol
- University of Minnesota College of Pharmacy, Minneapolis, MN, USA
| | - Meredith Clement
- Louisiana State University Health Sciences Center-New Orleans, New Orleans, LA, USA
| | - Elizabeth Anne Bukusi
- Kenya Medical Research Institute, Centre for Microbiology Research, Nairobi, Kenya; Departments of Global Health and Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Linda-Gail Bekker
- The Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - Jean-Michel Molina
- University of Paris Cité, INSERM U944, Paris, France; Department of Infectious Diseases, Hôpital Saint-Louis and Lariboisière, Paris, France
| | - Jenell Stewart
- Division of Infectious Diseases, Hennepin Healthcare, Minneapolis, MN, USA; University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
6
|
Meisner A, Xia F, Chan KCG, Mayer K, Wheeler D, Zangeneh S, Donnell D. Estimating the effect of pre-exposure prophylaxis in Black men who have sex with men. Int J Epidemiol 2024; 54:dyae170. [PMID: 39916344 PMCID: PMC11802470 DOI: 10.1093/ije/dyae170] [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: 04/03/2024] [Accepted: 12/11/2024] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Black men who have sex with men (MSM) are disproportionately burdened by the HIV epidemic in the USA. The effectiveness of pre-exposure prophylaxis (PrEP) in preventing HIV infection has been demonstrated through randomized placebo-controlled clinical trials in several populations. Importantly, no such trial has been conducted exclusively among Black MSM in the USA, and it would be unethical and infeasible to do so now. METHODS To estimate the causal effects of PrEP access, initiation, and adherence on HIV risk, we utilized causal inference methods to combine data from two non-randomized studies that exclusively enrolled Black MSM. RESULTS The estimated relative risks of HIV were: (i) 0.52 (95% confidence interval: 0.21, 1.22) for individuals with versus without PrEP access, (ii) 0.48 (0.12, 0.89) for individuals who initiated PrEP but were not adherent versus those who did not initiate, and (iii) 0.23 (0.02, 0.80) for individuals who were adherent to PrEP versus those who did not initiate. CONCLUSION Beyond addressing the knowledge gap around the effect of PrEP in Black MSM in the USA, which may have ramifications for public health, we have provided a framework to combine data from multiple non-randomized studies to estimate causal effects, which has broad utility.
Collapse
Affiliation(s)
- Allison Meisner
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Fan Xia
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States
| | - Kwun C G Chan
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, United States
| | - Kenneth Mayer
- Harvard Medical School, Boston, MA, United States
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, MA, United States
- The Fenway Institute, Boston, MA, United States
- Infectious Diseases Division, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Darrell Wheeler
- State University of New York at New Paltz, New Paltz, NY, United States
| | - Sahar Zangeneh
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, United States
- RTI International, Research Triangle Park, NC, United States
| | - Deborah Donnell
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| |
Collapse
|
7
|
Zewdie K, Muwonge T, Ssebuliba T, Bambia F, Nampewo O, Stein G, Mugwanya KK, Thomas KK, Wyatt C, Yin MT, Wang G, Gandhi M, Mujugira A, Heffron R. A point-of-care tenofovir urine test improves accuracy of self-reported preexposure prophylaxis adherence and increases condomless sex reporting among young women. AIDS 2024; 38:1965-1971. [PMID: 39088310 PMCID: PMC11524765 DOI: 10.1097/qad.0000000000003988] [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: 02/06/2024] [Revised: 05/14/2024] [Accepted: 05/23/2024] [Indexed: 08/03/2024]
Abstract
OBJECTIVES We evaluated a recently developed and validated point-of-care urine tenofovir (POC TFV) test to determine whether its use improves the accuracy of self-reported adherence to preexposure prophylaxis (PrEP) and sexual behavior. DESIGN We enrolled sexually active HIV-negative women ages 16 to 25 years in Kampala, Uganda. METHODS Women were followed quarterly for 24 months with HIV prevention counseling, PrEP dispensation, and adherence counseling. Midway through the study, the POC TFV test was introduced as part of routine study procedures. We examined changes in self-reported PrEP adherence, sexual behavior, and accuracy of self-reported PrEP adherence before and after the introduction of the POC TFV test. RESULTS A total of 146 women receiving PrEP refills had at least one visit with a POC TFV test administered before the study exit. At baseline, the median age was 19 years [interquartile range (IQR): 18-21] and the majority (76%) reported having condomless sex within the last 3 months. Participants more frequently self-reported low PrEP adherence [odds ratio (OR): 2.96, 95% confidence interval (CI): 1.89-4.67, P = 0.001] and condomless sex (OR: 1.47, 95% CI: 1.04-2.06, P = 0.03) during visits using the test compared with visits without the test. The accuracy of self-reported PrEP adherence (determined by concordance with TFV-diphosphate levels) was greater when the test was used (61 versus 24%, OR: 4.86, 95% CI: 2.85-8.30, P < 0.001). CONCLUSION When the POC TFV test was used, we observed greater reporting of condomless sex, low PrEP adherence, and more accurate reports of PrEP adherence. The test could facilitate honest conversations between clients and providers and warrant further investigation.
Collapse
Affiliation(s)
- Kidist Zewdie
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Timothy Muwonge
- Infectious Diseases Institute, Makerere University, Kampala, Uganda
| | | | - Felix Bambia
- Infectious Diseases Institute, Makerere University, Kampala, Uganda
| | - Olivia Nampewo
- Infectious Diseases Institute, Makerere University, Kampala, Uganda
| | - Gabrielle Stein
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Kenneth K. Mugwanya
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA
| | | | - Christina Wyatt
- Department of Medicine, Division of Nephrology, Duke University School of Medicine, Durham, NC
| | - Michael T. Yin
- Department of Medicine, Division of Infectious Disease, Columbia University, New York, NY
| | | | - Monica Gandhi
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Andrew Mujugira
- Infectious Diseases Institute, Makerere University, Kampala, Uganda
- Department of Global Health, University of Washington, Seattle, WA
| | - Renee Heffron
- Department of Global Health, University of Washington, Seattle, WA
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
8
|
Allan‐Blitz L, Mayer KH. Updated guidelines on HIV post-exposure prophylaxis: continued efforts towards increased accessibility. J Int AIDS Soc 2024; 27:e26393. [PMID: 39576221 PMCID: PMC11583823 DOI: 10.1002/jia2.26393] [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: 08/05/2024] [Accepted: 11/04/2024] [Indexed: 11/24/2024] Open
Abstract
INTRODUCTION HIV transmission is ongoing in both high- and low-resource settings. Post-exposure prophylaxis (PEP) remains an important tool in preventing HIV; however, PEP is significantly underutilized. The multitude of barriers to PEP implementation include low patient and provider awareness and acceptability, limited access to treatment and prevention services, and high rates of stigma. The World Health Organization (WHO) recently released updated guidance on the delivery of HIV PEP. This commentary aims to highlight the salient changes, evaluate how such recommendations can overcome the existing barriers to PEP implementation and discuss strategies needed to put the updated guidance into practice. DISCUSSION The 2024 WHO PEP guidelines continue a trend towards increasing access to PEP. Most notably, the WHO now provides strong recommendations that: (1) PEP be delivered in community settings (e.g. pharmacies, police stations and online platforms), and (2) PEP delivery and monitoring be done via task sharing involving non-specialist health workers (e.g. pharmacists or community health workers). The guidelines also emphasize that the PEP encounter is an important educable moment whereby a transition to pre-exposure prophylaxis among individuals at continued risk for HIV infection should be discussed. The decentralization of PEP delivery has the potential to overcome numerous barriers to PEP implementation, reduce time to initiation and support adherence with the 28-day course. To translate the recommendations into delivery programmes, however, much more work is needed. Detailed templates can help overcome the heterogeneity of both the community settings in which PEP can now be provided and the populations (e.g. survivors of sexual assault, healthcare workers, sex workers, etc.) among whom PEP may be indicated. Training of the workforce will be essential, which should include, as emphasized by the WHO, training in trauma-based care. Novel formulations of and delivery mechanisms for PEP are also emerging, and how such iterations can synergize with decentralized PEP delivery programmes remains to be seen. CONCLUSIONS The updated WHO PEP guidelines make major strides towards increasing access to PEP. Realization of such aims will require ongoing evaluation and support given the heterogeneity in who benefits most from PEP.
Collapse
Affiliation(s)
- Lao‐Tzu Allan‐Blitz
- Division of Global Health Equity, Department of MedicineBrigham and Women's HospitalBostonMassachusettsUSA
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Kenneth H. Mayer
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
- The Fenway Institute of Fenway HealthBostonMassachusettsUSA
- Fenway HealthBostonMassachusettsUSA
- Division of Infectious Diseases, Department of MedicineBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| |
Collapse
|
9
|
Cooper SE, Zhang S, Haines D, Mayer KH, Amico KR, Landovitz RJ, Hendrix CW, Marzinke MA, Chege W, McCauley M, Gulick RM. Pharmacologic Drug Detection and Self-Reported Adherence in the HPTN069/ACTG5305 Phase II PrEP Trial. AIDS Behav 2024; 28:3710-3718. [PMID: 39083151 PMCID: PMC11471692 DOI: 10.1007/s10461-024-04451-7] [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] [Accepted: 07/20/2024] [Indexed: 09/14/2024]
Abstract
Adherence drives efficacy in PrEP clinical trials. We compared drug concentrations and self-reported adherence in HPTN069/ACTG5305, a double-blinded, randomized trial of the safety and tolerability of candidate PrEP regimens that included maraviroc (MVC), tenofovir (TDF), and emtricitabine (FTC). Plasma drug concentrations and self-reported adherence by computer-assisted self-interview (CASI) were assessed at study weeks 24 and 48. Descriptive statistics and a generalized linear model were used to assess the association between selected demographic factors, self-report of daily medication adherence and plasma drug concentrations consistent with daily adherence. Among 718 paired observations from 370 participants, 43% (306/718) reported daily adherence by CASI, 65% (467/718) had drug concentrations consistent with daily adherence and 11% (81/718) had CASI responses that reported daily adherence despite having drug concentrations consistent with less-than-daily adherence. In adjusted analyses, participants who were assigned male at birth (aOR 1.42 [95% CI 1.02, 1.97]), older (5-year increments aOR 1.10 [95% CI 1.09, 1.11]), White (aOR 2.2 [95% CI 1.88, 2.56]), had advanced education (aOR 3.89 [95% CI 2.97, 5.09]), were employed (aOR 1.89 [95% CI 1.50, 2.40]), or partnered/married (aOR 2 [95% CI 1.72, 2.32]) were more likely to have drug concentrations consistent with daily adherence. Participants who were not employed (aOR 2.7 [95% CI 1.31, 5.55]) or who were single/not partnered (aOR 2.33 [CI 95% 1.25, 4.34]) were more likely to have drug concentrations that did not reflect daily adherence despite self-reported PrEP adherence. These findings support the need for ongoing adherence counseling in clinical trials of new PrEP regimens.
Collapse
Affiliation(s)
- Stanley E Cooper
- Division of Infectious Diseases, Weill Cornell Medicine, 525 East 68th Street, Baker 24, New York, NY, 10065, USA.
| | | | | | | | | | | | - Craig W Hendrix
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mark A Marzinke
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | - Roy M Gulick
- Division of Infectious Diseases, Weill Cornell Medicine, 525 East 68th Street, Baker 24, New York, NY, 10065, USA
| |
Collapse
|
10
|
Wu L, Niu X, Brunelli MK, Mugwanya KK. Adherence and HIV Protection Thresholds for Emtricitabine and Tenofovir Disoproxil Fumarate Preexposure Prophylaxis among Cisgender Women: A Systematic Review. Curr HIV/AIDS Rep 2024; 21:264-281. [PMID: 39120667 DOI: 10.1007/s11904-024-00705-0] [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] [Accepted: 07/08/2024] [Indexed: 08/10/2024]
Abstract
PURPOSE OF REVIEW Adherence-concentration-efficacy benchmarks have not been fully characterized for cisgender women using emtricitabine/tenofovir disoproxil fumarate (FTC/TDF) oral daily pre-exposure prophylaxis (PrEP) for HIV prevention. RECENT FINDINGS We conducted a systematic review to investigate current evidence on the adherence-concentration-efficacy relationship of tenofovir-diphosphate (TFV-DP) derived from FTC/TDF PrEP in dried blood spots (DBS) and peripheral mononuclear cells (PBMC) in cisgender women without HIV, including during pregnancy. We searched for completed and ongoing studies published before May 2024 in PubMed, Embase, Cochrane Library, CINAHL, and clinicaltrial.gov. Overall, 11 studies assessing adherence benchmarks focusing on (n = 5) or involving (n = 6) cisgender women were included. Women-specific median steady-state TFV-DP concentration for daily dosing ranged from 17 to 51 fmol/106 in PBMC and 1389 to 1685 fmol/punch in DBS in non-pregnant women; 50 to 71 fmol/106 in PBMC and 583 to 965 fmol/punch in DBS in pregnant women; and 618 to 1406 fmol/punch in DBS in postpartum women. DBS TFV-DP levels were 14-43% lower in pregnancy versus postpartum or non-pregnant periods, but PBMC TFV-DP levels appear to be comparable. Clinical and modeling studies demonstrate effective HIV protection for women taking at least four doses/week of oral TDF-based PrEP, and emerging evidence suggests that systemic drug levels are more likely to be predictive of efficacy than local tissue levels at the site of exposure. The preponderance of emerging evidence points to comparable efficacy and similar adherence requirement for women as men among those with detectable drug levels, although there was an indication that the highest achievable efficacy may be reached at a lower adherence level in men than women. In this review, we found evidence that women-specific TFV-DP adherence benchmarks in DBS and PBMC are within range of US-based historical thresholds derived from healthy men and women. Emerging evidence suggests that imperfect but adequate adherence to oral FTC/TDF PrEP with at least four doses/week provides sufficient HIV protection in cisgender women as it does in MSM, but more data are still needed to refine intrinsic achievable efficacy estimates for cisgender women.
Collapse
Affiliation(s)
- Linxuan Wu
- Department of Global Health, University of Washington, 908 Jefferson Street, Seattle, WA, 98104, USA
- Department of Epidemiology, University of Washington, 3980 15th Ave NE, Seattle, WA, 98195, USA
| | - Xin Niu
- Department of Global Health, University of Washington, 908 Jefferson Street, Seattle, WA, 98104, USA
- Department of Epidemiology, University of Washington, 3980 15th Ave NE, Seattle, WA, 98195, USA
| | | | - Kenneth K Mugwanya
- Department of Global Health, University of Washington, 908 Jefferson Street, Seattle, WA, 98104, USA.
- Department of Epidemiology, University of Washington, 3980 15th Ave NE, Seattle, WA, 98195, USA.
| |
Collapse
|
11
|
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
|
12
|
Gelé T, Gouget H, Dimant N, Furlan V, Collins J, Scholz EMB, Parry CM, Le Grand R, Lambotte O, Desjardins D, Barrail-Tran A. Whole-body distribution of tenofovir, emtricitabine and dolutegravir in non-human primates. J Antimicrob Chemother 2024; 79:2213-2220. [PMID: 39086094 DOI: 10.1093/jac/dkae216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 05/23/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND One major barrier to HIV cure is the persistence of virus, possibly linked to an insufficient antiretroviral drug (ARV) distribution into tissues. OBJECTIVES To draw the whole-body distribution of three antiretroviral drugs-tenofovir disoproxil fumarate, emtricitabine and dolutegravir-in non-human primates (NHPs). METHODS Eight uninfected NHPs received a single injection of a solution containing the three ARVs. Forty-five different tissues were sampled 24 h after injection. RESULTS Median tissue penetration factors (TPFs) were 45.4, 5.8 and 0.5 for tenofovir, emtricitabine and dolutegravir, respectively, and were statistically different between the three ARVs. Tissues were grouped by system, because TPFs were consistent according to these groups, and ranked in order of decreasing TPFs. The digestive system was the system with the highest tissue concentrations. Next came the two main sites of elimination, the liver and the kidney, as well as the tissues of the cardiopulmonary and urinary systems. Then, it was the whole lymphatic system. The next group included the reproductive system, the adipose tissue and the skin. The last two systems were the muscle and the CNS. The intra-tissue variability was rather low with a median coefficient of variation of the concentrations around 15% and no value greater than 80%. CONCLUSIONS Overall, this study determines the first whole-body distribution in a validated NHP model. These data have important implications for future preclinical and clinical studies for the development of novel HIV therapies towards an HIV cure.
