101
|
Pharmacokinetics and Pharmacodynamics of Tenofovir Reduced-Glycerin 1% Gel in the Rectal and Vaginal Compartments in Women: A Cross-Compartmental Study With Directly Observed Dosing. J Acquir Immune Defic Syndr 2019; 78:175-182. [PMID: 29767639 DOI: 10.1097/qai.0000000000001655] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Evidence is lacking regarding whether vaginal pre-exposure prophylaxis with topical tenofovir (TFV) reduces the risk of rectal HIV acquisition. SETTING Bronx, NY. METHODS MTN-014 was a phase 1, cross-over, randomized sequence trial comparing the cross-compartment pharmacokinetics and pharmacodynamics of daily TFV reduced-glycerin 1% gel after 14 days each of rectal and vaginal application, with directly observed dosing and a 6-week washout period between phases. RESULTS Fourteen HIV-uninfected women enrolled; 91% of doses were observed and 13 women completed all study procedures. TFV and TFV diphosphate (TFV-DP) were detected in most samples collected from the dosing compartment. After vaginal dosing, TFV was detected in 10/14 samples of rectal fluid (RF) (median 4.4 ng/sponge) and 1/13 rectal tissue samples (0.2 ng/mg); TFV-DP was detected in 2/13 rectal tissue samples at 59.8 and 76.5 fmol/mg. After rectal dosing, TFV was detected in 9/14 samples of vaginal fluid (median 1.1 ng/swab) and in 6/14 vaginal tissue samples (median below limit of quantification); TFV-DP was detected in 3/14 vaginal tissue samples at 17.3, 87.6, and 77.1 fmol/mg. Neither cervicovaginal lavage fluid nor RF collected 24 hours after rectal or vaginal dosing resulted in a statistically significant suppression of viral replication. CONCLUSIONS In this study of 14 days each of vaginal and rectal application of TFV reduced-glycerin 1% gel, we found only a small degree of cross-compartment distribution of TFV in RF and vaginal fluids and no pharmacodynamic activity in ex vivo testing. Although high TFV concentrations in the dosing compartment may be protective, low cross-compartment tissue concentrations are not likely to be protective.
Collapse
|
102
|
Predictors of Daily Adherence to HIV Pre-exposure Prophylaxis in Gay/Bisexual Men in the PRELUDE Demonstration Project. AIDS Behav 2019; 23:1287-1296. [PMID: 30523489 DOI: 10.1007/s10461-018-2353-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Adequate adherence to pre-exposure prophylaxis (PrEP) is critical to prevent HIV infection, but accurately measuring adherence remains challenging. We compared two biological [blood drug concentrations in plasma and peripheral blood mononuclear cells (PBMC)] and two self-reported measures (facilitated recall to clinicians and self-report in online surveys) and identified predictors of daily PrEP adherence among gay and bisexual men (GBM) in their first 12 months on PRELUDE, an open-label, single-arm PrEP demonstration project in New South Wales, Australia. 327 participants were enrolled; 263 GBM attended their 12-month follow-up visit (81% retention). Overall, 91% of blood samples had plasma drug concentrations indicative of taking 7 pills/week, and 99% had protective drug concentrations (≥ 4 pills/week). Facilitated recall to clinicians identified 99% of participants with protective adherence as measured by PBMC drug concentrations. Daily adherence measured by facilitated recall was associated with behavioural practices including group sex (aOR 1.33, 95% CI 1.15-1.53, p < 0.001). Retained participants maintained high adherence to daily PrEP over 12 months, confirmed by four different measures. Facilitated recall to clinicians is a suitable measure for assessing PrEP adherence in populations engaged in care where there is established trust and rapport with patients. Trial registration: ClinicalTrials.gov NCT02206555.
Collapse
|
103
|
Gandhi M, Bacchetti P, SpinelliI MA, Okochi H, Baeten JM, Siriprakaisil O, Klinbuayaem V, Rodrigues WC, Wang G, Vincent M, Cressey TR, Drain PK. Brief Report: Validation of a Urine Tenofovir Immunoassay for Adherence Monitoring to PrEP and ART and Establishing the Cutoff for a Point-of-Care Test. J Acquir Immune Defic Syndr 2019; 81:72-77. [PMID: 30664078 PMCID: PMC6456396 DOI: 10.1097/qai.0000000000001971] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Current pharmacologic adherence monitoring for antiretrovirals involves expensive, labor-intensive liquid chromatography/tandem mass spectrometry (LC-MS/MS)-based methods. Antibody-based assays can monitor and support adherence in real time. We developed a tenofovir (TFV)-based immunoassay and further validated it in a directly observed therapy (DOT) study. DESIGN Pharmacologic DOT study of TFV disoproxil fumarate (TDF)/emtricitabine (FTC) administered to HIV-noninfected volunteers. METHODS The TARGET study provided directly observed TDF 300 mg/FTC 200 mg 7 (high adherence), 4 (moderate), and 2 doses/week (low) to 30 volunteers (10/group) in Thailand, collecting a total of 637 urine samples over 6 weeks of administration and during washout. ELISA measured urine TFV levels by the immunoassay and LC-MS/MS-based concentrations served as the gold standard. A mixed-effects regression model evaluated cutoffs for a point-of-care assay. Performance characteristics of the immunoassay were compared with LC-MS/MS at a chosen cutoff. RESULTS Median TFV levels were 12,000 ng/mL by the immunoassay 1 day after dosing; 5000 ng/mL 2 days after dosing; 1500 ng/mL 3 days after dosing; and below the lower limit of quantification thereafter (≥4 days). An immunoassay cutoff of 1500 ng/mL accurately classified 98% of patients who took a dose 24 hours ago as adherent. The specificity and sensitivity of the immunoassay compared with LC-MS/MS at the 1500 ng/mL cutoff were 99% and 94%; the correlation between TFV levels by the 2 assays was high (0.92, P < 0.00001). CONCLUSIONS We have developed a novel TFV immunoassay that is highly specific, sensitive, and correlates strongly with LC-MS/MS measurements in a large DOT study. Adherence benchmarks from this DOT study will guide the development of a low-cost rapid point-of-care test for pre-exposure prophylaxis and antiretroviral treatment adherence monitoring and interventions.
Collapse
Affiliation(s)
- Monica Gandhi
- Division of HIV, Infectious Disease, and Global Medicine, Department of Medicine, University of California, San Francisco (UCSF), San Francisco, CA
| | - Peter Bacchetti
- Department of Epidemiology and Biostatistics, UCSF, San Francisco, CA
| | - Matthew A. SpinelliI
- Division of HIV, Infectious Disease, and Global Medicine, Department of Medicine, University of California, San Francisco (UCSF), San Francisco, CA
| | - Hideaki Okochi
- Department of Bioengineering and Therapeutic Sciences, UCSF, San Francisco, CA
| | - Jared M. Baeten
- Departments of Global Health, Medicine and Epidemiology, University of Washington, Seattle, WA
| | | | | | - Warren C. Rodrigues
- Alere Rapid Diagnostics/Abbott Rapid Diagnostics Division (ARDx), Pomona, CA
| | - Guohang Wang
- Alere Rapid Diagnostics/Abbott Rapid Diagnostics Division (ARDx), Pomona, CA
| | - Michael Vincent
- Alere Rapid Diagnostics/Abbott Rapid Diagnostics Division (ARDx), Pomona, CA
| | - Tim R. Cressey
- Department of Medicine, Chiang Mai University, Chiang Mai, Thailand; Department of Immunology & Infectious Diseases, Boston, Harvard T.H Chan School of Public Health, MA, USA; Department of Molecular & Clinical Pharmacology, University of Liverpool, UK
| | - Paul K. Drain
- Departments of Global Health, Medicine and Epidemiology, University of Washington, Seattle, WA
| |
Collapse
|
104
|
McGowan I, Wilkin T, Landovitz RJ, Wu C, Chen Y, Marzinke MA, Hendrix CW, Richardson P, Eshleman SH, Andrade A, Chege W, Anderson PL, McCauley M, Farley J, Mayer KH, Anton P, Brand RM, Cranston RD, Gulick R. The pharmacokinetics, pharmacodynamics, and mucosal responses to maraviroc-containing pre-exposure prophylaxis regimens in MSM. AIDS 2019; 33:237-246. [PMID: 30557160 DOI: 10.1097/qad.0000000000002038] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE HIV Prevention Trials Network 069/AIDS Clinical Trials Group A5305 was a study of 48-week oral pre-exposure prophylaxis (PrEP) regimens in MSM and transgender women. A rectal substudy was included to evaluate drug concentrations in rectal compartment vs. blood, gut-associated lymphoid tissue (GALT) responses to four antiretroviral PrEP regimens [maraviroc (MVC), MVC + emtricitabine (FTC), MVC + tenofovir (TFV) disoproxil fumarate, and TFV disoproxil fumarate + FTC], and to determine whether ARV exposure was associated with ex-vivo suppression of HIV infection in colorectal explants. METHODS C-C chemokine receptor type 5 (CCR5) genotype was characterized using PCR. At baseline and at Weeks 24, 48, and 49, GALT phenotype was characterized by flow cytometry, rectal biopsies were challenged with HIV-1BaL, and tissue and plasma pharmacokinetics were measured via mass spectrometry. RESULTS Exposure to MVC was not associated with increased expression of CD4+/CCR5+ HIV target T cells. Significant ex-vivo viral suppression compared with baseline was seen at Weeks 24 and 48, ranging from 1.4 to 1.8 log10 for all study regimens except the MVC-alone arm which did not show statistically significant viral suppression at Week 48. Tissue concentrations of TFV, TFV-diphosphate, and FTC were correlated with viral suppression. CONCLUSION MVC-containing HIV PrEP regimens did not increase GALT CD4+ T-cell activation or the CD4+/CCR5+ phenotype. No virologic suppression was seen with MVC-alone at Week 48 compared with combination regimens, suggesting MVC monotherapy might be less effective than combination antiretroviral PrEP regimens.
Collapse
|
105
|
Wheeler DP, Fields SD, Beauchamp G, Chen YQ, Emel LM, Hightow‐Weidman L, Hucks‐Ortiz C, Kuo I, Lucas J, Magnus M, Mayer KH, Nelson LE, Hendrix CW, Piwowar‐Manning E, Shoptaw S, Watkins P, Watson CC, Wilton L. Pre-exposure prophylaxis initiation and adherence among Black men who have sex with men (MSM) in three US cities: results from the HPTN 073 study. J Int AIDS Soc 2019; 22:e25223. [PMID: 30768776 PMCID: PMC6376611 DOI: 10.1002/jia2.25223] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 11/23/2018] [Indexed: 11/05/2022] Open
Abstract
INTRODUCTION Randomized clinical trials have demonstrated the efficacy of antiretroviral pre-exposure prophylaxis (PrEP) in preventing HIV acquisition among men who have sex with men (MSM). However, limited research has examined initiation and adherence to PrEP among Black MSM (BMSM) in the United States (US) who are disproportionately represented among newly HIV infected and late to care individuals. This research reports on the HIV Prevention Trials Network 073 (HPTN 073) study aimed to examine PrEP initiation, utilization and adherence among Black MSM utilizing the theoretically principled, culturally informed and client-centered care coordination (C4) model. METHODS The HPTN 073 study enrolled and followed 226 HIV-uninfected Black MSM in three US cities (Los Angeles, CA; Washington DC; and Chapel Hill, NC) from February 2013 through September 2015. Study participants were offered once daily oral emtricitabine/tenofovir (FTC/TDF) PrEP combined with C4 and followed up for 52 weeks. Participants received HIV testing, risk reduction education and clinical monitoring. RESULTS Of the 226 men enrolled, 178 participants initiated PrEP (79%), and of these 64% demonstrated PrEP utilization at week 26 (mid-point of the study) based on pharmacokinetic testing. Condomless anal sex with an HIV-infected or unknown status casual male partner was statistically significantly associated with a greater likelihood of PrEP initiation (adjusted odds ratio (OR) 4.4, 95% confidence interval (CI) 1.7, 11.7). Greater age (≥25 vs. <25, OR 2.95, 95% CI 1.37 -6.37), perception of having enough money (OR 3.6, 95% CI 1.7 to 7.7) and knowledge of male partner taking PrEP before sex (OR 2.22, 95% CI 1.03 to 4.79) were statistically significantly associated with increased likelihood of PrEP adherence at week 26. Annualized HIV incidence was 2.9 (95% CI 1.2 to 7.9) among those who initiated PrEP, compared to 7.7 (95% CI 2.5 to 24.1) among those who did not initiate PrEP (p = 0.18). CONCLUSIONS Results suggest a high level of PrEP initiation among at-risk Black MSM, a group historically characterized as hard to reach. The data support the importance of addressing contextual factors that affect PrEP initiation and adherence, and of additional research on the ultimate benefit of PrEP in HIV prevention among Black MSM.
Collapse
Affiliation(s)
| | - Sheldon D Fields
- School of Health ProfessionsNew York Institute of TechnologyNew YorkNYUSA
| | - Geetha Beauchamp
- Statistical Center for HIV/AIDS Research & Prevention (SCHARP)Vaccine and Infectious Disease Division (VIDD)Fred Hutchinson Cancer Research CenterSeattleWAUSA
| | - Ying Q Chen
- Statistical Center for HIV/AIDS Research & Prevention (SCHARP)Vaccine and Infectious Disease Division (VIDD) and Public Health Sciences DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
| | - Lynda M Emel
- Statistical Center for HIV/AIDS Research & Prevention (SCHARP)Vaccine and Infectious Disease Division (VIDD)Fred Hutchinson Cancer Research CenterSeattleWAUSA
| | - Lisa Hightow‐Weidman
- Division of Infectious DiseasesUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | | | - Irene Kuo
- Department of Epidemiology and BiostatisticsMilken Institute School of Public HealthGeorge Washington UniversityWashingtonDCUSA
| | | | - Manya Magnus
- Department of Epidemiology and BiostatisticsMilken Institute School of Public HealthGeorge Washington UniversityWashingtonDCUSA
| | - Kenneth H Mayer
- The Fenway InstituteFenway HealthBostonMAUSA
- Harvard Medical SchoolBostonMAUSA
- Beth Israel Deaconess Medical CenterBostonMAUSA
| | - LaRon E Nelson
- School of NursingUniversity of RochesterRochesterNYUSA
- Centre for Urban Health SolutionsLi Ka Shing Knowledge InstituteSt. Michael's HospitalTorontoONCanada
| | - Craig W Hendrix
- Department of Medicine (Clinical Pharmacology)John Hopkins School of MedicineBaltimoreMDUSA
| | - Estelle Piwowar‐Manning
- Department of PathologyJohn Hopkins School of MedicineBaltimoreMDUSA
- Present address:
CRS director for the GWU site and HPTN Black Caucus Chair
| | - Steven Shoptaw
- Department of Family MedicineDavid Geffen School of MedicineUniversity of California Los Angeles (UCLA)Los AngelesCAUSA
| | | | | | - Leo Wilton
- Department of Human DevelopmentState University of New York at BinghamtonBinghamtonNYUSA
- Faculty of HumanitiesUniversity of JohannesburgJohannesburgSouth Africa
| |
Collapse
|
106
|
Aung WS, Bakshi RP, Breakey J, Johnson JE, Hendrix CW, Weld E, Fuchs EJ, Marzinke MA. Fecal Coliform Bacterial Detection to Assess Enema Adherence in HIV Prevention Clinical Studies. AIDS Behav 2019; 23:252-258. [PMID: 29971733 DOI: 10.1007/s10461-018-2211-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Evaluating the efficacy of any HIV prevention strategy is dependent on ensuring and objectively monitoring adherence to the intervention. Medicated rectal enemas are a potential method for providing topical, episodic HIV prophylaxis during receptive anal intercourse. Assessing adherence to recommended enema dosing regimens is essential in evaluating the utility of this strategy. We utilized fecal coliform bacteria on used enema tips as a marker for enema use. Enema tip coliforms were tested by repurposing a microtiter plate-based water quality test designed to detect fecal contamination of water. Coliform detection occurred with 100% sensitivity and specificity when tips were assayed on day of use. The assay performed well post-7 day sample storage at room temperature, yielding a sensitivity of 80% and specificity of 93%. All (n = 64) samples collected in a subset of the DREAM-01 rectal microbicide enema clinical trial tested positive, even when tips were evaluated > 7 days post-reported use. The coliform-based enema tip assay allows monitoring of adherence in interventions involving rectal enemas in a sensitive, specific and inexpensive manner. The test performs well in clinical trial settings.
Collapse
|
107
|
Patel RR, Crane JS, López J, Chan PA, Liu AY, Tooba R, James AS. Pre-exposure prophylaxis for HIV prevention preferences among young adult African American men who have sex with men. PLoS One 2018; 13:e0209484. [PMID: 30592756 PMCID: PMC6310267 DOI: 10.1371/journal.pone.0209484] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 12/06/2018] [Indexed: 11/18/2022] Open
Abstract
Pre-exposure prophylaxis (PrEP) is effective in preventing HIV infections among men who have sex with men (MSM). PrEP uptake and adherence remain low and product preferences are unknown, especially among young African American MSM who are most at-risk. We conducted 26 qualitative interviews from 2014-2016 among young adult HIV-negative African American MSM regarding PrEP product preferences in Missouri. While the pill and injectable were most liked of all modalities, about a quarter preferred rectal products or patches. Most participants preferred a long-acting injectable (LAI) to daily oral pills due to better medication adherence and a dislike for taking pills. Many participants preferred daily oral pills to on-demand oral PrEP due to the inability to predict sex and the perception that insufficient time or medication would not achieve HIV protection with on-demand. A fear of needles and the perception that there would not be therapeutic levels for a long duration were concerns with injectable PrEP. Study findings highlight the need for a range of prevention options for African American MSM and can inform PrEP product development as well as dissemination and implementation efforts.
Collapse
Affiliation(s)
- Rupa R. Patel
- Division of Infectious Diseases, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - John S. Crane
- Division of Infectious Diseases, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Julia López
- Department of Obstetrics and Gynecology, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Philip A. Chan
- Division of Infectious Diseases, Brown University, Providence, Rhode Island, United States of America
| | - Albert Y. Liu
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- Bridge HIV, San Francisco Department of Public Health, San Francisco, California, United States of America
| | - Rubabin Tooba
- Washington University School of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Aimee S. James
- Division of Public Health Sciences, Washington University in St. Louis, St. Louis, Missouri, United States of America
| |
Collapse
|
108
|
Hendrix CW. HIV Antiretroviral Pre-Exposure Prophylaxis: Development Challenges and Pipeline Promise. Clin Pharmacol Ther 2018; 104:1082-1097. [PMID: 30199098 PMCID: PMC6925668 DOI: 10.1002/cpt.1227] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 08/20/2018] [Indexed: 12/17/2022]
Abstract
The US Food and Drug Administration (FDA) approved oral daily tenofovir/emtricitabine (Truvada) for pre-exposure prophylaxis of human immunodeficiency virus (HIV) infection in 2012 on the basis of two randomized controlled trials (RCTs), one in men who have sex with men (MSM) and another in HIV serodiscordant heterosexual couples. Subsequently, even greater efficacy has been demonstrated in MSM with rapid population-level incidence reductions in some locations. In contrast, studies of antiretroviral pre-exposure prophylaxis (PrEP) in heterosexual women showed only modest or no efficacy, largely attributed to low adherence. The mixed results of antiretroviral-based PrEP bear witness to unique drug development challenges at this complicated intersection of sexual behavior, public health, and drug development. Multiple innovative methods and formulation strategies followed to address unmet medical needs of persons struggling with daily oral PrEP adherence or preference for nonsystemic PrEP options. Clinical pharmacology plays essential roles throughout this PrEP development process, especially in early product development and through pharmacologically informed enhancement and interpretation of clinical trials.