Collapse
Affiliation(s)
- Thibaut Gelé
- Immunologie des Maladies Virales, Auto-immunes, Hématologiques et Bactériennes, Université Paris-Saclay, Inserm, CEA, 92265 Fontenay-aux-Roses, France
| | - Hélène Gouget
- Immunologie des Maladies Virales, Auto-immunes, Hématologiques et Bactériennes, Université Paris-Saclay, Inserm, CEA, 92265 Fontenay-aux-Roses, France
| | - Nastasia Dimant
- Immunologie des Maladies Virales, Auto-immunes, Hématologiques et Bactériennes, Université Paris-Saclay, Inserm, CEA, 92265 Fontenay-aux-Roses, France
| | - Valérie Furlan
- Service de Pharmacologie-Toxicologie, AP-HP. Université Paris-Saclay, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Jon Collins
- Research & Development, ViiV Healthcare, Blackwell Street, Durham, NC, USA
| | - Erin M B Scholz
- Research & Development, GlaxoSmithKline, Research Triangle Park, Durham, NC, USA
| | - Chris M Parry
- Research & Development, ViiV Healthcare, 980 Great West Road, London TW8 9GS, UK
| | - Roger Le Grand
- Immunologie des Maladies Virales, Auto-immunes, Hématologiques et Bactériennes, Université Paris-Saclay, Inserm, CEA, 92265 Fontenay-aux-Roses, France
| | - Olivier Lambotte
- Immunologie des Maladies Virales, Auto-immunes, Hématologiques et Bactériennes, Université Paris-Saclay, Inserm, CEA, 92265 Fontenay-aux-Roses, France
- Service de Médecine Interne Immunologie Clinique, AP-HP. Université Paris-Saclay, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Delphine Desjardins
- Immunologie des Maladies Virales, Auto-immunes, Hématologiques et Bactériennes, Université Paris-Saclay, Inserm, CEA, 92265 Fontenay-aux-Roses, France
| | - Aurélie Barrail-Tran
- Immunologie des Maladies Virales, Auto-immunes, Hématologiques et Bactériennes, Université Paris-Saclay, Inserm, CEA, 92265 Fontenay-aux-Roses, France
- Service de Pharmacie, AP-HP. Université Paris-Saclay, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| |
Collapse
|
13
|
Matthews LT, Jaggernath M, Kriel Y, Smith PM, Haberer JE, Baeten JM, Hendrix CW, Ware NC, Moodley P, Pillay M, Bennett K, Bassler J, Psaros C, Hurwitz KE, Bangsberg DR, Smit JA. Oral preexposure prophylaxis uptake, adherence, and persistence during periconception periods among women in South Africa. AIDS 2024; 38:1342-1354. [PMID: 38752557 PMCID: PMC11211057 DOI: 10.1097/qad.0000000000003925] [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: 01/18/2024] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 05/28/2024]
Abstract
OBJECTIVE We developed the Healthy Families-PrEP intervention to support HIV-prevention during periconception and pregnancy. We evaluated preexposure prophylaxis (PrEP) use with three objective measures. DESIGN This single-arm intervention study enrolled women in KwaZulu-Natal, South Africa, who were HIV-uninfected, not pregnant, in a relationship with a partner with HIV or unknown-serostatus, and with pregnancy plans. PrEP was offered as part of a comprehensive HIV prevention intervention. Participants were followed for 12 months. METHODS We evaluated periconception PrEP uptake and adherence using quarterly plasma tenofovir concentrations. We modeled factors associated with PrEP uptake and high plasma tenofovir (past day dosing). Patterns of use were analyzed using electronic pillcap data. Dried blood spots to measure intracellular tenofovir product (past 2 months dosing) were analyzed for a subset of women. RESULTS Three hundred thirty women with median age 24 (IQR: 22-27) years enrolled. Partner HIV-serostatus was unknown by 96% ( N = 316); 60% (195) initiated PrEP. High plasma tenofovir concentrations were seen in 35, 25, 22, and 20% of samples at 3, 6, 9, and 12 months, respectively. Similar adherence was measured by pillcap and dried blood spots. In adjusted models, lower income, alcohol use, and higher HIV stigma were associated with high plasma tenofovir. Eleven HIV-seroconversions were observed (incidence rate: 4.04/100 person-years [95% confidence interval: 2.24-7.30]). None had detectable plasma tenofovir. CONCLUSION The Healthy Families-PrEP intervention supported women in PrEP use. We observed high interest in periconception PrEP and over one-third adhered to PrEP in the first quarter; one-fifth were adherent over a year. High HIV incidence highlights the importance of strategies to reduce HIV incidence among periconception women. CLINICAL TRIAL NUMBER NCT03194308.
Collapse
Affiliation(s)
- Lynn T. Matthews
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Manjeetha Jaggernath
- Maternal Adolescent and Child Health Research Unit (MRU), Department of Obstetrics and Gynaecology, University of the Witwatersrand, Faculty of Health Sciences, Durban, South Africa
| | - Yolandie Kriel
- Maternal Adolescent and Child Health Research Unit (MRU), Department of Obstetrics and Gynaecology, University of the Witwatersrand, Faculty of Health Sciences, Durban, South Africa
| | - Patricia M. Smith
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jessica E. Haberer
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jared M. Baeten
- Department of Global Health
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington
| | - Craig W. Hendrix
- Department of Medicine (Clinical Pharmacology), Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Norma C. Ware
- Department of Global Health & Social Medicine, Harvard Medical School
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Pravi Moodley
- University of KwaZulu-Natal, School of Laboratory Medicine and Medical Sciences, National Health Laboratory Service
| | - Melendhran Pillay
- Department of Virology, National Health Laboratory Service, Durban, South Africa
| | | | - John Bassler
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama
| | - Christina Psaros
- Department of Psychiatry, Behavioural Medicine Program, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | - Jennifer A. Smit
- Maternal Adolescent and Child Health Research Unit (MRU), Department of Obstetrics and Gynaecology, University of the Witwatersrand, Faculty of Health Sciences, Durban, South Africa
| |
Collapse
|
14
|
Adeyemi OA, Nowak RG, Morgan D, Sam-Agudu NA, Craddock J, Zhan M, Crowell TA, Baral S, Adebajo S, Charurat ME. Risk Compensation After Initiation of Daily Oral Pre-exposure Prophylaxis Among Sexual and Gender Minorities in Nigeria. ARCHIVES OF SEXUAL BEHAVIOR 2024; 53:2807-2816. [PMID: 38684621 PMCID: PMC11758484 DOI: 10.1007/s10508-024-02859-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 01/21/2024] [Accepted: 03/21/2024] [Indexed: 05/02/2024]
Abstract
Pre-exposure prophylaxis (PrEP) use may be associated with risk compensation. We enrolled and provided PreP to sexual and gender minorities (SGM) in Abuja, Nigeria between April 2018 and May 2019. Behavioral information and samples for urogenital and anorectal Chlamydia trachomatis and Neisseria gonorrhoeae sexually transmitted infections (STIs) were collected at baseline. Blood samples for PrEP assay and self-reported adherence were collected at three-monthly follow-up visits. STIs were detected using Aptima Combo2 assay. We estimated the odds ratios (ORs) of PCR-diagnosed bacterial STIs and self-reported behavioral outcomes (condomless anal intercourse [CAI] and concurrent sexual relationships) with conditional logistic regression. Of 400 SGM who initiated PrEP, 206 were eligible for analysis, and had a median age of 24 years (IQR 22-27). In multivariable analysis, participants in the PrEP period had decreased odds of CAI (adjusted OR: 0.49, 95% CI 0.28, 0.84). PrEP use was not associated with risk compensation.
Collapse
Affiliation(s)
- Olusegun A Adeyemi
- Department of Public Health and Epidemiology, University of Maryland School of Medicine, 725 West Lombard Street, S422, Baltimore, MD, 21201, USA.
- International Research Center of Excellence, Institute of Human Virology Nigeria, Abuja, Nigeria.
| | - Rebecca G Nowak
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Daniel Morgan
- Department of Public Health and Epidemiology, University of Maryland School of Medicine, 725 West Lombard Street, S422, Baltimore, MD, 21201, USA
| | - Nadia A Sam-Agudu
- International Research Center of Excellence, Institute of Human Virology Nigeria, Abuja, Nigeria
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jaih Craddock
- School of Medicine, University of California, Irvine, CA, USA
| | - Min Zhan
- Division of Biostatistics and Bioinformatics, Department of Public Health and Epidemiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Trevor A Crowell
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- U.S Military HIV Research Program, Silver Spring, MD, USA
| | - Stefan Baral
- The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sylvia Adebajo
- Center for International Health, Education, and Biosecurity, Abuja, Nigeria
| | - Manhattan E Charurat
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
15
|
Massih SA, Atta MG, Thio CL, Tornheim JA, Fuchs EJ, Bakshi RP, Marzinke MA, Hendrix CW, Weld ED. Pharmacokinetics of tenofovir alafenamide, emtricitabine, and dolutegravir in a patient on peritoneal dialysis. AIDS Res Ther 2024; 21:34. [PMID: 38773606 PMCID: PMC11110198 DOI: 10.1186/s12981-024-00616-5] [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: 11/13/2023] [Accepted: 04/11/2024] [Indexed: 05/24/2024] Open
Abstract
INTRODUCTION Peritoneal dialysis (PD) is an effective renal replacement modality in people with HIV (PWH) with end-stage kidney disease (ESKD), particularly those with residual kidney function. Data on pharmacokinetics (PK) of antiretrovirals in patients on peritoneal dialysis are limited. METHODS A single-participant study was performed on a 49-year-old gentleman with ESKD on PD and controlled HIV on once daily dolutegravir (DTG) 50 mg + tenofovir alafenamide (TAF) 25 mg / emtricitabine (FTC) 200 mg. He underwent serial blood plasma, peripheral blood mononuclear cell, and urine PK measurements over 24 h after an observed DTG + FTC/TAF dose. RESULTS Plasma trough (Cmin) concentrations of TAF, tenofovir (TFV), FTC, and DTG were 0.05, 164, 1,006, and 718 ng/mL, respectively. Intracellular trough concentrations of TFV-DP and FTC-TP were 1142 and 11,201 fmol/million cells, respectively. Compared to published mean trough concentrations in PWH with normal kidney function, observed TFV and FTC trough concentrations were 15.5- and 20-fold higher, while intracellular trough concentrations of TFV-DP and FTC-TP were 2.2-fold and 5.4-fold higher, respectively. TFV and FTC urine levels were 20 times lower than in people with normal GFR. CONCLUSIONS In a single ESKD PWH on PD, daily TAF was associated with plasma TFV and intracellular TFV-DP trough concentrations 15-fold and 2-fold higher than those of people with uncompromised kidney function, potentially contributing to nephrotoxicity. This suggests that TFV accumulates on PD; thus, daily TAF in PD patients may require dose adjustment or regimen change to optimize treatment, minimize toxicity, and preserve residual kidney function.
Collapse
Affiliation(s)
- Sandra Abdul Massih
- Division of Clinical Pharmacology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mohamed G Atta
- Division of Nephrology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chloe L Thio
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins University School of Medicine Baltimore, Baltimore, MD, USA
| | - Jeffrey A Tornheim
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins University School of Medicine Baltimore, Baltimore, MD, USA
| | - Edward J Fuchs
- Division of Clinical Pharmacology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rahul P Bakshi
- Division of Clinical Pharmacology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mark A Marzinke
- Division of Clinical Pharmacology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Craig W Hendrix
- Division of Clinical Pharmacology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins University School of Medicine Baltimore, Baltimore, MD, USA
| | - Ethel D Weld
- Division of Clinical Pharmacology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins University School of Medicine Baltimore, Baltimore, MD, USA.
| |
Collapse
|
16
|
Weld ED, McGowan I, Anton P, Fuchs EJ, Ho K, Carballo-Dieguez A, Rohan LC, Giguere R, Brand R, Edick S, Bakshi RP, Parsons T, Manohar M, Seigel A, Engstrom J, Elliott J, Jacobson C, Bagia C, Wang L, Al-khouja A, Hartman DJ, Bumpus NN, Spiegel HML, Marzinke MA, Hendrix CW. Tenofovir Douche as HIV Preexposure Prophylaxis for Receptive Anal Intercourse: Safety, Acceptability, Pharmacokinetics, and Pharmacodynamics (DREAM 01). J Infect Dis 2024; 229:1131-1140. [PMID: 38019657 PMCID: PMC11011183 DOI: 10.1093/infdis/jiad535] [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: 09/02/2023] [Revised: 11/14/2023] [Accepted: 11/27/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Despite highly effective HIV preexposure prophylaxis (PrEP) options, no options provide on-demand, nonsystemic, behaviorally congruent PrEP that many desire. A tenofovir-medicated rectal douche before receptive anal intercourse may provide this option. METHODS Three tenofovir rectal douches-220 mg iso-osmolar product A, 660 mg iso-osmolar product B, and 660 mg hypo-osmolar product C-were studied in 21 HIV-negative men who have sex with men. We sampled blood and colorectal tissue to assess safety, acceptability, pharmacokinetics, and pharmacodynamics. RESULTS The douches had high acceptability without toxicity. Median plasma tenofovir peak concentrations for all products were several-fold below trough concentrations associated with oral tenofovir disoproxil fumarate (TDF). Median colon tissue mucosal mononuclear cell (MMC) tenofovir-diphosphate concentrations exceeded target concentrations from 1 hour through 3 to 7 days after dosing. For 6-7 days after a single product C dose, MMC tenofovir-diphosphate exceeded concentrations expected with steady-state oral TDF 300 mg on-demand 2-1-1 dosing. Compared to predrug baseline, HIV replication after ex vivo colon tissue HIV challenge demonstrated a concentration-response relationship with 1.9 log10 maximal effect. CONCLUSIONS All 3 tenofovir douches achieved tissue tenofovir-diphosphate concentrations and colorectal antiviral effect exceeding oral TDF and with lower systemic tenofovir. Tenofovir douches may provide a single-dose, on-demand, behaviorally congruent PrEP option, and warrant continued development. Clinical Trials Registration . NCT02750540.
Collapse
Affiliation(s)
- Ethel D Weld
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ian McGowan
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Orion Biotechnology, Ottawa, Ontario, Canada
| | - Peter Anton
- Division of Gastroenterology, Department of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Edward J Fuchs
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ken Ho
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alex Carballo-Dieguez
- HIV Center for Clinical and Behavioral Studies, Columbia University and NewYork State Psychiatric Institute, New York, New York, USA
| | - Lisa C Rohan
- Magee Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Rebecca Giguere
- HIV Center for Clinical and Behavioral Studies, Columbia University and NewYork State Psychiatric Institute, New York, New York, USA
| | - Rhonda Brand
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Magee Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Stacey Edick
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rahul P Bakshi
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Teresa Parsons
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Madhuri Manohar
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aaron Seigel
- Magee Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Jared Engstrom
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Julie Elliott
- Division of Gastroenterology, Department of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Cindy Jacobson
- Magee Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Christina Bagia
- Magee Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Lin Wang
- Magee Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Amer Al-khouja
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Douglas J Hartman
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Namandje N Bumpus
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hans M L Spiegel
- Kelly Government Solutions, Contractor to Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Mark A Marzinke
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Craig W Hendrix
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
17
|
Meisner A, Xia F, Chan KCG, Mayer K, Wheeler D, Zangeneh S, Donnell D. Estimating the Effect of PrEP in Black Men Who Have Sex with Men: A Framework to Utilize Data from Multiple Non-Randomized Studies to Estimate Causal Effects. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.10.24301113. [PMID: 38260494 PMCID: PMC10802753 DOI: 10.1101/2024.01.10.24301113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Black men who have sex with men (MSM) are disproportionately burdened by the HIV epidemic in the US. The effectiveness of pre-exposure prophylaxis (PrEP) in preventing HIV infection has been demonstrated through randomized placebo-controlled clinical trials in several populations. Importantly, no such trial has been conducted exclusively among Black MSM in the US, and it would be unethical and infeasible to do so now. To estimate the causal effects of PrEP access, initiation, and adherence on HIV risk, we utilized causal inference methods to combine data from two non-randomized studies that exclusively enrolled Black MSM. The estimated relative risks of HIV were: (i) 0.52 (95% confidence interval: 0.21, 1.22) for individuals with versus without PrEP access, (ii) 0.48 (0.12, 0.89) for individuals who initiated PrEP but were not adherent versus those who did not initiate, and (iii) 0.23 (0.02, 0.80) for individuals who were adherent to PrEP versus those who did not initiate. Beyond addressing the knowledge gap around the effect of PrEP in Black MSM in the US, which may have ramifications for public health, we have provided a framework to combine data from multiple non-randomized studies to estimate causal effects, which has broad utility.