Collapse
Affiliation(s)
- Craig W Hendrix
- 1Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
109
|
Cohen SE, Sachdev D, Lee SA, Scheer S, Bacon O, Chen MJ, Okochi H, Anderson PL, Kearney MF, Coffey S, Scott H, Grant RM, Havlir D, Gandhi M. Acquisition of tenofovir-susceptible, emtricitabine-resistant HIV despite high adherence to daily pre-exposure prophylaxis: a case report. Lancet HIV 2018; 6:S2352-3018(18)30288-1. [PMID: 30503324 PMCID: PMC6541554 DOI: 10.1016/s2352-3018(18)30288-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/27/2018] [Accepted: 10/03/2018] [Indexed: 04/08/2023]
Abstract
BACKGROUND Pre-exposure prophylaxis (PrEP) with emtricitabine and tenofovir disoproxil fumarate is highly protective against HIV infection. We report a case of tenofovir-susceptible, emtricitabine-resistant HIV acquisition despite high adherence to daily PrEP. METHODS Adherence to PrEP was assessed by measuring concentrations of emtricitabine and tenofovir disoproxil fumarate or their metabolites in plasma, dried blood spots, and hair. After seroconversion, genotypic and phenotypic resistance of the acquired virus was determined by standard clinical tests and by single-genome sequencing of proviral genomes. HIV partner services identified the likely transmission partner. FINDINGS A 21-year-old Latino man tested positive for HIV infection 13 months after PrEP initiation. He had a negative HIV antibody test, but detectable HIV RNA with 559 copies per mL. He reported good adherence to daily PrEP. He was linked to care and immediately started antiretroviral therapy, at which point his RNA was 1544 copies per mL and his HIV antibody test was positive. The HIV genotype revealed Met184Val, Leu74Val, Leu100Ile, and Lys103Asn mutations in reverse transcriptase, and the phenotype showed susceptibility to tenofovir disoproxil fumarate and resistance to emtricitabine. Segmental hair analysis of tenofovir disoproxil fumarate concentrations measured in 1 cm segments of hair from the scalp indicated consistently high adherence to PrEP in each of the 6 months before HIV diagnosis (0·0672-0·0889 ng/mg). Concentrations of tenofovir diphosphate (1012 fmol per punch) and emtricitabine triphosphate (0·266 fmol per punch) in a dried blood spot indicated high adherence over the preceding 6 weeks. Concentrations of emtricitabine (870·5 ng/mL) and tenofovir disoproxil fumarate (188·2 ng/mL) measured in plasma 3 months before HIV seroconversion confirmed adherence in the days preceding that visit. The likely transmission partner was not engaged in HIV primary care and had a similar viral genotype. INTERPRETATION Acquisition of HIV virus that is susceptible to tenofovir disoproxil fumarate, but resistant to emtricitabine can occur despite high adherence to PrEP. Quarterly screening for HIV and sexually transmitted diseases facilitates early diagnosis in people on PrEP; when combined with prompt linkage to care and partner services this can prevent onward transmission of HIV. FUNDING US National Institutes of Health.
Collapse
Affiliation(s)
- Stephanie E Cohen
- Disease Prevention and Control Branch, San Francisco Department of Public Health, San Francisco, CA, USA.
| | - Darpun Sachdev
- Disease Prevention and Control Branch, San Francisco Department of Public Health, San Francisco, CA, USA
| | - Sulggi A Lee
- Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Susan Scheer
- HIV Epidemiology Section, San Francisco Department of Public Health, San Francisco, CA, USA
| | - Oliver Bacon
- Disease Prevention and Control Branch, San Francisco Department of Public Health, San Francisco, CA, USA
| | - Miao-Jung Chen
- HIV Epidemiology Section, San Francisco Department of Public Health, San Francisco, CA, USA
| | - Hideaki Okochi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Peter L Anderson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Mary F Kearney
- HIV Dynamics and Replication Program, National Cancer Institute at Frederick, Frederick, MD, USA
| | - Susa Coffey
- Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Hyman Scott
- Bridge HIV, San Francisco Department of Public Health, San Francisco, CA, USA; Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Robert M Grant
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Diane Havlir
- Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Monica Gandhi
- Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
110
|
Mboup A, Béhanzin L, Guédou FA, Geraldo N, Goma‐Matsétsé E, Giguère K, Aza‐Gnandji M, Kessou L, Diallo M, Kêkê RK, Bachabi M, Dramane K, Geidelberg L, Cianci F, Lafrance C, Affolabi D, Diabaté S, Gagnon M, Zannou DM, Gangbo F, Boily M, Vickerman P, Alary M. Early antiretroviral therapy and daily pre-exposure prophylaxis for HIV prevention among female sex workers in Cotonou, Benin: a prospective observational demonstration study. J Int AIDS Soc 2018; 21:e25208. [PMID: 31291057 PMCID: PMC6287093 DOI: 10.1002/jia2.25208] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/19/2018] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION In sub-Saharan Africa, HIV prevalence remains high, especially among key populations. In such situations, combination prevention including clinical, behavioural, structural and biological components, as well as adequate treatment are important. We conducted a demonstration project at the Dispensaire IST, a clinic dedicated to female sex workers (FSWs) in Cotonou, on early antiretroviral therapy (E-ART, or immediate "test-and-treat") and pre-exposure prophylaxis (PrEP). We present key indicators such as uptake, retention and adherence. METHODS In this prospective observational study, we recruited FSWs from October 4th 2014 to December 31st 2015 and followed them until December 31st 2016. FSWs were provided with daily tenofovir disoproxil fumarate/emtricitabine (Truvada® ) for PrEP or received a first-line antiretroviral regimen as per Benin guidelines. We used generalized estimating equations to assess trends in adherence and sexual behaviour. RESULTS Among FSWs in the catchment area, HIV testing coverage within the study framework was 95.5% (422/442). At baseline, HIV prevalence was 26.3% (111/422). Among eligible FSWs, 95.5% (105/110) were recruited for E-ART and 88.3% (256/290) for PrEP. Overall retention at the end of the study was 59.0% (62/105) for E-ART and 47.3% (121/256) for PrEP. Mean (±SD) duration of follow-up was 13.4 (±7.9) months for E-ART and 11.8 (±7.9) months for PrEP. Self-reported adherence was over 90% among most E-ART participants. For PrEP, adherence was lower and the proportion with 100% adherence decreased over time from 78.4% to 56.7% (p-trend < 0.0001). During the 250.1 person-years of follow-up among PrEP initiators, two seroconversions occurred (incidence 0.8/100 person-years (95% confidence interval: 0.3 to 1.9/100 person-years)). The two seroconverters had stopped using PrEP for at least six months before being found HIV-infected. In both groups, there was no evidence of reduced condom use. CONCLUSIONS This study provides data on key indicators for the integration of E-ART and PrEP into the HIV prevention combination package already offered to FSWs in Benin. PrEP may be more useful as an individual intervention for adherent FSWs rather than a specific public health intervention. E-ART was a more successful intervention in terms of retention and adherence and is now offered to all key populations in Benin. STUDY REGISTRATION ClinicalTrials.gov NCT02237.
Collapse
Affiliation(s)
- Aminata Mboup
- Département de médecine sociale et préventiveUniversité LavalQuébecQCCanada
- Axe Santé des populations et pratiques optimales en santéCentre de recherche du CHU de Québec – Université LavalQuébecQCCanada
| | - Luc Béhanzin
- Axe Santé des populations et pratiques optimales en santéCentre de recherche du CHU de Québec – Université LavalQuébecQCCanada
- Dispensaire ISTCentre de santé communal de Cotonou 1CotonouBénin
- École Nationale de Formation des Techniciens Supérieurs en Santé Publique et en Surveillance ÉpidémiologiqueUniversité de ParakouParakouBénin
| | - Fernand A Guédou
- Axe Santé des populations et pratiques optimales en santéCentre de recherche du CHU de Québec – Université LavalQuébecQCCanada
- Dispensaire ISTCentre de santé communal de Cotonou 1CotonouBénin
| | - Nassirou Geraldo
- Dispensaire ISTCentre de santé communal de Cotonou 1CotonouBénin
| | | | - Katia Giguère
- Département de médecine sociale et préventiveUniversité LavalQuébecQCCanada
- Axe Santé des populations et pratiques optimales en santéCentre de recherche du CHU de Québec – Université LavalQuébecQCCanada
| | | | - Léon Kessou
- Service de Consultance et Expertise Nouvelle en Afrique (SCEN AFRIK)CotonouBénin
| | - Mamadou Diallo
- Département de médecine sociale et préventiveUniversité LavalQuébecQCCanada
- Axe Santé des populations et pratiques optimales en santéCentre de recherche du CHU de Québec – Université LavalQuébecQCCanada
| | - René K Kêkê
- Programme Santé de Lutte contre le Sida (PSLS)CotonouBénin
| | - Moussa Bachabi
- Programme Santé de Lutte contre le Sida (PSLS)CotonouBénin
| | - Kania Dramane
- Laboratoire de virologie du Centre MurazBobo‐DioulassoBurkina Faso
| | - Lily Geidelberg
- Department of infectious diseaseImperial College LondonLondonUK
| | - Fiona Cianci
- University of BristolBristolUK
- London School of Hygiene and Tropical MedicineLondonUK
| | - Christian Lafrance
- Axe Santé des populations et pratiques optimales en santéCentre de recherche du CHU de Québec – Université LavalQuébecQCCanada
| | - Dissou Affolabi
- Faculté des sciences de la santéUniversité d'Abomey‐CalaviCotonouBénin
- Centre national hospitalier universitaire HMK de CotonouCotonouBénin
| | - Souleymane Diabaté
- Département de médecine sociale et préventiveUniversité LavalQuébecQCCanada
- Axe Santé des populations et pratiques optimales en santéCentre de recherche du CHU de Québec – Université LavalQuébecQCCanada
- Université Alassane OuattaraBouakéCôte d'Ivoire
| | - Marie‐Pierre Gagnon
- Axe Santé des populations et pratiques optimales en santéCentre de recherche du CHU de Québec – Université LavalQuébecQCCanada
- Faculté des sciences infirmièresUniversité LavalQuébecQCCanada
| | - Djimon M Zannou
- Faculté des sciences de la santéUniversité d'Abomey‐CalaviCotonouBénin
- Centre national hospitalier universitaire HMK de CotonouCotonouBénin
| | - Flore Gangbo
- Programme Santé de Lutte contre le Sida (PSLS)CotonouBénin
- Faculté des sciences de la santéUniversité d'Abomey‐CalaviCotonouBénin
- Centre national hospitalier universitaire HMK de CotonouCotonouBénin
| | - Marie‐Claude Boily
- Axe Santé des populations et pratiques optimales en santéCentre de recherche du CHU de Québec – Université LavalQuébecQCCanada
- Department of infectious diseaseImperial College LondonLondonUK
| | | | - Michel Alary
- Département de médecine sociale et préventiveUniversité LavalQuébecQCCanada
- Axe Santé des populations et pratiques optimales en santéCentre de recherche du CHU de Québec – Université LavalQuébecQCCanada
- Institut national de santé publique du QuébecQuébecQCCanada
| |
Collapse
|
111
|
Zhang Y, Sivay MV, Hudelson SE, Clarke W, Breaud A, Wang J, Piwowar-Manning E, Agyei Y, Fogel JM, Hamilton EL, Selin A, MacPhail C, Kahn K, Gomez-Olive FX, Hughes JP, Pettifor A, Eshleman SH. Antiretroviral Drug Use and HIV Drug Resistance Among Young Women in Rural South Africa: HPTN 068. J Acquir Immune Defic Syndr 2018; 79:315-322. [PMID: 29985265 PMCID: PMC6185777 DOI: 10.1097/qai.0000000000001793] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Antiretroviral (ARV) drugs are used for HIV treatment and prevention. We analyzed ARV drug use and HIV drug resistance in a cohort of young women in rural South Africa enrolled in the HIV Prevention Trials Network (HPTN) 068 study, which evaluated the use of a cash transfer conditional on school attendance to reduce HIV incidence. METHODS ARV drug testing was performed using plasma samples from 2526 young women. This included 2526 enrollment samples (80 HIV-infected and 2446 HIV-uninfected) and 162 seroconversion samples (first HIV-positive study visit). Testing was performed using a qualitative assay that detects 20 ARV drugs from 5 drug classes. HIV drug resistance testing was performed with the ViroSeq HIV-1 Genotyping System for samples that had HIV viral loads ≥400 copies per milliliter. RESULTS At enrollment, ARV drugs were detected in 10 (12.5%) of 80 HIV-infected young women. None of 2446 HIV-uninfected young women had ARV drugs detected at enrollment. ARV drugs were also detected in 16 (9.9%) of 162 seroconverters. At enrollment, 9 (13.4%) of 67 young women with genotyping results had HIV drug resistance; resistance was also detected in 9 (6.9%) of 131 seroconverters with genotyping results. CONCLUSIONS Most of the HIV-infected young women in this cohort from rural South Africa were not taking ARV drugs, suggesting they were unaware of their HIV status or were not in care. HIV drug resistance was detected in young women with both prevalent and new HIV infection.
Collapse
Affiliation(s)
- Yinfeng Zhang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mariya V. Sivay
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah E. Hudelson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - William Clarke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Autumn Breaud
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jing Wang
- Statistical Center for HIV/AIDS Research & Prevention (SCHARP), Seattle, WA, USA
| | | | - Yaw Agyei
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jessica M. Fogel
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Amanda Selin
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Catherine MacPhail
- School of Health and Society, University of Wollongong, Australia; MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Kathleen Kahn
- MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - F. Xavier Gomez-Olive
- MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - James P. Hughes
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Audrey Pettifor
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC; MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Susan H. Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
112
|
Seneviratne HK, Hendrix CW, Fuchs EJ, Bumpus NN. MALDI Mass Spectrometry Imaging Reveals Heterogeneous Distribution of Tenofovir and Tenofovir Diphosphate in Colorectal Tissue of Subjects Receiving a Tenofovir-Containing Enema. J Pharmacol Exp Ther 2018; 367:40-48. [PMID: 30037813 PMCID: PMC6123665 DOI: 10.1124/jpet.118.250357] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/18/2018] [Indexed: 01/21/2023] Open
Abstract
Efforts to prevent human immunodeficiency virus (HIV) infection via pre-exposure prophylaxis (PrEP) include the development of anti-HIV drugs as microbicides for topical application to the mucosal sites of infection; however, although understanding the distribution profiles of these drugs in target mucosal tissues is of critical importance to guiding their optimization, data in this regard are largely lacking. With this in mind, we developed a matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) approach to visualize tenofovir (TFV), an HIV nucleotide analog reverse-transcriptase inhibitor under investigation for use as a topical microbicide, and its active metabolite TFV-diphosphate (TFV-DP) in colorectal biopsies obtained from healthy volunteers who received TFV-containing enemas. Application of MALDI MSI resulted in sufficient spatial resolution to visualize both TFV and TFV-DP and revealed heterogeneity in the distribution profiles of both analytes, including the presence of regions in which TFV and TFV-DP were undetectable, in colorectal tissue at two different time points and concentrations. Cell-specific staining for CD4 T and CD11c dendritic cells, which are important to the establishment of HIV infection, demonstrated that the TFV and TFV-DP distributions were independent of these cell types. MALDI MSI of endogenous lipids demonstrated that the heterogeneity observed for TFV and TFV-DP was not a function of tissue composition or processing. These data provide unique insight into the spatial distribution of TFV and TFV-DP in human colorectal tissue. In addition, this work establishes an approach that can be leveraged to directly detect and visualize these clinically important analytes more broadly in tissue.
Collapse
Affiliation(s)
- Herana Kamal Seneviratne
- Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Craig W Hendrix
- Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Edward J Fuchs
- Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Namandjé N Bumpus
- Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
113
|
Comparison of short messaging service self-reported adherence with other adherence measures in a demonstration project of HIV preexposure prophylaxis in Kenya and Uganda. AIDS 2018; 32:2237-2245. [PMID: 30005025 PMCID: PMC6755137 DOI: 10.1097/qad.0000000000001955] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVE Short messaging service (SMS) can collect adherence data on a frequent basis and is relatively anonymous, and therefore could potentially reduce recall and social desirability biases prevalent in other self-reported measures. METHODS We compared SMS self-reported adherence with three self-reported adherence questions (rating of ability to adhere, frequency of doses taken, percentage of doses taken) and two objective adherence measures [electronic adherence monitoring (EAM) and plasma tenofovir levels] using data from HIV-uninfected members of serodiscordant couples enrolled in a preexposure prophylaxis demonstration project in Kenya and Uganda. RESULTS Of 373 enrolled participants, 256 (69%) were male and median age at enrolment was 29 years (26, 35). Fifty-two percent were from Kenya and median education at enrolment was 10 years (7,12). Overall, median adherence was 90, 75, 85, 94 and 79%, respectively, for self-report by SMS, rating, frequency, percentage and EAM adherence. Spearman's correlation coefficient between SMS and interviewer-administered self-reported measures was 0.18 for rating and frequency, 0.22 for percentage and 0.14 for EAM (all P < 0.001). The estimated difference in average adherence between SMS and self-reported rating, frequency, percentage adherence and EAM was 8.1 (P < 0.001), 0.3 (P = 0.81), -5.2 (P < 0.001) and 9.5 (P < 0.001), respectively. Area under the receiver-operating curve assessing the ability of SMS self-report to discriminate between detectable and undetectable tenofovir was 0.51. CONCLUSION Our study found low correlation between SMS self-report and other self-reported and objective adherence measures and did not discriminate between detectable and undetectable plasma tenofovir levels. Future use of SMS self-report should explore alternative means for reducing potential biases.
Collapse
|
114
|
Koss CA, Liu AY, Castillo-Mancilla J, Bacchetti P, McHugh C, Kuncze K, Morrow M, Louie A, Seifert S, Okochi H, MaWhinney S, Gandhi M, Anderson PL. Similar tenofovir hair concentrations in men and women after directly observed dosing of tenofovir disoproxil fumarate/emtricitabine: implications for preexposure prophylaxis adherence monitoring. AIDS 2018; 32:2189-2194. [PMID: 30212404 PMCID: PMC6330141 DOI: 10.1097/qad.0000000000001935] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Women likely require higher adherence than men to preexposure prophylaxis (PrEP) with tenofovir disoproxil fumarate (TDF)/emtricitabine (FTC) for similar efficacy. Pharmacologic metrics of adherence predict efficacy better than self-report, but expected drug levels (adherence benchmarks) must be established using directly observed therapy. We sought to evaluate whether tenofovir hair concentrations differ between women and men receiving directly observed TDF/FTC. METHODS We assessed tenofovir hair concentrations in HIV-uninfected volunteers randomized to receive 100%, 67%, or 33% of daily dosing of TDF/FTC for 12 weeks (DOT-DBS, NCT02022657). Hair samples were collected at dosing weeks 4, 8, and 12 and every 3 weeks during a 12-week washout. Tenofovir concentrations in the proximal 1.5 cm of hair (representing ∼6 weeks of exposure) were analyzed using liquid chromatography/tandem mass spectrometry. Linear regression was used to model tenofovir hair concentrations in terms of sex, doses over the prior 6 weeks, and number of days since last dose. RESULTS A total of 264 hair samples were analyzed from 23 female and 24 male participants. Female participants had similar tenofovir hair concentrations to men (estimated fold-difference 0.92, 95% CI 0.75-1.13, P = 0.43). The estimated fold-difference in tenofovir levels for female versus male participants did not appreciably change when age (0.93, 95% CI 0.76-1.15), weight (0.89, 95% CI 0.71-1.11), or race/ethnicity (0.95, 95% CI 0.77-1.17) were added to the model. CONCLUSION Women and men have similar adherence benchmarks for tenofovir in hair samples. As pharmacokinetic metrics are increasingly used for PrEP monitoring, these findings provide guidance for assessing adherence via hair concentrations.