Collapse
Affiliation(s)
- Allison Meisner
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, US
| | - Fan Xia
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, US
| | - Kwun C G Chan
- Department of Biostatistics, University of Washington, Seattle, WA, US
| | - Kenneth Mayer
- Harvard Medical School, Boston, MA, US
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, MA, US
- The Fenway Institute, Boston, MA, US
- Infectious Diseases Division, Beth Israel Deaconess Medical Center, Boston, MA, US
| | - Darrell Wheeler
- State University of New York at New Paltz, New Paltz, NY, US
| | - Sahar Zangeneh
- RTI International, Research Triangle Park, NC, US
- School of Public Health, University of Washington, Seattle, WA, US
| | - Deborah Donnell
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, WA, US
- Department of Global Health, University of Washington, Seattle, WA, US
| |
Collapse
|
18
|
Liu AY, Gundacker H, Richardson B, Chen BA, Hoesley C, van der Straten A, Brown A, Beamer M, Robinson J, Jacobson CE, Scheckter R, Bunge K, Schwartz J, Thurman A, Piper JM, Marzinke MA. Phase 1 randomized pharmacokinetic and safety study of a 90-day tenofovir vaginal ring in the United States. J Int AIDS Soc 2024; 27:e26223. [PMID: 38444118 PMCID: PMC10935712 DOI: 10.1002/jia2.26223] [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: 05/19/2023] [Accepted: 02/05/2024] [Indexed: 03/07/2024] Open
Abstract
INTRODUCTION Tenofovir-based oral pre-exposure prophylaxis is currently approved for HIV prevention; however, adherence in women has been low. A vaginal gel containing tenofovir (TFV) demonstrated partial protection to HIV but protection was not confirmed in additional studies. Vaginal rings offer user-controlled long-acting HIV prevention that could overcome adherence and protection challenges. TFV may also help prevent herpes simplex virus type 2 acquisition when delivered intravaginally. We evaluated the pharmacokinetics, safety, adherence and acceptability of a 90-day TFV ring. METHODS Between January and June 2019, Microbicide Trials Network (MTN)-038 enrolled 49 HIV-negative participants into a phase 1, randomized (2:1) trial comparing a 90-day ring containing 1.4 grams (g) TFV to a placebo ring. TFV concentrations were quantified in plasma, cervicovaginal fluid (CVF), rectal fluid and cervical tissue, and TFV-diphosphate (TFV-DP) in cervical tissue. Used rings were analysed for residual TFV. Safety was assessed by adverse events (AEs); acceptability and adherence by self-report. RESULTS Mean age was 29.5; 46 identified as cisgender-female and three gender non-conforming. There were no differences in the proportion of participants with grade ≥2 genitourinary AEs in the TFV versus placebo arms (p = 0.41); no grade ≥3 AEs were reported. Geometric mean TFV concentrations increased through day 34 in CVF/rectal fluid and day 59 in plasma, but declined across compartments by day 91. Geometric mean TFV-DP tissue concentrations exceeded the 1000 fmol/mg target through day 56, but fell to 456 fmol/mg at day 91. Among 32 rings returned at the end of the study, 13 had no or low (<0.1 g) residual TFV. Residual TFV did not differ by socio-demographics, sexual activity, Nugent Score or vaginal microbiota. Most participants reported being fully adherent to ring use: 85% and 81% in the TFV and placebo arms, respectively (p = 1.00). A majority of participants reported liking the ring (median 8 on a 10-point Likert scale) and reported a high likelihood of using the ring in the future, if effective (median 9). CONCLUSIONS The 90-day TFV ring was well-tolerated, acceptable and exceeded target cervical tissue concentrations through day 56, but declined thereafter. Additional studies are needed to characterize the higher release from TFV rings in some participants and the optimal duration of use.
Collapse
Affiliation(s)
- Albert Y. Liu
- Bridge HIVSan Francisco Department of Public HealthSan FranciscoCaliforniaUSA
- Department of MedicineUniversity of CaliforniaSan FranciscoSan FranciscoCaliforniaUSA
| | - Holly Gundacker
- Statistical Center for HIV/AIDS Research & PreventionFred Hutchinson Cancer CenterSeattleWashingtonUSA
| | - Barbra Richardson
- Departments of Biostatistics and Global HealthUniversity of WashingtonSeattleWashingtonUSA
- Vaccine and Infectious Disease and Public Health Sciences DivisionsFred Hutchinson Cancer CenterSeattleWashingtonUSA
| | - Beatrice A. Chen
- Department of ObstetricsGynecologyand Reproductive SciencesUniversity of PittsburghPittsburghPennsylvaniaUSA
- Magee‐Womens Research InstitutePittsburghPennsylvaniaUSA
| | - Craig Hoesley
- University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Ariane van der Straten
- Department of MedicineUniversity of CaliforniaSan FranciscoSan FranciscoCaliforniaUSA
- ASTRA ConsultingKensingtonCaliforniaUSA
| | - Amanda Brown
- Statistical Center for HIV/AIDS Research & PreventionFred Hutchinson Cancer CenterSeattleWashingtonUSA
| | - May Beamer
- Magee‐Womens Research InstitutePittsburghPennsylvaniaUSA
| | - Jennifer Robinson
- Division of Gynecology and ObstetricsJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | | | | | - Katherine Bunge
- Department of ObstetricsGynecologyand Reproductive SciencesUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Jill Schwartz
- CONRADEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | | | - Jeanna M. Piper
- Division of AIDSNational Institutes of HealthBethesdaMarylandUSA
| | - Mark A. Marzinke
- Division of Clinical PharmacologyDepartment of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | | |
Collapse
|
19
|
Landovitz RJ, Hanscom BS, Clement ME, Tran HV, Kallas EG, Magnus M, Sued O, Sanchez J, Scott H, Eron JJ, Del Rio C, Fields SD, Marzinke MA, Eshleman SH, Donnell D, Spinelli MA, Kofron RM, Berman R, Piwowar-Manning EM, Richardson PA, Sullivan PA, Lucas JP, Anderson PL, Hendrix CW, Adeyeye A, Rooney JF, Rinehart AR, Cohen MS, McCauley M, Grinsztejn B. Efficacy and safety of long-acting cabotegravir compared with daily oral tenofovir disoproxil fumarate plus emtricitabine to prevent HIV infection in cisgender men and transgender women who have sex with men 1 year after study unblinding: a secondary analysis of the phase 2b and 3 HPTN 083 randomised controlled trial. Lancet HIV 2023; 10:e767-e778. [PMID: 37952550 PMCID: PMC11375758 DOI: 10.1016/s2352-3018(23)00261-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/27/2023] [Accepted: 10/03/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Injectable cabotegravir was superior to daily oral tenofovir disoproxil fumarate plus emtricitabine for HIV prevention in two clinical trials. Both trials had the primary aim of establishing the HIV prevention efficacy of long-acting injectable cabotegravir pre-exposure prophylaxis (PrEP) compared with tenofovir disoproxil fumarate plus emtricitabine daily oral PrEP. Long-acting PrEP was associated with diagnostic delays and integrase strand-transfer inhibitor (INSTI) resistance. This report presents findings from the first unblinded year of the HIV Prevention Trials Network (HPTN) 083 study. METHODS The HPTN 083 randomised controlled trial enrolled HIV-uninfected cisgender men and transgender women at elevated HIV risk who have sex with men, from 43 clinical research sites in Africa, Asia, Latin America, and the USA. Inclusion criteria included: a negative HIV serological test at the screening and study entry, undetectable HIV RNA levels within 14 days of study entry, age 18 years or older, overall good health as determined by clinical and laboratory evaluations, and a creatinine clearance of 60 mL/min or higher. Participants were randomly allocated to receive long-acting injectable cabotegravir or daily oral tenofovir disoproxil fumarate plus emtricitabine PrEP. After study unblinding, participants remained on their original regimen awaiting an extension study. HIV infections were characterised retrospectively at a central laboratory. Here we report the secondary analysis of efficacy and safety for the first unblinded year. The primary outcome was incident HIV infection. Efficacy analyses were done on the modified intention-to-treat population using a Cox regression model. Adverse events were compared across treatment groups and time periods (blinded vs unblinded). This trial is registered with ClinicalTrials.gov, NCT02720094. FINDINGS Of the 4488 participants who contributed person-time to the blinded analysis, 3290 contributed person-time to the first unblinded year analysis between May 15, 2020, and May 14, 2021. Updated HIV incidence in the blinded phase was 0·41 per 100 person-years for long-acting injectable cabotegravir PrEP and 1·29 per 100 person-years for daily oral tenofovir disoproxil fumarate plus emtricitabine PrEP (hazard ratio [HR] 0·31 [95% CI 0·17-0·58], p=0·0003). HIV incidence in the first unblinded year was 0·82 per 100 person-years for long-acting PrEP and 2·27 per 100 person-years for daily oral PrEP (HR 0·35 [0·18-0·69], p=0·002). Adherence to both study products decreased after study unblinding. Additional infections in the long-acting PrEP group included two with on-time injections; three with one or more delayed injections; two detected with long-acting PrEP reinitiation; and 11 more than 6 months after their last injection. Infection within 6 months of cabotegravir exposure was associated with diagnostic delays and INSTI resistance. Adverse events were generally consistent with previous reports; incident hypertension in the long-acting PrEP group requires further investigation. INTERPRETATION Long-acting injectable cabotegravir PrEP retained high efficacy for HIV prevention in men and transgender women who have sex with men during the first year of open-label follow-up, with a near-identical HR for HIV risk reduction between long-acting injectable cabotegravir and daily oral tenofovir disoproxil fumarate plus emtricitabine PrEP during the first year after unblinding compared with the blinded period. Extended follow-up further defined the risk period for diagnostic delays and emergence of INSTI resistance. FUNDING Division of AIDS at the National Institute of Allergy and Infectious Diseases, ViiV Healthcare, and Gilead Sciences.
Collapse
Affiliation(s)
- Raphael J Landovitz
- Center for Clinical AIDS Research and Education, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| | | | | | - Ha V Tran
- Department of Health Behavior, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Esper G Kallas
- Department of Parasitic and Infectious Diseases, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Manya Magnus
- Department of Epidemiology, Milken Institute School of Public Health, George Washington University, Washington, DC, USA
| | - Omar Sued
- Fundación Huésped, Buenos Aires, Argentina
| | - Jorge Sanchez
- Centro de Investigaciones Tecnologicas, Biomedicas y Medioambientales, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Hyman Scott
- San Francisco Department of Public Health, San Francisco, CA, USA
| | - Joe J Eron
- Department of Health Behavior, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Carlos Del Rio
- Emory University School of Medicine and Grady Health System, Atlanta, GA, USA
| | - Sheldon D Fields
- Ross and Carol Nese College of Nursing, Pennsylvania State University, University Park, PA, USA
| | - Mark A Marzinke
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | - Matthew A Spinelli
- Division of HIV, Infectious Diseases, and Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Ryan M Kofron
- Center for Clinical AIDS Research and Education, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Richard Berman
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | | | | | - Peter L Anderson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO, USA
| | - Craig W Hendrix
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Adeola Adeyeye
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | | | | | - Myron S Cohen
- Department of Health Behavior, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Beatriz Grinsztejn
- Instituto Nacional de Infectologia Evandro Chagas-Fiocruz, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Zhang L, Iannuzzi S, Chaturvedula A, Irungu E, Haberer JE, Hendrix CW, von Kleist M. Model-based predictions of protective HIV pre-exposure prophylaxis adherence levels in cisgender women. Nat Med 2023; 29:2753-2762. [PMID: 37957377 PMCID: PMC10667095 DOI: 10.1038/s41591-023-02615-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 09/26/2023] [Indexed: 11/15/2023]
Abstract
Most human immunodeficiency virus (HIV) infections occur in cisgender women in resource-limited settings. In women, self-protection with emtricitabine/tenofovir disoproxil fumarate pre-exposure prophylaxis (FTC/TDF-PrEP) constitutes a major pillar of HIV prevention. However, clinical trials in women had inconsistent outcomes, sparking uncertainty about adherence requirements and reluctance in evaluating on-demand regimens. We analyzed data from published FTC/TDF-PrEP trials to establish efficacy ranges in cisgender women. In a 'bottom-up' approach, we modeled hypotheses in the context of risk-group-specific, adherence-efficacy profiles and challenged those hypotheses with clinical data. We found that different clinical outcomes were related to the proportion of women taking the product, allowing coherent interpretation of the data. Our analysis showed that 90% protection was achieved when women took some product. We found that hypotheses of putative male/female differences were either not impactful or statistically inconsistent with clinical data. We propose that differing clinical outcomes could arise from pill-taking behavior rather than biological factors driving specific adherence requirements in cisgender women.
Collapse
Affiliation(s)
- Lanxin Zhang
- Project group 5 'Systems Medicine of Infectious Diseases', Robert Koch Institute, Berlin, Germany
| | - Sara Iannuzzi
- Project group 5 'Systems Medicine of Infectious Diseases', Robert Koch Institute, Berlin, Germany
- International Max-Planck Research School 'Biology and Computation', Max-Planck Institute for Molecular Genetics, Berlin, Germany
| | - Ayyappa Chaturvedula
- Department of Pharmacotherapy, University of North Texas Health Science Center, Fort Worth, TX, USA
| | | | - Jessica E Haberer
- Center for Global Health, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Craig W Hendrix
- Division of Clinical Pharmacology, Johns Hopkins University, Baltimore, MD, USA
| | - Max von Kleist
- Project group 5 'Systems Medicine of Infectious Diseases', Robert Koch Institute, Berlin, Germany.
- Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
21
|
Moore M, Stansfield S, Donnell DJ, Boily MC, Mitchell KM, Anderson PL, Delany-Moretlwe S, Bekker LG, Mgodi NM, Celum CL, Dimitrov D. Efficacy estimates of oral pre-exposure prophylaxis for HIV prevention in cisgender women with partial adherence. Nat Med 2023; 29:2748-2752. [PMID: 37798438 PMCID: PMC11386520 DOI: 10.1038/s41591-023-02564-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 08/23/2023] [Indexed: 10/07/2023]
Abstract
Pre-exposure prophylaxis (PrEP) with tenofovir (TFV) disoproxil fumarate and emtricitabine administered orally daily is effective in preventing human immunodeficiency virus (HIV) acquisition in both men and women with sufficient adherence; however, the adherence-efficacy relationship in cisgender women has not been well established. We calculated the adherence-efficacy curve for cisgender women by using HIV incidence and plasma TFV concentration data from three trials (FEM-PrEP, VOICE and Partners PrEP). We imputed TFV diphosphate (TFV-DP) concentrations, a measure of long-term adherence, from TFV quantification by using data from the HIV Prevention Trials Network 082 study, which measured both TFV-DP and TFV concentrations. Two, four and seven pills per week reduced HIV incidence by 59.3% (95% credible interval (CrI) 29.9-95.8%), 83.8% (95% CI 51.7-99.8%) and 95.9% (95% CI 72.6-100%), respectively. Our adherence-efficacy curve can be validated and updated by HIV prevention studies that directly measure TFV-DP concentrations. The curve suggests that high adherence confers high protection in cisgender women. However, the lower efficacy with partial adherence highlights the need for new PrEP products and interventions to increase adherence.
Collapse
Affiliation(s)
- Mia Moore
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- HPTN Modelling Centre, Imperial College London, London, UK.
| | - Sarah Stansfield
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- HPTN Modelling Centre, Imperial College London, London, UK
| | - Deborah J Donnell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Marie-Claude Boily
- HPTN Modelling Centre, Imperial College London, London, UK
- Medical Research Council Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
| | - Kate M Mitchell
- HPTN Modelling Centre, Imperial College London, London, UK
- Medical Research Council Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
| | - Peter L Anderson
- Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | | | - Linda-Gail Bekker
- Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - Nyaradzo M Mgodi
- College of Health Sciences Clinical Trials Research Centre, University of Zimbabwe, Harare, Zimbabwe
| | - Connie L Celum
- Departments of Global Health, Medicine and Epidemiology, University of Washington, Seattle, WA, USA
| | - Dobromir Dimitrov
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- HPTN Modelling Centre, Imperial College London, London, UK
- Department of Applied Mathematics, University of Washington, Seattle, WA, USA
| |
Collapse
|
22
|
Scott RK, Yu Y, Marzinke MA, Coleman JS, Hendrix CW, Bies R. Clinical trial simulation to evaluate tenofovir disoproxil fumarate/emtricitabine HIV pre-exposure prophylaxis dosing during pregnancy. FRONTIERS IN REPRODUCTIVE HEALTH 2023; 5:1224580. [PMID: 37830105 PMCID: PMC10565828 DOI: 10.3389/frph.2023.1224580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 09/04/2023] [Indexed: 10/14/2023] Open
Abstract
Objective To evaluate upward-adjustment of tenofovir disoproxil fumarate (TDF)/emtricitabine (FTC) pre-exposure prophylaxis (PrEP) dosing during pregnancy in order to maintain target plasma concentrations associated with HIV protection. Design Population pharmacokinetic (PK) modeling and clinical trial simulation (CTS). Material and methods We developed population pharmacokinetic models for TFV and FTC using data from the Partners Demonstration Project and a PK study of TDF/FTC among cisgender women by Coleman et al., and performed an in-silico simulation. Pregnancy-trimester was identified as a significant covariate on apparent clearance in the optimized final model. We simulated 1,000 pregnant individuals starting standard daily oral TDF/FTC (300 mg/200 mg) prior to pregnancy. Upon becoming pregnant, simulated patients were split into two study arms: one continuing standard-dose and the other receiving double standard-dose throughout pregnancy. Results Standard-dose trough TFV concentrations were significantly lower in pregnancy compared to pre-pregnancy, with 34.0%, 43.8%, and 65.1% of trough plasma concentrations below the lower bound of expected trough concentrations presumed to be the protective threshold in the 1st, 2nd, and 3rd trimesters, respectively. By comparison, in the simulated double-dose group, 10.7%, 14.4%, and 27.8% of trough concentrations fell below the estimated protective thresholds in the 1st, 2nd, and 3rd trimesters, respectively. The FTC trough plasma concentration during pregnancy was also lower than pre-pregnancy, with 45.2% of the steady-state trough concentrations below the estimated protective trough concentrations of FTC. In the pregnancy-adjusted double-dose group, 24.1% of trough plasma concentrations were lower than protective levels. Conclusions Our simulation shows >50% of research participants on standard dosing would have 3rd trimester trough plasma TFV concentrations below levels associated with protection. This simulation provides the quantitative basis for the design of prospective TDF/FTC studies during pregnancy to evaluate the safety and appropriateness of pregnancy-adjusted dosing.