Collapse
Affiliation(s)
- Catherine A Koss
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, California
| | - Albert Y Liu
- Bridge HIV, San Francisco Department of Public Health, California
| | - Jose Castillo-Mancilla
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado
| | - Peter Bacchetti
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Cricket McHugh
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus
| | - Karen Kuncze
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, California
| | - Mary Morrow
- Department of Biostatistics and Informatics, University of Colorado, Colorado School of Public Health, Colorado
| | - Alexander Louie
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, California
| | - Sharon Seifert
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus
| | - Hideaki Okochi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California, San Francisco, California, USA
| | - Samantha MaWhinney
- Department of Biostatistics and Informatics, University of Colorado, Colorado School of Public Health, Colorado
| | - Monica Gandhi
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, California
| | - Peter L Anderson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus
| |
Collapse
|
115
|
Vincent KL, Moss JA, Marzinke MA, Hendrix CW, Anton PA, Pyles RB, Guthrie KM, Dawson L, Olive TJ, Butkyavichene I, Churchman SA, Cortez JM, Fanter R, Gunawardana M, Miller CS, Yang F, Rosen RK, Vargas SE, Baum MM. Safety and pharmacokinetics of single, dual, and triple antiretroviral drug formulations delivered by pod-intravaginal rings designed for HIV-1 prevention: A Phase I trial. PLoS Med 2018; 15:e1002655. [PMID: 30265679 PMCID: PMC6161852 DOI: 10.1371/journal.pmed.1002655] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 08/17/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Intravaginal rings (IVRs) for HIV pre-exposure prophylaxis (PrEP) theoretically overcome some adherence concerns associated with frequent dosing that can occur with oral or vaginal film/gel regimens. An innovative pod-IVR, composed of an elastomer scaffold that can hold up to 10 polymer-coated drug cores (or "pods"), is distinct from other IVR designs as drug release from each pod can be controlled independently. A pod-IVR has been developed for the delivery of tenofovir (TFV) disoproxil fumarate (TDF) in combination with emtricitabine (FTC), as daily oral TDF-FTC is the only Food and Drug Administration (FDA)-approved regimen for HIV PrEP. A triple combination IVR building on this platform and delivering TDF-FTC along with the antiretroviral (ARV) agent maraviroc (MVC) also is under development. METHODOLOGY AND FINDINGS This pilot Phase I trial conducted between June 23, 2015, and July 15, 2016, evaluated the safety, pharmacokinetics (PKs), and acceptability of pod-IVRs delivering 3 different ARV regimens: 1) TDF only, 2) TDF-FTC, and 3) TDF-FTC-MVC over 7 d. The crossover, open-label portion of the trial (N = 6) consisted of 7 d of continuous TDF pod-IVR use, a wash-out phase, and 7 d of continuous TDF-FTC pod-IVR use. After a 3-mo pause to evaluate safety and PK of the TDF and TDF-FTC pod-IVRs, TDF-FTC-MVC pod-IVRs (N = 6) were evaluated over 7 d of continuous use. Safety was assessed by adverse events (AEs), colposcopy, and culture-independent analysis of the vaginal microbiome (VMB). Drug and drug metabolite concentrations in plasma, cervicovaginal fluids (CVFs), cervicovaginal lavages (CVLs), and vaginal tissue (VT) biopsies were determined via liquid chromatographic-tandem mass spectrometry (LC-MS/MS). Perceptibility and acceptability were assessed by surveys and interviews. Median participant age was as follows: TDF/TDF-FTC group, 26 y (range 24-35 y), 2 White, 2 Hispanic, and 2 African American; TDF-FTC-MVC group, 24.5 y (range 21-41 y), 3 White, 1 Hispanic, and 2 African American. Reported acceptability was high for all 3 products, and pod-IVR use was confirmed by residual drug levels in used IVRs. There were no serious adverse events (SAEs) during the study. There were 26 AEs reported during TDF/TDF-FTC IVR use (itching, discharge, discomfort), with no differences between TDF alone or in combination with FTC observed. In the TDF-FTC-MVC IVR group, there were 12 AEs (itching, discharge, discomfort) during IVR use regardless of attribution to study product. No epithelial disruption/thinning was seen by colposcopy, and no systematic VMB shifts were observed. Median (IQR) tenofovir diphosphate (TFV-DP) tissue concentrations of 303 (277-938) fmol/10(6) cells (TDF), 289 (110-603) fmol/10(6) cells (TDF-FTC), and 302 (177.1-823.8) fmol/10(6) cells (TDF-FTC-MVC) were sustained for 7 d, exceeding theoretical target concentrations for vaginal HIV prevention. The study's main limitations include the small sample size, short duration (7 d versus 28 d), and the lack of FTC triphosphate measurements in VT biopsies. CONCLUSIONS An innovative pod-IVR delivery device with 3 different formulations delivering different regimens of ARV drugs vaginally appeared to be safe and acceptable and provided drug concentrations in CVFs and tissues exceeding concentrations achieved by highly protective oral dosing, suggesting that efficacy for vaginal HIV PrEP is achievable. These results show that an alternate, more adherence-independent, longer-acting prevention device based on the only FDA-approved PrEP combination regimen can be advanced to safety and efficacy testing. TRIAL REGISTRATION ClinicalTrials.gov NCT02431273.
Collapse
Affiliation(s)
- Kathleen L. Vincent
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| | - John A. Moss
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| | - Mark A. Marzinke
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Craig W. Hendrix
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Peter A. Anton
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
- Center for HIV Prevention Research, Division of Digestive Diseases and UCLA AIDS Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Richard B. Pyles
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kate M. Guthrie
- The Centers for Behavioral & Preventive Medicine, The Miriam Hospital, Providence, Rhode Island, United States of America
- Department of Psychiatry & Human Behavior, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Lauren Dawson
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Trevelyn J. Olive
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Irina Butkyavichene
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| | - Scott A. Churchman
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| | - John M. Cortez
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| | - Rob Fanter
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| | - Manjula Gunawardana
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| | - Christine S. Miller
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| | - Flora Yang
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| | - Rochelle K. Rosen
- The Centers for Behavioral & Preventive Medicine, The Miriam Hospital, Providence, Rhode Island, United States of America
- Department of Behavioral & Social Sciences, Brown University School of Public Health, Providence, Rhode Island, United States of America
| | - Sara E. Vargas
- The Centers for Behavioral & Preventive Medicine, The Miriam Hospital, Providence, Rhode Island, United States of America
- Department of Psychiatry & Human Behavior, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Marc M. Baum
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| |
Collapse
|
116
|
Tenofovir and tenofovir-diphosphate concentrations during pregnancy among HIV-uninfected women using oral preexposure prophylaxis. AIDS 2018; 32:1891-1898. [PMID: 29894385 DOI: 10.1097/qad.0000000000001922] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVES Pregnancy is a time of increased HIV acquisition risk and pregnancy reduces concentrations of antiretrovirals used for treatment. We assessed whether pregnancy lowers concentrations of tenofovir (TFV) and tenofovir-diphosphate (TFV-DP) among HIV-uninfected women using oral preexposure prophylaxis (PrEP). METHODS We analyzed data from an open-label PrEP study, comparing concentrations of TFV in plasma and TFV-DP in dried blood spots (DBS) among 37 pregnant women and 97 nonpregnant women. Analyses controlled for adherence from daily electronic monitoring. RESULTS The average plasma concentration of TFV among pregnant women was 34.7 ng/ml with 22.2 average recorded doses over the prior month versus 86.5 ng/ml with 23.1 doses among nonpregnant women. After controlling for adherence, TFV concentrations were 58% lower among pregnant women, a statistically significant difference of -50.4 ng/ml (95% CI -68.3 to -32.5). The average TFV-DP concentration was 450.3 fmol/punch among pregnant women and 636.7 fmol/punch among nonpregnant women. This difference was not statistically significant after adjusting for adherence; however, among those with quantifiable TFV-DP, concentrations were 27% lower during pregnancy [-202 fmol/punch (95% CI -384 to -19)]. Among participants with samples before and during pregnancy, there were significant decreases during pregnancy, controlling for adherence: -28.1 ng/ml TFV (95% CI -52.3 to -4.0) and -289.2 fmol/punch TFV-DP (95% CI -439.0 to -139.3). CONCLUSION Consistent with studies among HIV-infected women on ART, we found TFV and TFV-DP concentrations were lower during pregnancy. There is no established TFV concentration threshold to achieve HIV prevention. Additional pharmacokinetic studies and studies of PrEP efficacy in pregnancy are needed.
Collapse
|
117
|
Vincent KL, Moss JA, Marzinke MA, Hendrix CW, Anton PA, Gunawardana M, Dawson LN, Olive TJ, Pyles RB, Baum MM. Phase I trial of pod-intravaginal rings delivering antiretroviral agents for HIV-1 prevention: Rectal drug exposure from vaginal dosing with tenofovir disoproxil fumarate, emtricitabine, and maraviroc. PLoS One 2018; 13:e0201952. [PMID: 30133534 PMCID: PMC6104940 DOI: 10.1371/journal.pone.0201952] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 05/22/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Intravaginal rings (IVRs) can deliver antiretroviral (ARV) agents for HIV pre-exposure prophylaxis (PrEP), theoretically overcoming adherence concerns associated with frequent dosing. However, topical vaginal ARV drug delivery has not simultaneously led to sufficient rectal drug exposure to likely protect from HIV infection as a result of receptive anal intercourse (RAI). Unprotected RAI has a higher risk of infection per sex act and, for women, also can be associated with vaginal exposure during a single sexual encounter, especially in higher-risk subsets of women. The physiologically inflamed, activated, immune-cell dense colorectal mucosa is increasingly appreciated as the sexual compartment with highly significant risk; this risk is increased in the setting of co-infections. Ex vivo studies have shown that colorectal tissue and rectal fluid concentrations correlated with HIV protection. Given these important results, efforts to document colorectal compartment ARV drug concentration from pod-IVR delivery was assessed to determine if vaginal application could provide protective ARV levels in both compartments. METHODOLOGY/PRINCIPAL FINDINGS A crossover clinical trial (N = 6) evaluated 7 d of continuous TDF pod-IVR use, a wash-out phase, followed by 7 d with a TDF-FTC pod-IVR. A subsequent clinical trial (N = 6) consisted of 7 d of continuous TDF-FTC-MVC pod-IVR use. Rectal fluids were collected on Day 7 at IVR removal in all three ARV-exposures (two Phase 1 trials) and drug concentrations quantified by LC-MS/MS. Median rectal fluid concentrations of TFV, the hydrolysis product of the prodrug TDF, were between 0.66 ng mg-1 (TDF pod-IVR group) and 1.11 ng mg-1 (TDF-FTC pod-IVR group), but below the analytical lower limit of quantitation in 5/6 samples in the TDF-FTC-MVC pod-IVR group. Unexpectedly, median FTC (TDF-FTC pod-IVR, 20.3 ng mg-1; TDF-FTC-MVC pod-IVR, 0.18 ng mg-1), and MVC rectal fluid concentrations (0.84 ng mg-1) were quantifiable and higher than their respective in vitro EC50 values in most samples. Due to participant burden in these exploratory trials, rectal fluid was used as a surrogate for rectal tissue, where drug concentrations are expected to be higher. CONCLUSIONS/SIGNIFICANCE The concentrations of FTC and MVC in rectal fluids obtained in two exploratory clinical trials of IVRs delivering ARV combinations exceeded levels associated with in vitro efficacy in HIV inhibition. Unexpectedly, MVC appeared to depress the distribution of TFV and FTC into the rectal lumen. Here we show that vaginal delivery of ARV combinations may provide adherence and coitally independent dual-compartment protection from HIV infection during both vaginal and receptive anal intercourse.