Collapse
Affiliation(s)
- Rachel K Scott
- Women's Health Research, MedStar Health Research Institute, Washington, DC, United States
| | - Yifan Yu
- Department of Pharmaceutical Sciences, University of Buffalo, Buffalo, NY, United States
| | - Mark A Marzinke
- Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jenell S Coleman
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Craig W Hendrix
- Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Robert Bies
- Department of Pharmaceutical Sciences, University of Buffalo, Buffalo, NY, United States
| |
Collapse
|
23
|
Srinivasula S, Degrange P, Perazzolo S, Bonvillain A, Tobery A, Kaplan J, Jang H, Turnier R, Davies M, Cottrell M, Ho RJY, Di Mascio M. Viral dissemination and immune activation modulate antiretroviral drug levels in lymph nodes of SIV-infected rhesus macaques. Front Immunol 2023; 14:1213455. [PMID: 37790938 PMCID: PMC10544331 DOI: 10.3389/fimmu.2023.1213455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 09/01/2023] [Indexed: 10/05/2023] Open
Abstract
Introduction and methods To understand the relationship between immunovirological factors and antiretroviral (ARV) drug levels in lymph nodes (LN) in HIV therapy, we analyzed drug levels in twenty-one SIV-infected rhesus macaques subcutaneously treated with daily tenofovir (TFV) and emtricitabine (FTC) for three months. Results The intracellular active drug-metabolite (IADM) levels (TFV-dp and FTC-tp) in lymph node mononuclear cells (LNMC) were significantly lower than in peripheral blood mononuclear cells (PBMC) (P≤0.005). Between Month 1 and Month 3, IADM levels increased in both LNMC (P≤0.001) and PBMC (P≤0.01), with a steeper increase in LNMC (P≤0.01). The viral dissemination in plasma, LN, and rectal tissue at ART initiation correlated negatively with IADM levels at Month 1. Physiologically-based pharmacokinetic model simulations suggest that, following subcutaneous ARV administration, ART-induced reduction of immune activation improves the formation of active drug-metabolites through modulation of kinase activity and/or through improved parent drug accessibility to LN cellular compartments. Conclusion These observations have broad implications for drugs that need to phosphorylate to exert their pharmacological activity, especially in the settings of the pre-/post-exposure prophylaxis and efficacy of antiviral therapies targeting pathogenic viruses such as HIV or SARS-CoV-2 replicating in highly inflammatory anatomic compartments.
Collapse
Affiliation(s)
- Sharat Srinivasula
- AIDS Imaging Research Section, Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Paula Degrange
- AIDS Imaging Research Section, Charles River Laboratories, Integrated Research Facility, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, MD, United States
| | - Simone Perazzolo
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States
| | - Andrew Bonvillain
- AIDS Imaging Research Section, Charles River Laboratories, Integrated Research Facility, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, MD, United States
| | - Amanda Tobery
- AIDS Imaging Research Section, Charles River Laboratories, Integrated Research Facility, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, MD, United States
| | - Jacob Kaplan
- AIDS Imaging Research Section, Division of Clinical Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Poolesville, MD, United States
| | - Hyukjin Jang
- AIDS Imaging Research Section, Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Refika Turnier
- Clinical Support Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Michael Davies
- Clinical Support Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Mackenzie Cottrell
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC, United States
| | - Rodney J. Y. Ho
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Michele Di Mascio
- AIDS Imaging Research Section, Division of Clinical Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Poolesville, MD, United States
| |
Collapse
|
24
|
Ramadhani HO, Crowell TA, Nowak RG, Adebajo S, Kayode BO, Ononaku U, Baral SD, Ndembi N, Charurat ME. Determinants of Preexposure Prophylaxis Cascade Among Sexual and Gender Minorities in Nigeria. Sex Transm Dis 2023; 50:559-566. [PMID: 37277897 PMCID: PMC10527664 DOI: 10.1097/olq.0000000000001841] [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: 06/07/2023]
Abstract
BACKGROUND Preexposure prophylaxis (PrEP) is effective in preventing human immunodeficiency virus (HIV) infection among sexual and gender minorities (SGMs). We evaluated the characteristics associated with engagement in 7 steps of the PrEP cascade among SGMs in Nigeria. METHODS Sexual and gender minorities without HIV from the Abuja site of TRUST/RV368 cohort who were surveyed on awareness of and willingness to use PrEP were approached for PrEP initiation upon availability of oral daily PrEP. To understand gaps in the uptake of oral daily PrEP, we categorized the HIV PrEP cascade as (i) education about PrEP, (ii) interest in PrEP, (iii) successful contact, (iv) appointment scheduled, (v) appointment attendance, (vi) PrEP initiation, and (vii) plasma protective levels of tenofovir disoproxil fumarate. Multivariable logistic regression models were used to determine factors associated with each of the 7 steps in the HIV PrEP cascade. RESULTS Of 788 participants, 718 (91.1%) showed interest in taking oral daily PrEP every day and/or after a sexual act, 542 (68.8%) were successfully contacted, 433 (54.9%) scheduled an appointment, 409 (51.9%) attended a scheduled appointment, 400 (50.8%) initiated oral daily PrEP, and 59 (7.4%) had protective levels of tenofovir disoproxil fumarate. Of initiators of PrEP, 23 (5.8%) seroconverted at a rate of 13.9 cases/100 person-years. Better social support, larger network density, and higher education were associated with engagement in 4 to 5 components of the cascade. CONCLUSIONS Our data highlight a gap between willingness and actual PrEP use. Despite PrEP's effectiveness in preventing HIV, the optimal impact of PrEP for SGMs in sub-Saharan Africa will require multifaceted approaches that combine social support, education, and destigmatization.
Collapse
Affiliation(s)
| | | | - Rebecca G Nowak
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore
| | - Sylvia Adebajo
- Center for International Health Education and Biosecurity, Maryland Global Initiatives Corporation (MGIC)-an affiliate of the University of Maryland Baltimore
| | - Blessing O Kayode
- Institute of Human Virology Nigeria, Abuja, Federal Capital Territory, Nigeria
| | - Uchenna Ononaku
- Institute of Human Virology Nigeria, Abuja, Federal Capital Territory, Nigeria
| | | | | | | |
Collapse
|
25
|
Rosen EP, White N, Gilliland WM, Gerona RR, Gandhi M, Amico KR, Mayer KH, Gulick RM, Kashuba ADM. Mass spectrometry imaging of hair identifies daily maraviroc adherence in HPTN 069/ACTG A5305. PLoS One 2023; 18:e0287449. [PMID: 37352285 PMCID: PMC10289441 DOI: 10.1371/journal.pone.0287449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 06/06/2023] [Indexed: 06/25/2023] Open
Abstract
Objective measures of adherence for antiretrovirals used as pre-exposure prophylaxis (PrEP) are critical for improving preventative efficacy in both clinical trials and real-world application. Current objective adherence measures either reflect only recent behavior (eg days for plasma or urine) or cumulative behavior (eg months for dried blood spots). Here, we measured the accumulation of the antiretroviral drug maraviroc (MVC) in hair strands by infrared matrix-assisted laser desorption electrospray ionization (IR-MALDESI) mass spectrometry imaging (MSI) to evaluate adherence behavior longitudinally at high temporal resolution. 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. We then used the benchmarked MSI assay to classify adherence to MVC-based PrEP regimens in hair samples collected throughout the 48-week HPTN069/ACTGA5305 study. We found that only ~32% of investigated hair samples collected during the study's active dosing period showed consistent daily PrEP adherence throughout a retrospective period of 30 days, and also found that profiles of daily individual adherence from MSI hair analysis could identify when patients were and were not taking study drug. The assessment of adherence from MSI hair strand analysis was 62% lower than adherence classified using paired plasma samples, the latter of which may be influenced by white-coat adherence. These findings demonstrate the ability of MSI hair analysis to examine daily variability of adherence behavior over a longer-term measurement and offer the potential for longitudinal comparison with risk behavior to target patient-specific adherence interventions and improve outcomes.
Collapse
Affiliation(s)
- Elias P. Rosen
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Nicole White
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - William M. Gilliland
- Department of Chemistry, Furman University, Greenville, South Carolina, United States of America
| | - Roy R. Gerona
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Monica Gandhi
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - K. Rivet Amico
- School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kenneth H. Mayer
- Fenway Health, Department of Medicine, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Roy M. Gulick
- Weill Cornell Medicine, New York, New York, United States of America
| | - Angela D. M. Kashuba
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill at Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
26
|
Donnell D, Gao F, Hughes JP, Hanscom B, Corey L, Cohen MS, Edupuganti S, Mgodi N, Rees H, Baeten JM, Gray G, Bekker L, Hosseinipour M, Delany‐Moretlwe S. Counterfactual estimation of efficacy against placebo for novel PrEP agents using external trial data: example of injectable cabotegravir and oral PrEP in women. J Int AIDS Soc 2023; 26:e26118. [PMID: 37363917 PMCID: PMC10292682 DOI: 10.1002/jia2.26118] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 05/12/2023] [Indexed: 06/28/2023] Open
Abstract
INTRODUCTION Multiple antiretroviral agents have demonstrated efficacy for human immunodeficiency virus (HIV) pre-exposure prophylaxis (PrEP). As a result, clinical trials of novel agents have transitioned from placebo- to active-controlled designs; however, active-controlled trials do not provide an estimate of efficacy versus no use of PrEP. Counterfactual placebo comparisons using other data sources could be employed to provide this information. METHODS We compared the active-controlled study (HPTN 084) of injectable cabotegravir (CAB-LA) versus daily oral emtricitabine/tenofovir disoproxil fumarate (FTC/TDF) among women from seven countries in Africa to three external, contemporaneous randomized HIV prevention trials from which we constructed counterfactual placebo estimates. We used direct standardization via analysis weights to achieve the same distribution of person-years between the external study and HPTN 084, across strata predictive of HIV risk (country and selected risk covariates). We estimated prevention efficacy against a counterfactual placebo to provide information on the use of CAB-LA and FTC/TDF compared to no intervention. We compared the counterfactual placebo findings for FTC/TDF to previous placebo-controlled trials, adjusted for observed adherence to daily pills. RESULTS Distribution of age and baseline prevalence of gonorrhoea and chlamydia were similar among matched counterfactual placebo and observed HPTN 084 arms after standardization. Counterfactual estimates of CAB-LA versus placebo in all three settings showed a consistent risk reduction of 93%-94%, with lower bounds of the confidence intervals above 72%. Observed adherence (quantifiable tenofovir in plasma) in HPTN 084 was 54%-56%, and estimated efficacy of daily oral FTC/TDF against a counterfactual placebo was consistent with a predicted risk reduction of 39%-40% for this level of daily pill use. CONCLUSIONS Counterfactual placebo rates of HIV acquisition derived from external trial data in similar locations and time can be used to support estimates of placebo-based efficacy of a novel HIV prevention agent. External trial data must be standardized to be representative of the clinical trial cohort testing the novel HIV prevention agent, accounting for confounders.
Collapse
Affiliation(s)
| | - Fei Gao
- Fred Hutchinson Cancer CenterSeattleWashingtonUSA
| | | | | | | | - Myron S. Cohen
- University of North CarolinaChapel HillNorth CarolinaUSA
| | | | - Nyaradzo Mgodi
- University of Zimbabwe Clinical Trials Research CentreHarareZimbabwe
| | | | | | - Glenda Gray
- South Africa Medical Research CouncilTygerbergSouth Africa
| | | | | | | |
Collapse
|
27
|
Zhang L, Iannuzzi S, Chaturvedula A, Haberer JE, Hendrix CW, von Kleist M. Synthesis of protective oral PrEP adherence levels in cisgender women using convergent clinical- and bottom-up modeling. RESEARCH SQUARE 2023:rs.3.rs-2772765. [PMID: 37131701 PMCID: PMC10153398 DOI: 10.21203/rs.3.rs-2772765/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Globally, most HIV infections occur in heterosexual women in resource-limited settings. In these settings, female self-protection with generic emtricitabine/tenofovir disoproxil fumarate pre-exposure prophylaxis (FTC/TDF-PrEP) may constitute a major pillar of the HIV prevention portfolio. However, clinical trials in women had inconsistent outcomes, sparking uncertainty regarding risk-group specific adherence requirements and causing reluctance in testing and recommending on-demand regimen in women. We analyzed all FTC/TDF-PrEP trials to establish PrEP efficacy ranges in women. In a 'bottom-up' approach, we modeled hypotheses corroborating risk-group specific adherence-efficacy profiles. Finally, we used the clinical efficacy ranges to (in-)validate hypotheses. We found that different clinical outcomes could solely be explained by the proportion of enrolled participants not taking the product, allowing, for the first time, to unify clinical observations. This analysis showed that 90% protection was achieved, when women took some of the product. Using 'bottom-up' modelling, we found that hypotheses of putative male/female differences were either irrelevant, or statistically inconsistent with clinical data. Furthermore, our multiscale modelling indicated that 90% protection was achieved if oral FTC/TDF was taken at least twice weekly.
Collapse
Affiliation(s)
- Lanxin Zhang
- Project group 5 “Systems Medicine of Infectious Disease”, Robert Koch Institute, Berlin, Germany
| | - Sara Iannuzzi
- Project group 5 “Systems Medicine of Infectious Disease”, Robert Koch Institute, Berlin, Germany
- International Max-Planck Research School “Biology and Computation” (IMPRS-BAC), Max-Planck Institute for Molecular Genetics, Berlin, Germany
| | - Ayyappa Chaturvedula
- The University of North Texas; Health Science Center, Fort Worth, United States
- Pumas-AI Inc
| | - Jessica E. Haberer
- Center for Global Health, Massachusetts General Hospital, Boston, United States
- Department of Medicine, Harvard Medical School, Boston, United States
| | - Craig W. Hendrix
- Division of Clinical Pharmacology, Johns-Hopkins University, Baltimore, United States
| | - Max von Kleist
- Project group 5 “Systems Medicine of Infectious Disease”, Robert Koch Institute, Berlin, Germany
- Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
28
|
Marzinke MA, Fogel JM, Wang Z, Piwowar-Manning E, Kofron R, Moser A, Bhandari P, Gollings R, Bushman LR, Weng L, Halvas EK, Mellors J, Anderson PL, Persaud D, Hendrix CW, McCauley M, Rinehart AR, St Clair M, Ford SL, Rooney JF, Adeyeye A, Chariyalertsak S, Mayer K, Arduino RC, Cohen MS, Grinsztejn B, Hanscom B, Landovitz RJ, Eshleman SH. Extended Analysis of HIV Infection in Cisgender Men and Transgender Women Who Have Sex with Men Receiving Injectable Cabotegravir for HIV Prevention: HPTN 083. Antimicrob Agents Chemother 2023; 67:e0005323. [PMID: 36995219 PMCID: PMC10112247 DOI: 10.1128/aac.00053-23] [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: 01/13/2023] [Accepted: 02/15/2023] [Indexed: 03/31/2023] Open
Abstract
HPTN 083 demonstrated that injectable cabotegravir (CAB) was superior to oral tenofovir disoproxil fumarate-emtricitabine (TDF-FTC) for HIV prevention in cisgender men and transgender women who have sex with men. We previously analyzed 58 infections in the blinded phase of HPTN 083 (16 in the CAB arm and 42 in the TDF-FTC arm). This report describes 52 additional infections that occurred up to 1 year after study unblinding (18 in the CAB arm and 34 in the TDF-FTC arm). Retrospective testing included HIV testing, viral load testing, quantification of study drug concentrations, and drug resistance testing. The new CAB arm infections included 7 with CAB administration within 6 months of the first HIV-positive visit (2 with on-time injections, 3 with ≥1 delayed injection, and 2 who restarted CAB) and 11 with no recent CAB administration. Three cases had integrase strand transfer inhibitor (INSTI) resistance (2 with on-time injections and 1 who restarted CAB). Among 34 CAB infections analyzed to date, diagnosis delays and INSTI resistance were significantly more common in infections with CAB administration within 6 months of the first HIV-positive visit. This report further characterizes HIV infections in persons receiving CAB preexposure prophylaxis and helps define the impact of CAB on the detection of infection and the emergence of INSTI resistance.
Collapse
Affiliation(s)
- Mark A. Marzinke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jessica M. Fogel
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zhe Wang
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Estelle Piwowar-Manning
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ryan Kofron
- Department of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Amber Moser
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Pradip Bhandari
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ryann Gollings
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lane R. Bushman
- Department of Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Lei Weng
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Elias K. Halvas
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John Mellors
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Peter L. Anderson
- Department of Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Deborah Persaud
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Craig W. Hendrix
- Department of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | - Marty St Clair
- ViiV Healthcare, Research Triangle Park, North Carolina, USA
| | - Susan L. Ford
- GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | | | - Adeola Adeyeye
- Prevention Science Program, Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Suwat Chariyalertsak
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Kenneth Mayer
- The Fenway Institute, Fenway Health, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Roberto C. Arduino
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Myron S. Cohen
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Beatriz Grinsztejn
- Instituto de Pesquisa Clinica Evandro Chagas-Fiocruz, Rio de Janeiro, Brazil
| | - Brett Hanscom
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Raphael J. Landovitz
- Center for Clinical AIDS Research and Education, University of California, Los Angeles, Los Angeles, California, USA
| | - Susan H. Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
29
|
Devanathan AS, Dumond JB, Anderson DJC, Moody K, Poliseno AJ, Schauer AP, Sykes C, Gay CL, Rosen EP, Kashuba ADM, Cottrell ML. A Novel Algorithm to Improve PrEP Adherence Monitoring Using Dried Blood Spots. Clin Pharmacol Ther 2023; 113:896-903. [PMID: 36622798 PMCID: PMC10023501 DOI: 10.1002/cpt.2845] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/03/2023] [Indexed: 01/10/2023]
Abstract
Tenofovir diphosphate (TFVdp; an active metabolite of oral HIV pre-exposure prophylaxis (PrEP)) is measured in dried blood spots (DBS) to estimate adherence. However, TFVdp's long half-life in whole blood may lead to misclassification following a recent change in adherence. PrEP's other metabolite, emtricitabine triphosphate (FTCtp), has a shorter half-life in whole blood but adherence thresholds are undefined. We characterized DBS TFVdp and FTCtp concentrations across many dosing scenarios. Population pharmacokinetic models were fit to TFVdp and FTCtp DBS concentrations from a directly observed therapy study (NCT03218592). Concentrations were simulated for 90 days of daily dosing followed by 90 days of 1 to 7 doses/week and for event-driven PrEP (edPrEP) scenarios. Thresholds of 1,000 and 200 fmol/punch, for TFVdp and FTCtp, respectively, were reflective of taking 4 doses/week (a minimum target for effective PrEP in men). TFVdp was < 1,000 fmol/punch for 17 days after initiating daily PrEP and > 1,000 fmol/punch for 62 days after decreasing to 3 doses/week. Respectively, FTCtp was < 200 fmol/punch for 4 days and > 200 fmol/punch for 6 days. Accuracy of edPrEP adherence classification depended on duration between last sex act and DBS sampling for both measures with misclassification ranging from 9-100%. These data demonstrate adherence misclassification by DBS TFVdp for 2 months following a decline in adherence, elucidating the need for FTCtp to estimate recent adherence. We provide proof of principle that individualized interpretation is needed to support edPrEP adherence monitoring. Our collective approach facilitates clinicians' ability to interpret DBS results and administer patient-centric interventions.
Collapse
Affiliation(s)
- Aaron S. Devanathan
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC, USA
| | - Julie B. Dumond
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC, USA
| | - Daijha JC Anderson
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC, USA
| | - Kristen Moody
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC, USA
| | - Amanda J. Poliseno
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC, USA
| | - Amanda P. Schauer
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC, USA
| | - Craig Sykes
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC, USA
| | - Cynthia L. Gay
- University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Elias P. Rosen
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC, USA
| | - Angela DM Kashuba
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC, USA
- University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | | |
Collapse
|
30
|
Stansfield SE, Moore M, Boily MC, Hughes JP, Donnell DJ, Dimitrov DT. Estimating benefits of using on-demand oral prep by MSM: A comparative modeling study of the US and Thailand. EClinicalMedicine 2023; 56:101776. [PMID: 36618897 PMCID: PMC9813675 DOI: 10.1016/j.eclinm.2022.101776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/18/2022] [Accepted: 11/18/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Daily and on-demand pre-exposure prophylaxis (PrEP) are effective at preventing HIV acquisition among men who have sex with men (MSM), but only daily PrEP is approved in the US. On-demand PrEP may improve uptake and adherence. We identify sub-groups of MSM who would benefit from on-demand PrEP and determine effectiveness achieved if individuals used their optimal regimens. METHODS Using data from the HPTN 067 study (study period 2012-2014), we created an individual-based stochastic model of HIV risk in two synthetic MSM populations with parameters separately estimated using data from Harlem, US, and Bangkok, Thailand. Agents were assigned daily and on-demand PrEP for six months each. Two personalized PrEP assignments: optimal, based on improved predicted effectiveness and reduced pill burden, and adherence-based, using daily PrEP adherence, were simulated for another six months. FINDINGS Simulated on-demand PrEP was optimal for approximately one-third of MSM. It was assigned mainly to those with low daily PrEP adherence (88% (Harlem), 95% (Bangkok) of MSM with daily PrEP adherence <40%). Mean effectiveness was slightly higher in the full synthetic population with optimal PrEP assignment compared to universal daily PrEP. Among MSM for whom on-demand PrEP was optimal, mean effectiveness improved by 18 (Harlem) and 7 percentage points (Bangkok). Comparable predicted effectiveness was achieved if on-demand PrEP was assigned to the population with daily PrEP adherence <50%. There was no advantage in assigning on-demand PrEP by sex act frequency. INTERPRETATION On-demand PrEP could benefit many MSM by increasing effectiveness or decreasing pill burden with similar effectiveness. On-demand PrEP may be an effective alternative to daily PrEP for individuals with difficulty taking daily PrEP consistently. Results were similar for Harlem and Bangkok, indicating that these conclusions were robust in populations with different overall adherence levels and may inform future public-health policies. FUNDING US NIH grant UM1 AI068617.
Collapse
Affiliation(s)
- Sarah E. Stansfield
- Fred Hutchinson Cancer Center, Seattle, WA, United States
- Corresponding author. Vaccine & Infectious Disease Division, Fred Hutchinson Cancer Center, United States.
| | - Mia Moore
- Fred Hutchinson Cancer Center, Seattle, WA, United States
| | | | | | | | | |
Collapse
|
31
|
Matthews LT, Atukunda EC, Owembabazi M, Kalyebera KP, Psaros C, Chitneni P, Hendrix CW, Marzinke MA, Anderson PL, Isehunwa OO, Hurwitz KE, Bennett K, Muyindike W, Bangsberg DR, Haberer JE, Marrazzo JM, Bwana MB. High PrEP uptake and objective longitudinal adherence among HIV-exposed women with personal or partner plans for pregnancy in rural Uganda: A cohort study. PLoS Med 2023; 20:e1004088. [PMID: 36795763 PMCID: PMC9983833 DOI: 10.1371/journal.pmed.1004088] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 03/03/2023] [Accepted: 01/23/2023] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND In Uganda, fertility rates and adult HIV prevalence are high, and many women conceive with partners living with HIV. Pre-exposure prophylaxis (PrEP) reduces HIV acquisition for women and, therefore, infants. We developed the Healthy Families-PrEP intervention to support PrEP use as part of HIV prevention during periconception and pregnancy periods. We conducted a longitudinal cohort study to evaluate oral PrEP use among women participating in the intervention. METHODS AND FINDINGS We enrolled HIV-negative women with plans for pregnancy with a partner living, or thought to be living, with HIV (2017 to 2020) to evaluate PrEP use among women participating in the Healthy Families-PrEP intervention. Quarterly study visits through 9 months included HIV and pregnancy testing and HIV prevention counseling. PrEP was provided in electronic pillboxes, providing the primary adherence measure ("high" adherence when pillbox was opened ≥80% of days). Enrollment questionnaires assessed factors associated with PrEP use. Plasma tenofovir (TFV) and intraerythrocytic TFV-diphosphate (TFV-DP) concentrations were determined quarterly for women who acquired HIV and a randomly selected subset of those who did not; concentrations TFV ≥40 ng/mL and TFV-DP ≥600 fmol/punch were categorized as "high." Women who became pregnant were initially exited from the cohort by design; from March 2019, women with incident pregnancy remained in the study with quarterly follow-up until pregnancy outcome. Primary outcomes included (1) PrEP uptake (proportion who initiated PrEP); and (2) PrEP adherence (proportion of days with pillbox openings during the first 3 months following PrEP initiation). We used univariable and multivariable-adjusted linear regression to evaluate baseline predictors selected based on our conceptual framework of mean adherence over 3 months. We also assessed mean monthly adherence over 9 months of follow-up and during pregnancy. We enrolled 131 women with mean age 28.7 years (95% CI: 27.8 to 29.5). Ninety-seven (74%) reported a partner with HIV and 79 (60%) reported condomless sex. Most women (N = 118; 90%) initiated PrEP. Mean electronic adherence during the 3 months following initiation was 87% (95% CI: 83%, 90%). No covariates were associated with 3-month pill-taking behavior. Concentrations of plasma TFV and TFV-DP were high among 66% and 47%, 56% and 41%, and 45% and 45% at months 3, 6, and 9, respectively. We observed 53 pregnancies among 131 women (1-year cumulative incidence 53% [95% CI: 43%, 62%]) and 1 HIV-seroconversion in a non-pregnant woman. Mean pillcap adherence for PrEP users with pregnancy follow-up (N = 17) was 98% (95% CI: 97%, 99%). Study design limitations include lack of a control group. CONCLUSIONS Women in Uganda with PrEP indications and planning for pregnancy chose to use PrEP. By electronic pillcap, most were able to sustain high adherence to daily oral PrEP prior to and during pregnancy. Differences in adherence measures highlight challenges with adherence assessment; serial measures of TFV-DP in whole blood suggest 41% to 47% of women took sufficient periconception PrEP to prevent HIV. These data suggest that women planning for and with pregnancy should be prioritized for PrEP implementation, particularly in settings with high fertility rates and generalized HIV epidemics. Future iterations of this work should compare the outcomes to current standard of care. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT03832530 https://clinicaltrials.gov/ct2/show/NCT03832530?term=lynn+matthews&cond=hiv&cntry=UG&draw=2&rank=1.
Collapse
Affiliation(s)
- Lynn T. Matthews
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| | | | | | - Kato Paul Kalyebera
- Mbarara University of Science and Technology, Mbarara, Uganda
- Mbarara Regional Referral Hospital, Mbarara, Uganda
| | - Christina Psaros
- Behavioral Medicine Program, Department of Psychiatry, Massachusetts General Hospital, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Pooja Chitneni
- Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Infectious Diseases and General Internal Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Craig W. Hendrix
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Mark A. Marzinke
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Peter L. Anderson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, United States of America
| | - Oluwaseyi O. Isehunwa
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Kathleen E. Hurwitz
- NoviSci, Inc., a Target RWE company, Durham, North Carolina, United States of America
| | - Kara Bennett
- Bennett Statistical Consulting Inc., Ballston Lake, New York, United States of America
| | - Winnie Muyindike
- Mbarara University of Science and Technology, Mbarara, Uganda
- Mbarara Regional Referral Hospital, Mbarara, Uganda
| | - David R. Bangsberg
- School of Public Health, Oregon Health Sciences University–Portland State University, Portland, Oregon, United States of America
| | - Jessica E. Haberer
- Harvard Medical School, Boston, Massachusetts, United States of America
- Center for Global Health, Massachusetts General Hospital, Boston, Boston, Massachusetts, United States of America
| | - Jeanne M. Marrazzo
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | | |
Collapse
|
32
|
Andrade A, Fuchs EJ, Marzinke MA, Abdul Massih S, Breakey J, Beselman S, McNicholl I, Hendrix CW. EVG/COBI/FTC/TAF Bioequivalence Comparing Whole Tablets with Tablets Dissolved in Tap Water. AIDS Res Hum Retroviruses 2023; 39:38-43. [PMID: 36301928 PMCID: PMC9910106 DOI: 10.1089/aid.2022.0085] [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] [Indexed: 02/04/2023] Open
Abstract
Medication adherence can be challenging for persons with difficulty swallowing tablets. We investigated the bioequivalence of a dissolved tablet when compared with that of a whole tablet of the fixed-dose combination elvitegravir (EVG)/cobicistat (COBI)/emtricitabine (FTC)/tenofovir (TFV) alafenamide fumarate (TAF). A within-subject fixed-order two-period open-label study was conducted in 12 HIV-negative research participants after obtaining informed consent. Participants took a single dose each of the whole tablet and dissolved tablet under direct observation, separated by a 14-day washout period. The dissolved tablet was prepared by adding one whole EVG/COBI/FTC/TAF tablet to 120 mL tap water and stirring. Both dosage types were taken with a standardized meal. Plasma samples were obtained for 72 h postdose. Plasma EVG, FTC, TAF, and TFV were analyzed with liquid chromatographic-tandem mass spectrometric methods. Peak plasma concentration (Cmax) and the area under the concentration-time curve extrapolated to infinity (AUC0-∞) were estimated using WinNonlin software (v.8.3). The primary outcome was bioequivalence consistent with FDA guidance using the 90% confidence interval or the geometric mean ratio. Of 12 participants, 7 were black (58%) and 5 were white (42%), 4 were women (33%), 8 were men (67%), and the mean age was 43.6 years (23-54). There were no complaints about taste with the dissolved tablet. Bioequivalence was established only for FTC. EVG Cmax and AUC0-∞ were higher by 18% and 12%, respectively, when taking the dissolved compared with the whole tablet. TAF AUC0-∞ and Cmax were both 8% lower, whereas TFV Cmax and AUC0-∞ were 8% and 5% lower, respectively, when taken after dissolution. EVG/COBI/FTC/TAF dissolved rapidly in water and had no unpleasant taste. Increases in EVG and decreases in TAF and TFV concentrations were observed when taking dissolved EVG/COBI/FTC/TAF. These changes were judged to be clinically insignificant. Dissolving EVG/COBI/FTC/TAF in water may be suitable for those with pill swallowing challenges. The trial was registered on (//clinicaltrials.gov NCT03717129).
Collapse
Affiliation(s)
| | - Edward J. Fuchs
- Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mark A. Marzinke
- Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sandra Abdul Massih
- Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jennifer Breakey
- Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sasha Beselman
- Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Craig W. Hendrix
- Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
33
|
Boyd DT, Nelson LE, Hill MJ, Whitfield D, Ramos SR, Akyirem S, Emel L, Wilton L, Hightow-Weidman L, Shoptaw S, Magnus M, Mayer KH, Piwowar-Manning E, Wallace SE, Fields SD, Wheeler DP. Examining the Role of Autonomy Support, Goal Setting, and Care Coordination Quality on HIV PrEP Adherence in Black Men Who Have Sex with Men: HPTN 073. AIDS Patient Care STDS 2023; 37:22-30. [PMID: 36626154 PMCID: PMC10024067 DOI: 10.1089/apc.2022.0139] [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: 01/11/2023] Open
Abstract
Autonomy support is a concept that is derived from self-determination theory. Autonomy refers to the freedom to act as one chooses. The current study aimed to examine if autonomy support was associated with dried blood spot validated pre-exposure prophylaxis (PrEP) adherence, and whether the association was mediated by PrEP adherence goal setting and progress toward PrEP adherence goals. Our sample was drawn from Black men who have sex with men (MSM) from across three cities (Chapel Hill, NC; Los Angeles, CA; and Washington, DC) in the United States between February 2013 and September 2014. We used logistic regression to evaluate associations between study variables and path analysis to test mediation effects. Participants were, on average, 28 [standard deviation (SD) = 1.12] years old and 25% were unemployed. We found that MSM who experienced high autonomy support were more likely to adhere to PrEP [odds ratio (OR) = 1.17; 95% confidence interval: 1.00-1.38]. MSM who set PrEP adherence goals were more likely to adhere to PrEP. Moreover, MSM who reported making progress toward their goals were also more likely to adhere to PrEP. Finally, client perception of coordination quality enhanced the magnitude of the association between goal setting and goal progress and the effect size of goal progress on PrEP adherence. Autonomy support, goal setting, goal monitoring/evaluation, and care coordination quality influenced PrEP adherence among Black MSM. Our findings indicate that while it is important to set goals for PrEP adherence, goal setting may need to be accompanied by progress monitoring to achieve the maximal effect.