Collapse
Affiliation(s)
- Kathleen Listiak Vincent
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - John A. Moss
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| | - Mark A. Marzinke
- Department of Medicine (Division of Clinical Pharmacology), Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Pathology, Johns Hopkins University, Sheikh Zayed Tower, Baltimore, Maryland, United States of America
| | - Craig W. Hendrix
- Department of Medicine (Division of Clinical Pharmacology), Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Peter A. Anton
- Center for HIV Prevention Research, Division of Digestive Diseases and UCLA AIDS Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Manjula Gunawardana
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| | - Lauren N. Dawson
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Trevelyn J. Olive
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Richard B. Pyles
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Marc M. Baum
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| |
Collapse
|
118
|
Transcutaneously refillable nanofluidic implant achieves sustained level of tenofovir diphosphate for HIV pre-exposure prophylaxis. J Control Release 2018; 286:315-325. [PMID: 30092254 DOI: 10.1016/j.jconrel.2018.08.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/20/2018] [Accepted: 08/03/2018] [Indexed: 02/07/2023]
Abstract
Pre-exposure prophylaxis (PrEP) with antiretroviral (ARV) drugs are effective at preventing human immunodeficiency virus (HIV) transmission. However, implementation of PrEP presents significant challenges due to poor user adherence, low accessibility to ARVs and multiple routes of HIV exposure. To address these challenges, we developed the nanochannel delivery implant (NDI), a subcutaneously implantable device for sustained and constant delivery of tenofovir alafenamide (TAF) and emtricitabine (FTC) for HIV PrEP. Unlike existing drug delivery platforms with finite depots, the NDI incorporates ports allowing for transcutaneous refilling upon drug exhaustion. NDI-mediated drug delivery in rhesus macaques resulted in sustained release of both TAF and FTC for 83 days, as indicated by concentrations of TAF, FTC and their respectively metabolites in plasma, PBMCs, rectal mononuclear cells and tissues associated with HIV transmission. Notably, clinically relevant preventative levels of tenofovir diphosphate were achieved as early as 3 days after NDI implantation. We also demonstrated the feasibility of transcutaneous drug refilling to extend the duration of PrEP drug delivery in NHPs. Overall, the NDI represents an innovative strategy for long-term HIV PrEP administration in both developed and developing countries.
Collapse
|
119
|
Gandhi M, Bacchetti P, Rodrigues WC, Spinelli M, Koss CA, Drain PK, Baeten JM, Mugo NR, Ngure K, Benet LZ, Okochi H, Wang G, Vincent M. Development and Validation of an Immunoassay for Tenofovir in Urine as a Real-Time Metric of Antiretroviral Adherence. EClinicalMedicine 2018; 2-3:22-28. [PMID: 30906930 PMCID: PMC6428441 DOI: 10.1016/j.eclinm.2018.08.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Pharmacologic adherence measures were critical to the interpretation of the tenofovir (TFV)-disoproxil-fumarate/emtricitabine (TDF/FTC) PrEP trials. These measures are being incorporated into PrEP demonstration projects, but currently-available metrics in plasma, cells, hair or urine involve expensive and time-intensive mass-spectrometry (MS)-based methods. No point-of-care method to assess PrEP adherence in real-time has yet been implemented. Antibody-based tests allow for low-cost, easy-to-perform, point-of-care drug detection. In this study, we developed an antibody-based TFV immunoassay and evaluated its test characteristics among individuals taking TDF/FTC. METHODS We synthesized possible immunogens based on TFV's molecular structure, injected rabbits with the conjugated derivatives, and bled them monthly for subsequent ELISA-testing for TFV-specific antibodies. We purified an antibody with specific TFV binding and created dose-response curves for ELISA-quantification. We then quantified TFV in urine from human participants not taking TDF/FTC and from individuals taking daily TDF/FTC 300 mg/200 mg for 7 days with a 7-day washout period using ELISA with this TFV-specific antibody. ELISA results were compared with the gold-standard test for TFV detection/quantification using liquid-chromatography-tandem-MS (LC-MS/MS). FINDINGS None of the urine samples from 115 participants not taking TDF/FTC showed ELISA- reactivity, indicating 100% specificity (95% CI 97-100%) of the immunoassay. Among participants taking TDF/FTC, 67 of 70 samples positive by LC-MS/MS were positive by the ELISA-immunoassay for an estimated diagnostic sensitivity of 96% (95% CI 88-99%). The precision of the assay was high (coefficient of variationb15%). The rank correlation between ELISA and LC-MS/MS values in the 70 quantitative urine TFV levels positive by LC-MS/MS across a wide range of concentrations among participants on TDF/FTC was high (r = 0.96). INTERPRETATION Our antibody-based immunoassay for measuring TFV in urine performed well compared to the gold-standard of LC-MS/MS among individuals taking TDF/FTC. A sensitive and specific immunoassay paves the way for real-time monitoring/feedback on recent adherence to TFV-based regimens, which should optimize interpretation and outcomes during PrEP and ART roll-out.
Collapse
Affiliation(s)
- Monica Gandhi
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco (UCSF), 995 Potrero Avenue, Building 80, 4th floor, San Francisco, CA 94110, United States of America
- Corresponding author.
| | - Peter Bacchetti
- Department of Epidemiology and Biostatistics, University of California, San Francisco (UCSF), Box 0560, 550 16th Street, 2538, San Francisco, CA 94158, United States of America
| | - Warren C. Rodrigues
- Alere Rapid Diagnostics/Abbott Rapid Diagnostics Division (ARDx), 829 Towne Center Drive, Pomona, CA 91767, United States of America
| | - Matthew Spinelli
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco (UCSF), 995 Potrero Avenue, Building 80, 4th floor, San Francisco, CA 94110, United States of America
| | - Catherine A. Koss
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco (UCSF), 995 Potrero Avenue, Building 80, 4th floor, San Francisco, CA 94110, United States of America
| | - Paul K. Drain
- Department of Global Health, University of Washington, 325 Ninth Avenue, Box 359927, Seattle, WA 98104, United States of America
| | - Jared M. Baeten
- Department of Epidemiology, University of Washington, 325 Ninth Avenue, Box 359927, Seattle, WA 98104, United States of America
| | - Nelly R. Mugo
- Department of Global Health, University of Washington, 325 Ninth Avenue, Box 359927, Seattle, WA 98104, United States of America
- Center for Clinical Research, Kenya Medical Research Institute, Section 9, Nairobi, Kenya
| | - Kenneth Ngure
- Department of Global Health, University of Washington, 325 Ninth Avenue, Box 359927, Seattle, WA 98104, United States of America
- Department of Community Health, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Leslie Z. Benet
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), 513 Parnassus Avenue, San Francisco, CA 94143, United States of America
| | - Hideaki Okochi
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), 513 Parnassus Avenue, San Francisco, CA 94143, United States of America
| | - Guohong Wang
- Alere Rapid Diagnostics/Abbott Rapid Diagnostics Division (ARDx), 829 Towne Center Drive, Pomona, CA 91767, United States of America
| | - Michael Vincent
- Alere Rapid Diagnostics/Abbott Rapid Diagnostics Division (ARDx), 829 Towne Center Drive, Pomona, CA 91767, United States of America
| |
Collapse
|
120
|
Bunge KE, Dezzutti CS, Hendrix CW, Marzinke MA, Spiegel HML, Moncla BJ, Schwartz JL, Meyn LA, Richardson‐Harman N, Rohan LC, Hillier SL. FAME-04: A Phase 1 trial to assess the safety, acceptability, pharmacokinetics and pharmacodynamics of film and gel formulations of tenofovir. J Int AIDS Soc 2018; 21:e25156. [PMID: 30101439 PMCID: PMC6088248 DOI: 10.1002/jia2.25156] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 06/01/2018] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Fast-dissolving vaginal film formulations release antiretroviral drugs directly into vaginal fluid and may be as efficient at drug delivery yet more acceptable to women than gels. In this Phase 1 vaginal film study, the safety, acceptability, pharmacokinetics and pharmacodynamics of two doses of tenofovir (TFV) film and TFV 1% gel were compared to corresponding placebo formulations. METHODS Seventy-eight healthy HIV negative women were randomized to self-insert daily vaginal film (10 mg TFV, 40 mg TFV or placebo) or 4 mL of vaginal gel (TFV 1% [40 mg] or placebo) for seven days. Grade 2 and higher adverse events (AEs) related to study product were compared across study arms using Fisher's exact test. Plasma TFV concentrations were measured before and 2 hours after last product use. Paired cervical and vaginal tissue biopsies obtained 2 hours after the last dose were measured to determine tenofovir diphosphate (TFV-DP) concentrations and exposed to HIV in an ex vivo challenge assay. Acceptability was assessed through questionnaire. RESULTS There was only one grade 2 or higher related AE, the primary endpoint; it occurred in the placebo gel arm. AEs occurred in 90% of participants; the majority (91%) were grade 1. AEs were similar across study arms. TFV concentrations in plasma and TFV-DP concentrations in cervical and vaginal tissues were comparable between 40 mg TFV film and the TFV gel groups. There was a significant relationship between reduced viral replication and TFV-DP concentrations in cervical tissues. Film users were less likely to report product leakage than gel users (66% vs. 100%, p < 0.001). CONCLUSIONS Films were safe and well tolerated. Furthermore, films delivered TFV to mucosal tissues at concentrations similar to gel and were sufficient to block HIV infection of genital tissue ex vivo.