Collapse
Affiliation(s)
- Donte T. Boyd
- College of Social Work, The Ohio State University, Columbus, Ohio, USA
- Yale School of Public Health, Center for Interdisciplinary Research on AIDS, New Haven, Connecticut, USA
| | - LaRon E. Nelson
- Yale School of Public Health, Center for Interdisciplinary Research on AIDS, New Haven, Connecticut, USA
- Yale School of Nursing, New Haven, Connecticut, USA
- Unity Health Toronto—St. Michael's, Li Ka Shing Knowledge Institute, MAP Centre for Urban Health Solutions, Toronto, Ontario, Canada
| | - Mandy J. Hill
- Department of Emergency Medicine, University of Texas Health Sciences Center at Houston, McGovern Medical School, Houston, Texas, USA
| | - Darren Whitfield
- School of Social Work, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - S. Raquel Ramos
- Yale School of Public Health, Center for Interdisciplinary Research on AIDS, New Haven, Connecticut, USA
- Yale School of Nursing, New Haven, Connecticut, USA
| | | | - Lynda Emel
- Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Leo Wilton
- State University of New York at Binghamton, Binghamton, New York, USA
- Department of Humanities, University of Johannesburg, Johannesburg, South Africa
| | - Lisa Hightow-Weidman
- Institute for Global Health and Infectious Diseases, School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, North Carolina, USA
| | - Steve Shoptaw
- Division of Family Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Manya Magnus
- Department of Epidemiology, Milken Institute School of Public Health, George Washington University, Washington, District of Columbia, USA
| | | | - Estelle Piwowar-Manning
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore Maryland, USA
| | - Stephaun E. Wallace
- Vaccine and Infectious Disease Division, Fred Hutch, Seattle, Washington, USA
| | - Sheldon D. Fields
- Ross and Carol Nese College of Nursing, Pennsylvania State University, University Park, Pennsylvania, USA
| | | |
Collapse
|
34
|
Thurman AR, Ouattara LA, Yousefieh N, Anderson PL, Bushman LR, Fang X, Hanif H, Clark M, Singh O, Doncel GF. A phase I study to assess safety, pharmacokinetics, and pharmacodynamics of a vaginal insert containing tenofovir alafenamide and elvitegravir. Front Cell Infect Microbiol 2023; 13:1130101. [PMID: 37153145 PMCID: PMC10154607 DOI: 10.3389/fcimb.2023.1130101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/31/2023] [Indexed: 05/09/2023] Open
Abstract
Background New multi-purpose prevention technology (MPT) products are needed to prevent human immunodeficiency virus (HIV) and herpes simplex virus type 2 (HSV2). In this study, we evaluated a fast-dissolve insert that may be used vaginally or rectally for prevention of infection. Objective To describe the safety, acceptability, multi-compartment pharmacokinetics (PK), and in vitro modeled pharmacodynamics (PD) after a single vaginal dose of an insert containing tenofovir alafenamide (TAF) and elvitegravir (EVG) in healthy women. Methods This was a Phase I, open-label, study. Women (n=16) applied one TAF (20mg)/EVG (16mg) vaginal insert and were randomized (1:1) to sample collection time groups for up to 7 days post dosing. Safety was assessed by treatment-emergent adverse events (TEAEs). EVG, TAF and tenofovir (TFV) concentrations were measured in plasma, vaginal fluid and tissue, and TFV-diphosphate (TFV-DP) concentration in vaginal tissue. PD was modeled in vitro by quantifying the change in inhibitory activity of vaginal fluid and vaginal tissue against HIV and HSV2 from baseline to after treatment. Acceptability data was collected by a quantitative survey at baseline and post treatment. Results The TAF/EVG insert was safe, with all TEAEs graded as mild, and acceptable to participants. Systemic plasma exposure was low, consistent with topical delivery, while high mucosal levels were detected, with median TFV vaginal fluid concentrations exceeding 200,000 ng/mL and 1,000 ng/mL for up to 24 hours and 7 days post dosing, respectively. All participants had vaginal tissue EVG concentrations of > 1 ng/mg at 4 and 24 hours post dosing. The majority had tissue TFV-DP concentrations exceeding 1000 fmol/mg by 24 - 72 hours post dosing. Vaginal fluid inhibition of HIV-1 and HSV-2 in vitro significantly increased from baseline and was similarly high at 4 and 24 hours post dosing. Consistent with high tissue TFV-DP concentrations, p24 HIV antigen production from ectocervical tissues infected ex vivo with HIV-1 significantly decreased from baseline at 4 hours post dosing. HSV-2 production from tissue also decreased post treatment. Conclusions A single dose of TAF/EVG inserts met PK benchmarks, with PK data supporting an extended window of high mucosal protection. PD modeling supports mucosal protection against both HIV-1 and HSV-2. The inserts were safe and highly acceptable. Clinical trial registration ClinicalTrials.gov, identifier NCT03762772.
Collapse
Affiliation(s)
- Andrea R. Thurman
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States
- *Correspondence: Andrea R. Thurman,
| | - Louise A. Ouattara
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States
| | - Nazita Yousefieh
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States
| | - Peter L. Anderson
- University of Colorado, Colorado Antiviral Pharmacology Lab, School of Pharmacy, Anschutz Medical Campus, Aurora, CO, United States
| | - Lane R. Bushman
- University of Colorado, Colorado Antiviral Pharmacology Lab, School of Pharmacy, Anschutz Medical Campus, Aurora, CO, United States
| | - Xi Fang
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States
| | - Homaira Hanif
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States
| | - Meredith Clark
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States
| | - Onkar Singh
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States
| | - Gustavo F. Doncel
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States
| |
Collapse
|
35
|
Adeyemi OA, Nowak RG, Marzinke M, Morgan D, Sam-Agudu N, Craddock J, Zhan M, Crowell TA, Baral S, Ndembi N, Adebajo S, Charurat ME. Correlates of self-reported and biomarker based adherence to daily oral HIV pre-exposure prophylaxis among a cohort of predominantly men who have sex with men in Nigeria. PLoS One 2023; 18:e0282999. [PMID: 36928630 PMCID: PMC10019705 DOI: 10.1371/journal.pone.0282999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 02/28/2023] [Indexed: 03/17/2023] Open
Abstract
INTRODUCTION HIV pre-exposure prophylaxis (PrEP) significantly reduces the risk of HIV acquisition. However, studies have demonstrated discordance between self-reported measures and biomedical benchmarks of PrEP adherence. We estimated the correlation between self-reported PrEP adherence and PrEP biomarkers and explored factors associated with adherence among men who have sex with men (MSM) in Nigeria. METHODS TRUST-PrEP, an open-label, prospective study; conducted in Abuja between April 2018 and May 2019. MSM ≥ 18 years with substantial HIV risk were enrolled. Participants reported PrEP adherence in the last month using a 4-point scale from "poor" to "perfect" and serum samples for PrEP biomarkers were collected at months 3 and 9. Serum tenofovir concentration was measured by liquid chromatography-tandem mass spectrometry and considered protective for adherence if ≥ 4.2 ng/ml. Spearman's rank correlation was used to estimate correlation between self-reported adherence and measured tenofovir levels. Generalized estimating equations with a logit link was used to estimate adjusted odds ratios (aORs) and 95% confidence intervals (CIs) for associations between self-reported adherence and laboratory-measured adherence. RESULTS A total of 219 MSM with median age 23 (interquartile range 20-27) years had at least one PrEP biomarker assay. Only 66/219 (30%) had at least one record of protective tenofovir concentration. Correlation between tenofovir and self-reported adherence at 3 and 9 months were 0.1 and 0.02 respectively. Furthermore, 17/219 (8%,) and 49/219 (22%) had serum tenofovir of 4.2-35.4 ng/mL and ≥ 35.5 ng/mL, corresponding to at least 4 and 7 days' PrEP use in a week, respectively. PrEP adherence was higher among participants introduced to PrEP in the clinics compared with communities (aOR: 8.35, 95%CI: [3.24, 21.5]) and those with same-sex practices family disclosure (aOR: 3.60 95% CI: [1.73, 7.51]). CONCLUSION Self-reported PrEP adherence poorly correlated with biomarkers. Facilitating clinic-based PrEP introduction and disclosure of same-sex practices to family among MSM may improve PrEP adherence.
Collapse
Affiliation(s)
- Olusegun A. Adeyemi
- Department of Public Health and Epidemiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- International Research Center of Excellence, Institute of Human Virology, Abuja, Nigeria
- * E-mail:
| | - Rebecca G. Nowak
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Mark Marzinke
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Daniel Morgan
- Department of Public Health and Epidemiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Nadia Sam-Agudu
- International Research Center of Excellence, Institute of Human Virology, Abuja, Nigeria
| | - Jaih Craddock
- School of Social Work, University of Maryland, Baltimore, MD, United States of America
| | - Min Zhan
- Division of Biostatistics and Bioinformatics, Department of Public Health and Epidemiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Trevor A. Crowell
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
- U.S Military HIV Research Program, Silver Spring, Maryland, United States of America
| | - Stefan Baral
- The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Nicaise Ndembi
- International Research Center of Excellence, Institute of Human Virology, Abuja, Nigeria
| | - Sylvia Adebajo
- Center for International Health, Education, and Biosecurity, Abuja, Nigeria
| | - Manhattan E. Charurat
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | | |
Collapse
|
36
|
Coleman JS, Diniz CP, Fuchs EJ, Marzinke MA, Aung W, Bakshi RP, Farzadegan H, Bream JH, Nilles TL, Hudson S, Bumpus NN, Schwartz GJ, Rosenblum MA, Rooney JF, Hendrix CW. Interaction of Depot Medroxyprogesterone Acetate and Tenofovir Disoproxil Fumarate/Emtricitabine on Peripheral Blood Mononuclear Cells and Cervical Tissue Susceptibility to HIV Infection and Pharmacokinetics. J Acquir Immune Defic Syndr 2023; 92:89-96. [PMID: 36305827 PMCID: PMC9742287 DOI: 10.1097/qai.0000000000003113] [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: 07/21/2022] [Accepted: 09/19/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Depot medroxyprogesterone acetate (DMPA) is a widely used contraceptive method. HIV pre-exposure prophylaxis with emtricitabine and tenofovir disoproxil fumarate (F/TDF) is highly effective in reducing HIV acquisition in women. We sought to determine the impact of DMPA on F/TDF pharmacokinetics and pharmacodynamics. METHODS Twelve healthy premenopausal cisgender women were enrolled and each completed 4 sequential conditions: (1) baseline, (2) steady-state F/TDF alone, (3) steady-state F/TDF + DMPA, and (4) DMPA alone. Assessments included clinical, pharmacokinetic, viral infectivity (ex vivo challenge of peripheral blood mononuclear cells by X4- and R5-tropic green fluorescent protein pseudoviruses and cervical tissue by HIV BaL ), endocrine, immune cell phenotyping, and renal function. RESULTS Compared with baseline, F/TDF (± DMPA) significantly decreased both %R5- and X4-infected CD4 T cells and F/TDF + DMPA decreased cervical explant p24 (all P < 0.05). The %R5- and X4-infected CD4 T cells were higher during DMPA alone than during F/TDF periods and lower than baseline (not statistically significant). Cervical explant p24 fell between baseline and F/TDF values (not statistically significant). There were neither statistically significant differences in F/TDF pharmacokinetics, including total or renal clearance of either antiviral drug, nor changes in glomerular filtration rate with the addition of DMPA. There were few immune cell phenotypic differences across conditions. CONCLUSIONS F/TDF decreased HIV infection in both challenge assays, whereas DMPA alone did not enhance HIV infection in either challenge assay. DMPA did not alter F/TDF pharmacokinetics or renal function.
Collapse
Affiliation(s)
- Jenell S Coleman
- Department of Gynecology and Obstetrics, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Clarissa P Diniz
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Edward J Fuchs
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Mark A Marzinke
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Wutyi Aung
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Rahul P Bakshi
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Homayoon Farzadegan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Jay H Bream
- Department of Molecular Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Tricia L Nilles
- Department of Molecular Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Sherry Hudson
- Department of Molecular Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Namandjé N Bumpus
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pharmacology and Molecular Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - George J Schwartz
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY
| | - Michael A Rosenblum
- Department of Biostatistics, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD; and
| | | | - Craig W Hendrix
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
37
|
Xiao P, Gumber S, Marzinke MA, Hoang T, Myers R, Date AA, Hanes J, Ensign LM, Wang L, Rohan LC, Cone R, Fuchs EJ, Hendrix CW, Villinger F. Hypo-osmolar rectal douche tenofovir formulation prevents simian/human immunodeficiency virus acquisition in macaques. JCI Insight 2022; 7:161577. [PMID: 36477356 PMCID: PMC9746910 DOI: 10.1172/jci.insight.161577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 10/19/2022] [Indexed: 12/12/2022] Open
Abstract
In spite of the rollout of oral pre-exposure prophylaxis (PrEP), the rate of new HIV infections remains a major health crisis. In the United States, new infections occur predominantly in men having sex with men (MSM) in rural settings where access to PrEP can be limited. As an alternative congruent with MSM sexual behavior, we have optimized and tested tenofovir (TFV) and analog-based iso-osmolar and hypo-osmolar (HOsm) rectal douches for efficacy against rectal simian/human immunodeficiency virus (SHIV) infection of macaques. Single TFV HOsm high-dose douches achieved peak plasma TFV levels similar to daily oral PrEP, while other formulations yielded lower concentrations. Rectal tissue TFV-diphosphate (TFV-DP) concentrations at the portal of virus entry, however, were markedly higher after HOsm douching than daily oral PrEP. Repeated douches led to significantly higher plasma TFV and higher TFV-DP concentrations in rectal tissue at 24 hours compared with single douches, without detectable mucosal or systemic toxicity. Using stringent repeated intrarectal SHIV exposures, single HOsm high-dose douches delivered greater protection from virus acquisition for more than 24 hours compared with oral PrEP. Our results demonstrate a rapid delivery of protective TFV doses to the rectal portal of virus entry as a potential low-cost and safe PrEP alternative.
Collapse
Affiliation(s)
- Peng Xiao
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, Louisiana, USA
| | - Sanjeev Gumber
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Mark A Marzinke
- Division of Clinical Pharmacology, Department of Medicine.,Department of Pathology
| | - Thuy Hoang
- Center for Nanomedicine; and.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rohan Myers
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, Louisiana, USA
| | - Abhijit A Date
- Center for Nanomedicine; and.,Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona, USA.,Department of Ophthalmology and Vision Science, University of Arizona, Tucson, Arizona, USA
| | - Justin Hanes
- Center for Nanomedicine; and.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Laura M Ensign
- Center for Nanomedicine; and.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Chemical & Biomolecular Engineering, Johns Hopkins Whiting School of Engineering, Baltimore, Maryland, USA
| | - Lin Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Lisa C Rohan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Richard Cone
- Division of Clinical Pharmacology, Department of Medicine
| | - Edward J Fuchs
- Division of Clinical Pharmacology, Department of Medicine
| | - Craig W Hendrix
- Division of Clinical Pharmacology, Department of Medicine.,Center for Nanomedicine; and.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Francois Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, Louisiana, USA
| |
Collapse
|
38
|
Mayer KH, Yuhas K, Amico KR, Wilkin T, Landovitz RJ, Richardson P, Marzinke MA, Hendrix CW, Eshleman SH, Cottle LM, Marcus C, Chege W, Rinehart AR, Rooney JF, Andrew P, Salata RA, Magnus M, Farley JE, Liu AY, Frank I, Ho K, Santana J, Stekler JD, Chen YQ, McCauley M, Gulick RM. Sexual behavior and medication adherence in men who have sex with men participating in a pre-exposure prophylaxis study of combinations of Maraviroc, Tenofovir Disoproxil Fumarate and/or Emtricitabine (HPTN 069/ACTG 5305). AIDS Behav 2022; 26:4107-4114. [PMID: 35687192 PMCID: PMC10265494 DOI: 10.1007/s10461-022-03736-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2022] [Indexed: 11/01/2022]
Abstract
HPTN 069/ACTG 5305 was designed to evaluate potential new PrEP regimens that included maraviroc, tenofovir disoproxil fumarate, and/or emtricitabine. The current analyses assessed antiretroviral (ARV) plasma concentrations in relation to sexual behavior in 224 cisgender men who have sex with men and 2 transgender women at risk for HIV. Poisson generalized estimating equations (GEE) regression were used to test for associations between self-reported sexual behavior, sociodemographic, behavioral variables, and study drug levels The median (IQR) age was 30 [25, 37] years old; 48.2% had completed college; 27.4% were Black and 21.7% Latino. At weeks 24 and 48, one third of participants reported condomless anal sex (CAS) in the prior month with more than one partner. CAS was associated with daily ARV drug use (χ2 = 12.64, p = 0.002). Older individuals and those with greater education were more likely to ingest ARV drugs daily (χ2 = 9.36, p = 0.009 and χ2 = 8.63, p = 0.013, respectively), while neither race nor ethnicity was associated with daily ARV drug use. Participants who reported recent condomless anal sex and/or advanced education had higher rates of daily ARV drug use. These data support the need for ongoing adherence counseling in clinical trials of new PrEP modalities.
Collapse
Affiliation(s)
- Kenneth H Mayer
- The Fenway Institute, Fenway Health, Harvard Medical School, Harvard University, 1340 Boylston St, 02215, Boston, MA, United States.
| | - Krista Yuhas
- Statistical Center for HIV/AIDS Research (SCHARP), Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - K Rivet Amico
- University of Michigan School of Public Health, Ann Arbor, MI, United States
| | | | - Raphael J Landovitz
- UCLA Center for Clinical AIDS Research & Education, Los Angeles, CA, United States
| | - Paul Richardson
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Mark A Marzinke
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Craig W Hendrix
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Susan H Eshleman
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | | | - Cheryl Marcus
- HIV Clinical Trials Unit, University of North Carolina, Chapel Hill, NC, United States
| | - Wairimu Chege
- Clinical Prevention Research Branch, Prevention Sciences Program, Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Alex R Rinehart
- Global HIV Prevention Strategy, ViiV Healthcare - Research Triangle Park, Research Triangle Park, NC, United States
| | - James F Rooney
- Medical Affairs, Gilead Sciences, 12. FHI 360, Foster City, Durham, CA, NC, United States
| | | | - Robert A Salata
- Department of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Manya Magnus
- Milken Institute School of Public Health, George Washington University, Washington, DC, United States
| | - Jason E Farley
- Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Albert Y Liu
- San Francisco Department of Public Health, Bridge HIV, San Francisco, CA, United States
| | - Ian Frank
- Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Ken Ho
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | | | - Joanne D Stekler
- University of Washington School of Medicine, Seattle, WA, United States
| | - Ying Q Chen
- Statistical Center for HIV/AIDS Research (SCHARP), Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | | | - Roy M Gulick
- Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
39
|
Nelson LE, Boyd DT, Beauchamp G, Emel L, Wilton L, Whitfield D, Ramos SR, Ajiboye W, Hill MJ, Conserve DF, Thomas P, Hightow-Weidman L, Shoptaw S, Magnus M, Mayer KH, Piwowar-Manning E, Fields SD, Wheeler DP. Freedom as Prevention: Mechanisms of Autonomy Support for Promoting HIV Pre-Exposure Prophylaxis Use and Condom Use among Black MSM in 3 US Cities-HPTN 073. J Urban Health 2022; 99:1157-1169. [PMID: 35939181 PMCID: PMC9727017 DOI: 10.1007/s11524-022-00666-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/08/2022] [Indexed: 02/04/2023]
Abstract
Healthcare providers who use controlling or coercive strategies may compel short-term enactment of HIV and sexually transmitted infection prevention behaviors but may inadvertently undermine their client's motivation to maintain those behaviors in the absence of external pressure. Autonomous motivation refers to the self-emanating and self-determined drive for engaging in health behaviors. It is associated with long-term maintenance of health behaviors. We used structural equation modeling to investigate whether autonomy support was associated with increased odds of therapeutic serum levels of pre-exposure prophylaxis, through a pathway that satisfies basic psychological needs for autonomous self-regulation and competence regarding pre-exposure prophylaxis use. We also investigated whether autonomy support was associated with decreased odds of condomless anal intercourse via the same psychological needs-satisfaction pathway of autonomous self-regulation and competence regarding condom use. We tested these two theorized pathways using secondary data from a longitudinal sample of Black men who have sex with men from across three cities in the US (N = 226). Data from the sample fit the theorized models regarding the pathways by which autonomy support leads to the presence of therapeutic PrEP levels in serum (χ2 = 0.56; RMSEA = 0.04; CFI = .99, TLI = 0.98) and how it also leads to decreased odds of condomless anal intercourse (χ2 = 0.58; RMSEA = 0.03; CFI = 0.99; TLI = 0.98). These findings provide scientific evidence for the utility of self-determination theory as a model to guide intervention approaches to optimize the implementation and impact of PrEP for Black men who have sex with men.