Collapse
Affiliation(s)
- Katherine E Bunge
- Department of Obstetrics, Gynecology, and Reproductive SciencesUniversity of PittsburghPittsburghPAUSA
| | - Charlene S Dezzutti
- Department of Obstetrics, Gynecology, and Reproductive SciencesUniversity of PittsburghPittsburghPAUSA
- Magee‐Womens Research InstitutePittsburghPAUSA
| | - Craig W Hendrix
- Division of Clinical PharmacologyDepartment of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Mark A Marzinke
- Division of Clinical PharmacologyDepartment of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Hans M L Spiegel
- Department of Health and Human ServicesKelly Government SolutionsContractor to National Institute of Allergy and Infectious DiseasesNational Institutes of HealthRockvilleMDUSA
| | - Bernard J Moncla
- Department of Obstetrics, Gynecology, and Reproductive SciencesUniversity of PittsburghPittsburghPAUSA
- Magee‐Womens Research InstitutePittsburghPAUSA
| | | | - Leslie A Meyn
- Department of Obstetrics, Gynecology, and Reproductive SciencesUniversity of PittsburghPittsburghPAUSA
| | | | - Lisa C Rohan
- Department of Obstetrics, Gynecology, and Reproductive SciencesUniversity of PittsburghPittsburghPAUSA
- Magee‐Womens Research InstitutePittsburghPAUSA
- Department of Pharmaceutical SciencesUniversity of PittsburghPittsburghPAUSA
| | - Sharon L Hillier
- Department of Obstetrics, Gynecology, and Reproductive SciencesUniversity of PittsburghPittsburghPAUSA
- Magee‐Womens Research InstitutePittsburghPAUSA
| |
Collapse
|
121
|
Anderson PL. What Can Urine Tell Us About Medication Adherence? EClinicalMedicine 2018; 2-3:5-6. [PMID: 31193677 PMCID: PMC6537573 DOI: 10.1016/j.eclinm.2018.09.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 09/07/2018] [Indexed: 11/13/2022] Open
|
122
|
Heffron R, Mugo N, Hong T, Celum C, Marzinke MA, Ngure K, Asiimwe S, Katabira E, Bukusi EA, Odoyo J, Tindimwebwa E, Bulya N, Baeten JM. Pregnancy outcomes and infant growth among babies with in-utero exposure to tenofovir-based preexposure prophylaxis for HIV prevention. AIDS 2018; 32:1707-1713. [PMID: 30001244 PMCID: PMC6086376 DOI: 10.1097/qad.0000000000001867] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Global guidelines recommend preexposure prophylaxis (PrEP) use by women at risk for HIV, including during pregnancy, a period with heightened HIV risk. However, data to support safety of PrEP use during pregnancy are limited, particularly from women using PrEP throughout pregnancy. METHODS In an open-label delivery study of PrEP integrated with ART for high-risk HIV serodiscordant couples in Kenya and Uganda (the Partners Demonstration Project), women who became pregnant while using PrEP were offered the option to continue PrEP throughout pregnancy. We compared pregnancy outcomes and 1-year infant growth from pregnancies with exposure to PrEP throughout pregnancy to those without any exposure, with data from the placebo arm of a prior efficacy trial of PrEP conducted in the same setting. RESULTS Outcomes from 30 women who elected to continue PrEP throughout pregnancy were compared with those from 96 pregnancies among PrEP-unexposed women. There were small nonsignificant decreases in the frequency of pregnancy loss [16.7% PrEP-exposed versus 23.5% PrEP-unexposed, adjusted odds ratio (aOR) = 0.59, P = 0.4] and preterm delivery [0 versus 7.7%, (aOR) = 0.54, exact P = 0.6]. No congenital anomalies occurred among PrEP-exposed infants. PrEP-exposed infants had slightly lower adjusted mean z-scores for length (-1.73 versus -0.79, P = 0.05) and head circumference (0.24 versus 1.07, P = 0.04) 1 month after birth but were comparable to PrEP-unexposed infants in these measurements 1 year after birth. CONCLUSION This first evaluation among women using PrEP throughout pregnancy indicates no greater frequency of adverse pregnancy outcomes or restricted infant growth; these findings support recommendations permitting PrEP use during pregnancy.
Collapse
Affiliation(s)
- Renee Heffron
- Department of Global Health
- Department of Epidemiology
| | - Nelly Mugo
- Department of Global Health
- Centres for Clinical Research
| | | | - Connie Celum
- Department of Global Health
- Department of Epidemiology
- Department of Medicine
| | - Mark A Marzinke
- Department of Pathology
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Kenneth Ngure
- Department of Global Health
- College of Health Sciences, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | | | - Elly Katabira
- Infectious Disease Institute, Makerere University, Kampala, Uganda
| | - Elizabeth A Bukusi
- Department of Global Health
- Department of Obstetrics & Gynecology, University of Washington, Seattle, USA
- Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Josephine Odoyo
- Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | | | - Nulu Bulya
- Infectious Disease Institute, Makerere University, Kampala, Uganda
| | - Jared M Baeten
- Department of Global Health
- Department of Epidemiology
- Department of Medicine
| |
Collapse
|
123
|
Thurman AR, Schwartz JL, Brache V, Clark MR, McCormick T, Chandra N, Marzinke MA, Stanczyk FZ, Dezzutti CS, Hillier SL, Herold BC, Fichorova R, Asin SN, Rollenhagen C, Weiner D, Kiser P, Doncel GF. Randomized, placebo controlled phase I trial of safety, pharmacokinetics, pharmacodynamics and acceptability of tenofovir and tenofovir plus levonorgestrel vaginal rings in women. PLoS One 2018; 13:e0199778. [PMID: 29953547 PMCID: PMC6023238 DOI: 10.1371/journal.pone.0199778] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 06/07/2018] [Indexed: 01/28/2023] Open
Abstract
To prevent the global health burdens of human immunodeficiency virus [HIV] and unintended/mistimed pregnancies, we developed an intravaginal ring [IVR] that delivers tenofovir [TFV] at ~10mg/day alone or with levonorgestrel [LNG] at ~20μg/day for 90 days. We present safety, pharmacokinetics, pharmacodynamics, acceptability and drug release data in healthy women. CONRAD A13-128 was a randomized, placebo controlled phase I study. We screened 86 women; 51 were randomized to TFV, TFV/LNG or placebo IVR [2:2:1] and 50 completed all visits, using the IVR for approximately 15 days. We assessed safety by adverse events, colposcopy, vaginal microbiota, epithelial integrity, mucosal histology and immune cell numbers and phenotype, cervicovaginal [CV] cytokines and antimicrobial proteins and changes in systemic laboratory measurements, and LNG and TFV pharmacokinetics in multiple compartments. TFV pharmacodynamic activity was measured by evaluating CV fluid [CVF] and tissue for antiviral activity using in vitro models. LNG pharmacodynamic assessments were timed based on peak urinary luteinizing hormone levels. All IVRs were safe with no significant colposcopic, mucosal, immune and microbiota changes and were acceptable. Among TFV containing IVR users, median and mean CV aspirate TFV concentrations remained above 100,000 ng/mL 4 hours post IVR insertion and mean TFV-diphosphate [DP] concentrations in vaginal tissue remained above 1,000 fmol/mg even 3 days post IVR removal. CVF of women using TFV-containing IVRs completely inhibited [94-100%] HIV infection in vitro. TFV/LNG IVR users had mean serum LNG concentrations exceeding 300 pg/mL within 1 hour, remaining high throughout IVR use. All LNG IVR users had a cervical mucus Insler score <10 and the majority [95%] were anovulatory or had abnormal cervical mucus sperm penetration. Estimated in vivo TFV and LNG release rates were within expected ranges. All IVRs were safe with the active ones delivering sustained high concentrations of TFV locally. LNG caused changes in cervical mucus, sperm penetration, and ovulation compatible with contraceptive efficacy. The TFV and TFV/LNG rings are ready for expanded 90 day clinical testing. Trial registration ClinicalTrials.gov #NCT02235662.
Collapse
Affiliation(s)
- Andrea Ries Thurman
- CONRAD, Eastern Virginia Medical School, Arlington, Virginia, United States of America
| | - Jill L. Schwartz
- CONRAD, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | | | - Meredith R. Clark
- CONRAD, Eastern Virginia Medical School, Arlington, Virginia, United States of America
| | - Timothy McCormick
- CONRAD, Eastern Virginia Medical School, Arlington, Virginia, United States of America
| | - Neelima Chandra
- CONRAD, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - Mark A. Marzinke
- Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Frank Z. Stanczyk
- University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| | - Charlene S. Dezzutti
- University of Pittsburgh, Department of Obstetrics, Gynecology & Reproductive Sciences, Department of Infectious Diseases & Microbiology, Graduate School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Sharon L. Hillier
- University of Pittsburgh School of Medicine, Departments of Obstetrics, Gynecology and Reproductive Sciences and Microbiology and Molecular Genetics, Pittsburgh, Pennsylvania, United States of America
| | - Betsy C. Herold
- Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Raina Fichorova
- Laboratory of Genital Tract Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Susana N. Asin
- V.A. Medical Center, White River Junction, VT and Geisel School of Medicine at Dartmouth, New Hampshire
| | - Christiane Rollenhagen
- V.A. Medical Center, White River Junction, VT and Geisel School of Medicine at Dartmouth, New Hampshire
| | - Debra Weiner
- FHI360, Durham, North Carolina, United States of America
| | - Patrick Kiser
- Northwestern University, Evanston, Illinois, United States of America
| | - Gustavo F. Doncel
- CONRAD, Eastern Virginia Medical School, Arlington, Virginia, United States of America
| |
Collapse
|
124
|
Development of rectal enema as microbicide (DREAM): Preclinical progressive selection of a tenofovir prodrug enema. Eur J Pharm Biopharm 2018; 138:23-29. [PMID: 29802984 DOI: 10.1016/j.ejpb.2018.05.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/14/2018] [Accepted: 05/22/2018] [Indexed: 12/22/2022]
Abstract
HIV pre-exposure prophylaxis (PrEP) strategies have the potential to prevent millions of incident HIV infections each year. However, the efficacy of PrEP strategies has been plagued by issues of non-adherence, likely because of the difficulty in motivating otherwise healthy people to adhere to treatment regimens that require significant behavioral changes and daily discipline. An alternative approach to PrEP is to focus on strategies that fit in to normal, and even desirable, sexual behaviors, such as the use of cleansing enemas by men who have sex with men (MSM) prior to receptive anal intercourse (RAI). Here, we describe preclinical efforts toward optimizing a tenofovir (TFV)-based enema formulation for rectal PrEP. Using a murine model, we compared the plasma and tissue pharmacokinetics of TFV and various TFV prodrugs, including tenofovir disoproxil fumarate (TDF), tenofovir alafenamide (TAF), and hexadecyloxypropyl tenofovir (CMX157), after dosing as enema formulations with varying osmolality and ion content. We observed that the enema vehicle composition played a more important role than the TFV prodrug properties in achieving rapid and therapeutically relevant tenofovir diphosphate (TFV-DP) concentrations in mouse colorectal tissue. Our results support the next steps, which are further preclinical (non-human primate) and clinical development of a hypo-osmolar TFV enema product for rectal PrEP.