Collapse
Affiliation(s)
- LaRon E Nelson
- School of Nursing, Yale University, 400 West Campus Drive, New Haven, CT, USA.
- Center for Interdisciplinary Research On AIDS, School of Public Health, Yale University, New Haven, CT, USA.
- MAP Centre for Urban Health Solutions, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.
| | - Donte T Boyd
- Center for Interdisciplinary Research On AIDS, School of Public Health, Yale University, New Haven, CT, USA
- College of Social Work, The Ohio State University, Columbus, OH, USA
| | - Geetha Beauchamp
- Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Lynda Emel
- Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Leo Wilton
- Department of Human Development, College of Community and Public Affairs, State University of New York at Binghamton, Binghamton, NY, USA
- Department of Humanities, University of Johannesburg, Johannesburg, South Africa
| | - Darren Whitfield
- School of Social Work, University of Maryland Baltimore, Baltimore, MD, USA
| | - S Raquel Ramos
- School of Nursing, Yale University, 400 West Campus Drive, New Haven, CT, USA
- Center for Interdisciplinary Research On AIDS, School of Public Health, Yale University, New Haven, CT, USA
| | - Wale Ajiboye
- MAP Centre for Urban Health Solutions, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Mandy J Hill
- Department of Emergency Medicine, McGovern Medical School, University of Texas Health, Houston, TX, USA
| | - Donaldson F Conserve
- Department of Prevention and Community Health, Milken Institute School of Public Health, George Washington Universty, Washington, DC, USA
| | - Portia Thomas
- Center for Interdisciplinary Research On AIDS, School of Public Health, Yale University, New Haven, CT, USA
- Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Lisa Hightow-Weidman
- Institute for Global Health and Infectious Diseases, School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, NC, USA
| | - Steve Shoptaw
- Division of Family Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Manya Magnus
- Department of Epidemiology, Milken Institute School of Public Health, George Washington University, Washington, DC, USA
| | | | | | - Sheldon D Fields
- Ross and Carol Nese College of Nursing, Pennsylvania State University, University Park, PA, USA
| | | |
Collapse
|
40
|
Yager J, Brooks KM, Brothers J, Mulligan K, Landovitz R, Reirden D, Glenny C, Malhotra M, Anderson PL, Hosek S. Pharmacokinetics of Emtricitabine/Tenofovir Disoproxil Fumarate Among Transgender Adolescents and Young Adults Without HIV Receiving Gender Affirming Hormones. AIDS Res Hum Retroviruses 2022; 38:840-846. [PMID: 35943868 PMCID: PMC9910104 DOI: 10.1089/aid.2022.0043] [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: 01/25/2023] Open
Abstract
The transgender community has expressed concerns regarding drug-drug interactions between HIV-pre-exposure prophylaxis (PrEP) and gender-affirming hormones. In this study, we evaluated emtricitabine (F, FTC)/tenofovir (TFV) disoporoxil fumarate (TDF) pharmacokinetics (PK) among adolescent and young adult transgender persons receiving gender-affirming hormone therapy (GAHT). This was a prospective, observational study among transgender women (TW) and men (TM) without HIV, 15-24 years of age, receiving GAHT (estradiol with/without spironolactone, or testosterone). Participants received 1 month of directly observed daily F/TDF. Weekly convenience blood samples were collected for plasma TFV and FTC, and intracellular TFV-diphosphate (TFV-DP) and FTC-triphosphate (FTC-TP) in peripheral blood mononuclear cells (PBMC) and dried blood spots (DBS). After 2-3 weeks of F/TDF dosing, intensive PK sampling was conducted. PK parameters were estimated using noncompartmental methods. Data were log-transformed and compared between TM and TW, and to historical data among cisgender adults. Plasma TFV exposures were similar between TM and TW [geometric mean ratio (GMR); confidence interval (95% CI): 1.06 (0.89-1.28)], whereas FTC plasma exposures were 21% higher in TM versus TW (95% CI: 1.07-1.38). TFV-DP in PBMC and DBS and FTC-TP in DBS did not differ between TM versus TW after controlling for creatinine clearance (CrCl), but FTC-TP in PBMC remained 46% (95% CI: 1.15-1.86) higher in TM versus TW. All PK exposures were within expected ranges based on historical studies. TM had higher FTC exposures compared with TW, but overall plasma and intracellular exposures for both drugs were within the range of historical studies, suggesting high PrEP efficacy will be retained in adolescent and young adult transgender persons. Registered at ClinicalTrials.gov (NCT03652623).
Collapse
Affiliation(s)
- Jenna Yager
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Kristina M. Brooks
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Jennifer Brothers
- Department of Psychiatry, Stroger Hospital of Cook County, Chicago, Illinois, USA
| | - Kathleen Mulligan
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Francisco, San Francisco, California, USA
| | - Raphael Landovitz
- Department of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Daniel Reirden
- Department of Adolescent Medicine, Children's Hospital of Colorado, Aurora, Colorado, USA
| | - Carrie Glenny
- Department of Adolescent Medicine, Children's Hospital of Colorado, Aurora, Colorado, USA
| | - Meena Malhotra
- Department of Psychiatry, Stroger Hospital of Cook County, Chicago, Illinois, USA
| | - Peter L. Anderson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Sybil Hosek
- Department of Psychiatry, Stroger Hospital of Cook County, Chicago, Illinois, USA
| |
Collapse
|
41
|
Thurman AR, Brache V, Cochon L, Ouattara LA, Chandra N, Jacot T, Yousefieh N, Clark MR, Peet M, Hanif H, Schwartz JL, Ju S, Marzinke MA, Erikson DW, Parikh U, Herold BC, Fichorova RN, Tolley E, Doncel GF. Randomized, placebo controlled phase I trial of the safety, pharmacokinetics, pharmacodynamics and acceptability of a 90 day tenofovir plus levonorgestrel vaginal ring used continuously or cyclically in women: The CONRAD 138 study. PLoS One 2022; 17:e0275794. [PMID: 36215267 PMCID: PMC9550080 DOI: 10.1371/journal.pone.0275794] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 08/29/2022] [Indexed: 11/04/2022] Open
Abstract
Multipurpose prevention technologies (MPTs), which prevent sexually transmitted infection(s) and unintended pregnancy, are highly desirable to women. In this randomized, placebo-controlled, phase I study, women used a placebo or tenofovir (TFV) and levonorgestrel (LNG) intravaginal ring (IVR), either continuously or cyclically (three, 28-day cycles with a 3 day interruption in between each cycle), for 90 days. Sixty-eight women were screened; 47 were randomized to 4 arms: TFV/LNG or placebo IVRs used continuously or cyclically (4:4:1:1). Safety was assessed by adverse events and changes from baseline in mucosal histology and immune mediators. TFV concentrations were evaluated in multiple compartments. LNG concentration was determined in serum. Modeled TFV pharmacodynamic antiviral activity was evaluated in vaginal and rectal fluids and cervicovaginal tissue ex vivo. LNG pharmacodynamics was assessed with cervical mucus quality and anovulation. All IVRs were safe with no serious adverse events nor significant changes in genital tract histology, immune cell density or secreted soluble proteins from baseline. Median vaginal fluid TFV concentrations were >500 ng/mg throughout 90d. TFV-diphosphate tissue concentrations exceeded 1,000 fmol/mg within 72hrs of IVR insertion. Mean serum LNG concentrations exceeded 200 pg/mL within 2h of TFV/LNG use, decreasing quickly after IVR removal. Vaginal fluid of women using TFV-containing IVRs had significantly greater inhibitory activity (87-98% versus 10% at baseline; p<0.01) against HIV replication in vitro. There was a >10-fold reduction in HIV p24 antigen production from ectocervical tissues after TFV/LNG exposure. TFV/LNG IVR users had significantly higher rates of anovulation, lower Insler scores and poorer/abnormal cervical mucus sperm penetration. Most TFV/LNG IVR users reported no change in menstrual cycles or fewer days of and/or lighter bleeding. All IVRs were safe. Active rings delivered high TFV concentrations locally. LNG caused changes in cervical mucus, sperm penetration, and ovulation compatible with contraceptive efficacy. Trial registration: ClinicalTrials.gov #NCT03279120.
Collapse
Affiliation(s)
- Andrea R. Thurman
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States of America
- * E-mail:
| | | | | | - Louise A. Ouattara
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States of America
| | - Neelima Chandra
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States of America
| | - Terry Jacot
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States of America
| | - Nazita Yousefieh
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States of America
| | - Meredith R. Clark
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States of America
| | - Melissa Peet
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States of America
| | - Homaira Hanif
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States of America
| | - Jill L. Schwartz
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States of America
| | - Susan Ju
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States of America
| | - Mark A. Marzinke
- Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - David W. Erikson
- Endocrine Technologies Core (ETC), Oregon National Primate Research Center (ONPRC), Beaverton, OR, United States of America
| | - Urvi Parikh
- Department of Medicine, Division of Infectious Diseases and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Betsy C. Herold
- Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Raina N. Fichorova
- Laboratory of Genital Tract Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Elizabeth Tolley
- Family Health International 360, Research Triangle, NC, United States of America
| | - Gustavo F. Doncel
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States of America
| |
Collapse
|
42
|
Beesham I, Mansoor LE, Joseph Davey DL, Palanee-Phillips T, Smit J, Ahmed K, Selepe P, Louw C, Singata-Madliki M, Kotze P, Heffron R, Parikh UM, Wiesner L, Rees H, Baeten JM, Beksinska M. Brief Report: Quantifiable Plasma Tenofovir Among South African Women Using Daily Oral Pre-exposure Prophylaxis During the ECHO Trial. J Acquir Immune Defic Syndr 2022; 91:26-30. [PMID: 35972853 PMCID: PMC9377486 DOI: 10.1097/qai.0000000000003023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/16/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND HIV endpoint-driven clinical trials provide oral pre-exposure prophylaxis (PrEP) as HIV prevention standard of care. We evaluated quantifiable plasma tenofovir among South African women who used oral PrEP during the Evidence for Contraceptive Options and HIV Outcomes (ECHO) Trial. METHODS ECHO, a randomized trial conducted in 4 African countries between 2015 and 2018, assessed HIV incidence among HIV-uninfected women, aged 16-35 years, randomized to 1 of 3 contraceptives. Oral PrEP was offered onsite as part of the HIV prevention package at the South African trial sites. We measured tenofovir in plasma samples collected at the final trial visit among women reporting ongoing PrEP use. We used bivariate and multivariate logistical regression to assess demographic and sexual risk factors associated with plasma tenofovir quantification. RESULTS Of 260 women included, 52% were ≤24 years and 22% had Chlamydia trachomatis at enrollment. At PrEP initiation, 68% reported inconsistent/nonuse of condoms. The median duration of PrEP use was 90 days (IQR: 83-104). Tenofovir was quantified in 36% (n = 94) of samples. Women >24 years had twice the odds of having tenofovir quantified vs younger women (OR = 2.12; 95% confidence interval = 1.27 to 3.56). Women who reported inconsistent/nonuse of condoms had lower odds of tenofovir quantification (age-adjusted OR = 0.47; 95% confidence interval = 0.26 to 0.83). CONCLUSIONS Over a third of women initiating PrEP and reporting ongoing use at the final trial visit had evidence of recent drug exposure. Clinical trials may serve as an entry point for PrEP initiation among women at substantial risk for HIV infection with referral to local facilities for ongoing access at trial end. CLINICAL TRIAL NUMBER NCT02550067.
Collapse
Affiliation(s)
- Ivana Beesham
- MatCH Research Unit (MRU), Department of Obstetrics and Gynaecology, Faculty of Health Sciences, University of the Witwatersrand, Durban, South Africa
| | - Leila E. Mansoor
- Centre for AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
| | - Dvora L. Joseph Davey
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA
| | - Thesla Palanee-Phillips
- Wits Reproductive Health and HIV Institute (Wits RHI), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jenni Smit
- MatCH Research Unit (MRU), Department of Obstetrics and Gynaecology, Faculty of Health Sciences, University of the Witwatersrand, Durban, South Africa
| | - Khatija Ahmed
- Setshaba Research Centre, Soshanguve, South Africa
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | | | - Cheryl Louw
- Madibeng Centre for Research, Brits, South Africa
- Department of Family Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Mandisa Singata-Madliki
- Effective Care Research Unit, Universities of the Witwatersrand and Fort Hare and Eastern Cape Department of Health, East London, South Africa
| | - Philip Kotze
- Qhakaza Mbokodo Research Clinic, Ladysmith, South Africa;
| | - Renee Heffron
- Department of Global Health and Department of Epidemiology, University of Washington, Seattle, WA
| | - Urvi M. Parikh
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Lubbe Wiesner
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa;
| | - Helen Rees
- Wits Reproductive Health and HIV Institute (Wits RHI), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jared M. Baeten
- Department of Global Health, Department of Epidemiology, Department of Medicine, University of Washington, Seattle, WA; and
- Gilead Sciences, Foster City, CA
| | - Mags Beksinska
- MatCH Research Unit (MRU), Department of Obstetrics and Gynaecology, Faculty of Health Sciences, University of the Witwatersrand, Durban, South Africa
| |
Collapse
|
43
|
Niu X, Kubiak RW, Siriprakaisil O, Klinbuyaem V, Sukrakanchana PO, Cressey R, Okochi H, Gandhi M, Cressey TR, Drain PK. Tenofovir-Diphosphate in Dried Blood Spots vs Tenofovir in Urine/Plasma for Oral Preexposure Prophylaxis Adherence Monitoring. Open Forum Infect Dis 2022; 9:ofac405. [PMID: 36004315 PMCID: PMC9394764 DOI: 10.1093/ofid/ofac405] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 08/08/2022] [Indexed: 12/02/2022] Open
Abstract
Background Tenofovir-diphosphate (TFV-DP) measured in dried blood spots (DBS) and tenofovir (TFV) measured in urine/plasma have been used to measure TFV-based oral pre-exposure prophylaxis (PrEP) adherence. However, there are limited data comparing these 3 metrics and their appropriate use for PrEP adherence monitoring. Methods We collected DBS, urine, and plasma samples from HIV-negative adults randomized to a low (2 doses/week), moderate (4 doses/week), or perfect (7 doses/week) adherence group (via directly observed therapy) of tenofovir disoproxil fumarate/emtricitabine (TDF/FTC) for 6 weeks, followed by a 4-week washout phase. Drug concentrations were measured using liquid chromatography tandem mass spectrometry. Linear mixed-effects modeling was used to examine associations between drug concentrations and dosing time. Results Among 28 participants, the median age was 33 years, and 12 (43%) were female. At steady state, 25th percentile TFV-DP concentrations were 466, 779, and 1375 fmol/3 mm punch in the low, moderate, and perfect adherence group, respectively. Correlation was stronger between quantifiable TFV-DP and plasma TFV (r = 0.65; P < .01) than between TFV-DP and urine TFV (r = 0.50; P < .01). Among all participants, each additional week of cumulative dosing on average led to a mean increase of 158 fmol/3 mm punch (P < .001) in TFV-DP during the dosing phase. Each additional day after the last dose was associated with 43 fmol/3 mm punch lower TFV-DP (P = .07). Conclusions TFV-DP levels in DBS provide valuable insight into both dosing recency and cumulative doses from variable adherence patterns. Our observed benchmark TFV-DP concentrations were slightly higher than prior predicted estimates based on convenience samples.
Collapse
Affiliation(s)
- Xin Niu
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Rachel W Kubiak
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | | | | | - Pra ornsuda Sukrakanchana
- AMS/IRD Research Collaboration, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Ratchada Cressey
- Division of Clinical Chemistry, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Hideaki Okochi
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Monica Gandhi
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Tim R Cressey
- AMS/IRD Research Collaboration, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | - Paul K Drain
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
44
|
Gatto GJ, Krovi A, Li L, Massud I, Holder A, Gary J, Mills P, Mitchell J, Luecke E, Demkovich ZR, Heneine W, García-Lerma JG, Marzinke MA, Brand RM, Dobard CW, Johnson LM, Van Der Straten A. Comparative Pharmacokinetics and Local Tolerance of Tenofovir Alafenamide (TAF) From Subcutaneous Implant in Rabbits, Dogs, and Macaques. Front Pharmacol 2022; 13:923954. [PMID: 35928266 PMCID: PMC9343794 DOI: 10.3389/fphar.2022.923954] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/16/2022] [Indexed: 01/18/2023] Open
Abstract
The administration of antiretrovirals (ARVs) for HIV pre-exposure prophylaxis (PrEP) is highly efficacious and may benefit from new long-acting (LA) drug delivery approaches. This paper describes a subcutaneous, reservoir-style implant for the LA delivery of tenofovir alafenamide (TAF) and documents the preclinical assessment of implant safety and pharmacokinetics (PK) in New Zealand White (NZW) rabbits (3 groups of n = 5), beagle dogs (2 groups of n = 6), and rhesus macaques (2 groups of n = 3). Placebo implants were placed in rabbits (n = 10) and dogs (n = 12). Implant parameters, including selection of the TAF form, choice of excipient, and PCL formulation were tuned to achieve targeted concentrations of the active anabolite of TAF, tenofovir diphosphate (TFV-DP), within peripheral blood mononuclear cells (PBMCs) and mucosal tissues relevant to HIV transmission. Sustained concentrations of TFV-DP in PBMCs over 100 fmol/106 cells were achieved in all animal species indicating that the implants effectively delivered TAF for 3-6 months. Unlike placebo implants without TAF, all active implants resulted in local adverse events (AEs) proximal to the implant ranging in severity from mild to moderate and included dermal inflammation and necrosis across all species. Despite these AEs, the implant performed as designed and achieved a constant drug release profile, supporting the continued development of this drug delivery platform.
Collapse
Affiliation(s)
- G. J. Gatto
- RTI International, Research Triangle Park, NC, United States
| | - A. Krovi
- RTI International, Research Triangle Park, NC, United States
| | - L. Li
- RTI International, Research Triangle Park, NC, United States
| | - I. Massud
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - A. Holder
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - J. Gary
- Neuropathology, StageBio, Frederick, MD, United States
| | - P. Mills
- Department of Comparative Medicine, Tulane University, New Orleans, LA, United States
| | - J. Mitchell
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - E. Luecke
- RTI International, Research Triangle Park, NC, United States
| | - Z. R. Demkovich
- RTI International, Research Triangle Park, NC, United States
| | - W. Heneine
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - J. G. García-Lerma
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - M. A. Marzinke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - R. M. Brand
- Department of Internal Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - C. W. Dobard
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - L. M. Johnson
- RTI International, Research Triangle Park, NC, United States
| | - A. Van Der Straten
- Department of Medicine, Center for AIDS Prevention Studies (CAPS), University of California, San Francisco, CA, United States
- ASTRA Consulting, Kensington, CA, United States
| |
Collapse
|
45
|
Delany-Moretlwe S, Hughes JP, Bock P, Ouma SG, Hunidzarira P, Kalonji D, Kayange N, Makhema J, Mandima P, Mathew C, Spooner E, Mpendo J, Mukwekwerere P, Mgodi N, Ntege PN, Nair G, Nakabiito C, Nuwagaba-Biribonwoha H, Panchia R, Singh N, Siziba B, Farrior J, Rose S, Anderson PL, Eshleman SH, Marzinke MA, Hendrix CW, Beigel-Orme S, Hosek S, Tolley E, Sista N, Adeyeye A, Rooney JF, Rinehart A, Spreen WR, Smith K, Hanscom B, Cohen MS, Hosseinipour MC. Cabotegravir for the prevention of HIV-1 in women: results from HPTN 084, a phase 3, randomised clinical trial. Lancet 2022; 399:1779-1789. [PMID: 35378077 PMCID: PMC9077443 DOI: 10.1016/s0140-6736(22)00538-4] [Citation(s) in RCA: 287] [Impact Index Per Article: 95.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/12/2022] [Accepted: 03/14/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Oral pre-exposure prophylaxis has been introduced in more than 70 countries, including many in sub-Saharan Africa, but women experience considerable barriers to daily pill-taking, such as stigma, judgement, and the fear of violence. Safe and effective long-acting agents for HIV prevention are needed for women. We aimed to evaluate the safety and efficacy of injectable cabotegravir compared with daily oral tenofovir diphosphate plus emtricitabine (TDF-FTC) for HIV prevention in HIV-uninfected women. METHODS HPTN 084 was a phase 3, randomised, double-blind, double-dummy, active-controlled, superiority trial in 20 clinical research sites in seven countries in sub-Saharan Africa. Participants were eligible for enrolment if they were assigned female sex at birth, were aged 18-45 years, reported at least two episodes of vaginal intercourse in the previous 30 days, were at risk of HIV infection based on an HIV risk score, and agreed to use a long-acting reversible contraceptive method. Participants were randomly assigned (1:1) to either active cabotegravir with TDF-FTC placebo (cabotegravir group) or active TDF-FTC with cabotegravir placebo (TDF-FTC group). Study staff and participants were masked to study group allocation, with the exception of the site pharmacist who was responsible for study product preparation. Participants were prescribed 5 weeks of daily oral product followed by intramuscular injections every 8 weeks after an initial 4-week interval load, alongside daily oral pills. Participants who discontinued injections were offered open-label daily TDF-FTC for 48 weeks. The primary endpoints of the study were incident HIV infection in the intention-to-treat population, and clinical and laboratory events that were grade 2 or higher in all women who had received at least one dose of study product. This study is registered with ClinicalTrials.gov, NCT03164564. FINDINGS From Nov 27, 2017, to Nov 4, 2020, we enrolled 3224 participants (1614 in the cabotegravir group and 1610 in the TDF-FTC group). Median age was 25 years (IQR 22-30); 1755 (54·7%) of 3209 had two or more partners in the preceding month. 40 incident infections were observed over 3898 person-years (HIV incidence 1·0% [95% CI 0·73-1·40]); four in the cabotegravir group (HIV incidence 0·2 cases per 100 person-years [0·06-0·52]) and 36 in the TDF-FTC group (1·85 cases per 100 person-years [1·3-2·57]; hazard ratio 0·12 [0·05-0·31]; p<0·0001; risk difference -1·6% [-1·0% to -2·3%]. In a random subset of 405 TDF-FTC participants, 812 (42·1%) of 1929 plasma samples had tenofovir concentrations consistent with daily use. Injection coverage was 93% of the total number of person-years. Adverse event rates were similar across both groups, apart from injection site reactions, which were more frequent in the cabotegravir group than in the TDF-FTC group (577 [38·0%] of 1519 vs 162 [10·7%] of 1516]) but did not result in injection discontinuation. Confirmed pregnancy incidence was 1·3 per 100 person-years (0·9-1·7); no congenital birth anomalies were reported. INTERPRETATION Although both products for HIV prevention were generally safe, well tolerated, and effective, cabotegravir was superior to TDF-FTC in preventing HIV infection in women. FUNDING National Institute of Allergy and Infectious Diseases, ViiV Healthcare, and the Bill & Melinda Gates Foundation. Additional support was provided through the National Institute of Mental Health, the National Institute on Drug Abuse, and the Eunice Kennedy Shriver National Institute of Child Health and Human Development. ViiV Healthcare and Gilead Sciences provided pharmaceutical support.
Collapse
Affiliation(s)
- Sinead Delany-Moretlwe
- Wits Reproductive Health and HIV Institute, University of the Witwatersrand, Johannesburg, South Africa.
| | - James P Hughes
- Statistical Centre for HIV/AIDS Research and Prevention, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Peter Bock
- Desmond Tutu TB Centre, University of Stellenbosch, Stellenbosch, South Africa
| | - Samuel Gurrion Ouma
- Kisumu Clinical Research Site, Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Portia Hunidzarira
- Clinical Trials Research Centre, University of Zimbabwe, Harare, Zimbabwe
| | - Dishiki Kalonji
- HIV and other Infectious Diseases Research Unit, South African Medical Research Council, Durban, South Africa
| | - Noel Kayange
- Blantyre Clinical Research Site, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Joseph Makhema
- Botswana Harvard AIDS Institute Partnership (BHP), Gaborone, Botswana
| | - Patricia Mandima
- Clinical Trials Research Centre, University of Zimbabwe, Harare, Zimbabwe
| | - Carrie Mathew
- Wits Reproductive Health and HIV Institute, University of the Witwatersrand, Johannesburg, South Africa
| | - Elizabeth Spooner
- HIV and other Infectious Diseases Research Unit, South African Medical Research Council, Durban, South Africa
| | - Juliet Mpendo
- International AIDS Vaccine Initiative, Uganda Virus Research Institute, Entebbe, Uganda
| | | | - Nyaradzo Mgodi
- Clinical Trials Research Centre, University of Zimbabwe, Harare, Zimbabwe
| | | | - Gonasagrie Nair
- Desmond Tutu Health Foundation, University of Cape Town, Cape Town, South Africa
| | - Clemensia Nakabiito
- Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda
| | - Harriet Nuwagaba-Biribonwoha
- Eswatini Prevention Center, International Center for AIDS Care and Treatment Program at Columbia University Mailman School of Public Health, New York, NY, USA
| | - Ravindre Panchia
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Nishanta Singh
- HIV and other Infectious Diseases Research Unit, South African Medical Research Council, Durban, South Africa
| | - Bekezela Siziba
- Clinical Trials Research Centre, University of Zimbabwe, Harare, Zimbabwe
| | | | | | - Peter L Anderson
- Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - Susan H Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mark A Marzinke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Craig W Hendrix
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephanie Beigel-Orme
- Statistical Centre for HIV/AIDS Research and Prevention, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sybil Hosek
- Department of Psychiatry, Stroger Hospital of Cook County, Chicago, IL, USA
| | | | | | - Adeola Adeyeye
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Rockville, MD, USA
| | | | | | | | | | - Brett Hanscom
- Statistical Centre for HIV/AIDS Research and Prevention, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Myron S Cohen
- University of North Carolina (UNC) at Chapel Hill, Chapel Hill, NC, USA
| | - Mina C Hosseinipour
- University of North Carolina (UNC) at Chapel Hill, Chapel Hill, NC, USA; UNC Project-Malawi, Lilongwe, Malawi
| |
Collapse
|
46
|
Mass Spectroscopy Imaging of Hair Strands Captures Short-Term and Long-Term Changes in Emtricitabine Adherence. Antimicrob Agents Chemother 2022; 66:e0217621. [PMID: 35266824 PMCID: PMC9017293 DOI: 10.1128/aac.02176-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Most measures of adherence to antiretroviral therapy require a blood sample, and none capture longitudinal daily adherence. A new noninvasive method for measuring daily adherence to antiretroviral regimens containing emtricitabine (FTC) was developed for intact hair strands using infrared matrix-assisted laser desorption electrospray ionization (IR-MALDESI) mass spectrometry imaging (MSI). A directly observed therapy study of daily and intermittent (3, 1, and 0 doses/week) FTC dosing (n = 12) benchmarked adherence in hair, revealing distinct accumulation patterns and median FTC signal abundance (1,702, 495, 352, and 0, respectively) with each dosing frequency. A threshold value of FTCsignal abundance of 500 differentiated daily dosing from 3 or fewer doses/week (specificity, 100%; sensitivity, 100% over 30 days and 80% over 60 days). Using these criteria, daily FTC hair adherence was classified in young men (n = 8) who have sex with men (YMSM) engaged in or initiating preexposure prophylaxis (PrEP). Four types of adherence profiles were observed in sequential 30-day periods: consistently high, occasional missed doses, improvement following study initiation, and intermittent. Discrete days of nonadherence were identified across the 60-day window, with the average number of consecutive days classified as nonadherent increasing across the four profile types (1, 2, 19, and 58 days, respectively). Additionally, cumulative FTC response in hair (60-day average) significantly correlated with dried blood spot tenofovir diphosphate concentrations collected simultaneously (rs = 0.79, P = 0.03). Based on these data, IR-MALDESI FTC adherence classification in hair strands can better delineate short-term changes in adherence behaviors over a long retrospective window, offering great potential for noninvasive adherence monitoring and quick supportive interventions.
Collapse
|
47
|
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 PMCID: PMC9048169 DOI: 10.1089/aid.2021.0115] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The 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
|
48
|
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 PMCID: PMC9048178 DOI: 10.1089/aid.2021.0071] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [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
|
49
|
Morris S, Cottrell M, Rawlings SA, Peterson S, Karris M, Pacheco D, Chaillon A, Kay A, Chow K, Anderson PL, Gianella S, Blumenthal J. Genital Inflammation Is Not Associated With Decreased Vaginal Tenofovir Concentrations in Women Taking Oral PrEP. J Acquir Immune Defic Syndr 2022; 89:390-395. [PMID: 35202047 PMCID: PMC8887788 DOI: 10.1097/qai.0000000000002884] [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] [Received: 07/02/2021] [Accepted: 10/04/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND We evaluated the association of inflammation and dysbosis on cervicovaginal fluid (CVF) tenofovir (TFV) concentrations in women taking oral tenofovir disoproxil fumarate/emtricitable for HIV pre-exposure prophylaxis (PrEP) in the United States. SETTING Thirty-five women in a HIV PrEP implementation study attended their week 24 visit at a San Diego research clinic and provided CVF specimens. METHODS Women in the Adherence Enhancement Guided by Individualized Texting and Drug Levels study had their CVF specimens evaluated for (1) sexually transmitted bacterial (Neisseria gonorrhoeae, Chlamydia trachomatis, Gardnerella, and Trichomonas vaginalis), viral (human papillomavirus, cytomegalovirus and herpes simplex virus-1/2) and fungal (Candida) infections; (2) microbiome composition by 16 S sequencing (V3-V4 region); and (3) cytokine profiles by enzyme-linked immunoassay (Interleukin-8, macrophage Inflammatory protein-1a, macrophage Inflammatory Protein-1b and interferon-γ-inducible protein-10). Univariate statistical analysis was used to determine factors associated with CVF TFV concentrations. CVF TFV of 100-1000 ng/mL benchmarked typical genital concentrations and TFV-diphosphate in dried blood spots of 700 fmol/punch was considered adequate adherence. RESULTS Thirty-five women had CVF specimens collected. No factor was associated with CVF TFV concentrations or discordance of blood and vaginal concentrations. Among 27 participants assessed for vaginosis (Candida, Gardnerella or Trichomonas), women with Gardnerella (n = 11) were more likely to have high (>1000 ng/mL) CVF TFV concentrations (82% versus 33%, P = 0.02). CONCLUSIONS Presence of genital viruses, cytokines, or vaginal community state types were not associated with low CVF TFV concentrations in cisgender women taking oral tenofovir disoproxil fumarate/emtricitable for PrEP. The surprising association observed between presence of Gardnerella and higher vaginal TFV concentrations needs further evaluation.
Collapse
Affiliation(s)
| | | | | | - Scott Peterson
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Maile Karris
- University of California San Diego, La Jolla, CA
| | | | | | - Alexis Kay
- University of California San Diego, La Jolla, CA
| | - Karen Chow
- University of California San Diego, La Jolla, CA
| | | | | | | |
Collapse
|
50
|
Little KM, Flomen L, Hanif H, Anderson SM, Thurman AR, Clark MR, Doncel GF. HIV Pre-exposure Prophylaxis Implant Stated Preferences and Priorities: Results of a Discrete Choice Experiment Among Women and Adolescent Girls in Gauteng Province, South Africa. AIDS Behav 2022; 26:3099-3109. [PMID: 35360893 PMCID: PMC9371991 DOI: 10.1007/s10461-022-03658-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2022] [Indexed: 12/14/2022]
Abstract
For adolescent girls (AG) and young women (YW), adherence barriers may limit the effectiveness of daily oral HIV pre-exposure prophylaxis (PrEP). Due to its low-burden and long-lasting product attributes, PrEP implants could remove some of the critical adherence barriers of oral PrEP products for individuals at risk of HIV. To explore stated preferences for a long-acting PrEP implant, we conducted a quantitative survey and discrete choice experiment with AG (ages 15-17), YW (18-34), and female sex workers (FSW; ≥ 18) in Gauteng Province, South Africa. We completed 600 quantitative surveys across the three subgroups of women. Respondents stated preference for an implant that provided longer HIV protection (24 months versus 6 months) and required a single insertion. They stated that they preferred a biodegradable implant that could be removed within 1 month of insertion. Respondents had no preference for a particular insertion location. Overall, 78% of respondents said they would be likely (33%) or very likely (45%) to use a PrEP implant were one available, with the majority (82%) stating preference for a product that would provide dual protection against HIV and unintended pregnancies. To reduce their risk of HIV, AG, YW, and FSW in our survey reported a strong willingness to use long-acting, highly-effective, dissolvable PrEP implants.
Collapse
Affiliation(s)
- Kristen M Little
- HIV/TB Department, Population Services International (PSI), Washington, DC, USA
| | - Lola Flomen
- Strategy & Insights Department, PSI, 1120 19th Street NW, Suite 600, Washington, DC, 20036, USA.
| | - Homaira Hanif
- CONRAD, Eastern Virginia Medical School, Norfolk, VA, USA
| | | | | | | | | |
Collapse
|