Collapse
|
125
|
Abaasa A, Hendrix C, Gandhi M, Anderson P, Kamali A, Kibengo F, Sanders EJ, Mutua G, Bumpus NN, Priddy F, Haberer JE. Utility of Different Adherence Measures for PrEP: Patterns and Incremental Value. AIDS Behav 2018; 22:1165-1173. [PMID: 29090394 PMCID: PMC5878836 DOI: 10.1007/s10461-017-1951-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Measuring PrEP adherence remains challenging. In 2009–2010, the International AIDS Vaccine Initiative randomized phase II trial participants to daily tenofovir disoproxil fumarate/emtricitabine or placebo in Uganda and Kenya. Adherence was measured by electronic monitoring (EM), self-report (SR), and drug concentrations in plasma and hair. Each adherence measure was categorised as low, moderate, or high and also considered continuously; the incremental value of combining measures was determined. Forty-five participants were followed over 4 months. Discrimination for EM adherence by area under receiver operating curves (AROC) was poor for SR (0.53) and best for hair (AROC 0.85). When combining hair with plasma or hair with self-report, discrimination was improved (AROC > 0.9). Self-reported adherence was of low utility by itself. Hair level was the single best PK measure to predict EM-assessed adherence; the other measurements had lower discrimination values. Combining short-term (plasma) and long-term (hair) metrics could be useful to assess patterns of drug-taking in the context of PrEP.
Collapse
Affiliation(s)
- Andrew Abaasa
- MRC/UVRI Uganda Research Unit on AIDS, P.O Box 49, Entebbe, Uganda.
| | - Craig Hendrix
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, ME, USA
| | - Monica Gandhi
- Divisions of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Peter Anderson
- Department of Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Anatoli Kamali
- International AIDS Vaccine Initiative, New York, NY, USA
| | - Freddie Kibengo
- MRC/UVRI Uganda Research Unit on AIDS, P.O Box 49, Entebbe, Uganda
| | - Eduard J Sanders
- Kenya Medical Research Institute, University of Oxford, Kilifi, Kenya
| | - Gaudensia Mutua
- Kenya AIDS Vaccine Initiative, University of Nairobi, Nairobi, Kenya
| | - Namandjé N Bumpus
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, ME, USA
| | - Frances Priddy
- International AIDS Vaccine Initiative, New York, NY, USA
| | - Jessica E Haberer
- Massachusetts General Hospital Global Health and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
126
|
Bakshi RP, Breakey J, Manohar M, Jois B, Fuchs EJ, Marzinke MA. Short Communication: Specimen Processing Impacts Tissue Tenofovir Pharmacokinetic Measurements. AIDS Res Hum Retroviruses 2018; 34:354-356. [PMID: 29258331 DOI: 10.1089/aid.2017.0231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Antiretroviral drug concentrations at sites of HIV exposure are important drivers that influence the development of HIV pre-exposure chemoprophylaxis strategies and regimens. We assessed the effect of collection method-in the presence or absence of tissue culture medium-on tenofovir (TFV) and tenofovir diphosphate (TFV-DP) concentrations in colonic biopsies. We find significant baseline interbiopsy variation in TFV (38% CV) and TFV-DP (33% CV) concentrations. Incubation in medium leads to a fluid absorption-driven twofold increase in tissue weight with a concomitant 75% decrease in weight-adjusted tissue TFV concentrations 120 min post-incubation. In contrast, adjusted TFV-DP concentrations decrease by only 25% during the same period, with this difference not achieving statistical significance. Although colonic biopsies should be collected in the absence of medium for accurate TFV concentrations, the presence of medium does not significantly impact TFV-DP-dependent pharmacokinetic or pharmacodynamic assays. Appropriate assessment of tissue drug concentrations should account for biopsy collection method and drug mechanism of action.
Collapse
Affiliation(s)
- Rahul P. Bakshi
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jennifer Breakey
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Madhuri Manohar
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Bhavna Jois
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Edward J. Fuchs
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mark A. Marzinke
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Division of Clinical Chemistry, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
127
|
Haaland RE, Holder A, Pau CP, Swaims-Kohlmeier A, Dawson C, Smith DK, Segolodi TM, Thigpen MC, Paxton LA, Parsons TL, Hendrix CW, Hart CE. Levels of Intracellular Phosphorylated Tenofovir and Emtricitabine Correlate With Natural Substrate Concentrations in Peripheral Blood Mononuclear Cells of Persons Prescribed Daily Oral Truvada for HIV Pre-exposure Prophylaxis. J Acquir Immune Defic Syndr 2018; 75:e86-e88. [PMID: 28225438 DOI: 10.1097/qai.0000000000001324] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Richard E Haaland
- *Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA †Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Robinson JA, Marzinke MA, Fuchs EJ, Bakshi RP, Spiegel HML, Coleman JS, Rohan LC, Hendrix CW. Comparison of the Pharmacokinetics and Pharmacodynamics of Single-Dose Tenofovir Vaginal Film and Gel Formulation (FAME 05). J Acquir Immune Defic Syndr 2018; 77:175-182. [PMID: 29135651 PMCID: PMC5821271 DOI: 10.1097/qai.0000000000001587] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Although preexposure prophylaxis with oral tenofovir (TFV) disoproxil fumarate/emtricitabine reduces HIV acquisition rates, poor adherence to and acceptability of daily vaginal gels have led to development of vaginal film formulations to improve adherence and, potentially, to enable episodic use. STUDY DESIGN In this 2-arm, cross-over study of a fast-dissolving tenofovir film (40 mg) compared with a previously studied semisolid tenofovir 1% gel (40 mg), 10 healthy women received a single vaginal dose of each study product. Clinical, pharmacokinetic, and antiviral assessments were performed over 1 week after dose. RESULTS Nine of 10 participants experienced mild to moderate adverse effects, similar between products, with no severe adverse events or events attributed to study products. TFV concentrations after film dosing exceeded concentrations after gel dosing in plasma between 8 and 24 hours (P ≤ 0.02). TFV concentrations in cervicovaginal fluid and both TFV and TFV diphosphate concentrations in cervical tissue homogenates were higher after film dosing (all P values < 0.04). The differences ranged from median (interquartile range) 2.9-fold (1.1, 9.0; midvaginal cervicovaginal fluid) to 4.4-fold (2.9, 7.7; plasma). Neither film nor gel demonstrated reduced cervical tissue biopsy infectivity after ex vivo HIV challenge. CONCLUSION Single-dose tenofovir film demonstrated consistently higher concentrations in plasma and cervicovaginal samples when compared with gel during the first day after dosing. Single-dose cervical tissue TFV-diphosphate concentrations at 5 hours exceeded steady-state concentrations previously reported with daily oral Truvada dosing. Tenofovir film may provide an alternative to tenofovir oral and gel formulations. Clinical efficacy remains to be tested.
Collapse
Affiliation(s)
- Jennifer A. Robinson
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, Maryland
- Department of Medicine (Clinical Pharmacology), Johns Hopkins University, Baltimore, Maryland
| | - Mark A. Marzinke
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
- Department of Medicine (Clinical Pharmacology), Johns Hopkins University, Baltimore, Maryland
| | - Edward J. Fuchs
- Department of Medicine (Clinical Pharmacology), Johns Hopkins University, Baltimore, Maryland
| | - Rahul P. Bakshi
- Department of Medicine (Clinical Pharmacology), Johns Hopkins University, Baltimore, Maryland
| | - Hans M. L. Spiegel
- Kelly Government Solutions, Contractor to Division of AIDS, PMPRB/Prevention Sciences Program, Division of AIDS, NIAID, NIH, Rockville, MD
| | - Jenell S. Coleman
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, Maryland
| | - Lisa C. Rohan
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Craig W. Hendrix
- Department of Medicine (Clinical Pharmacology), Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
129
|
Bekker LG, Roux S, Sebastien E, Yola N, Amico KR, Hughes JP, Marzinke MA, Hendrix CW, Anderson PL, Elharrar V, Stirratt M, Rooney JF, Piwowar-Manning E, Eshleman SH, McKinstry L, Li M, Dye BJ, Grant RM. Daily and non-daily pre-exposure prophylaxis in African women (HPTN 067/ADAPT Cape Town Trial): a randomised, open-label, phase 2 trial. Lancet HIV 2018; 5:e68-e78. [PMID: 28986029 PMCID: PMC6107917 DOI: 10.1016/s2352-3018(17)30156-x] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 06/30/2017] [Accepted: 07/13/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND The relative feasibility and acceptability of daily versus non-daily dosing of oral HIV pre-exposure prophylaxis (PrEP) among women are unknown. We aimed to investigate the feasibility of non-daily PrEP regimens in adult women. METHODS We did a randomised, open-label, phase 2 clinical trial (HPTN 067/ADAPT) of oral PrEP with emtricitabine plus tenofovir disoproxil fumarate at a research centre in Cape Town, South Africa. Participants were adult women (age ≥18 years) who received directly observed dosing once a week for 5 weeks followed by random assignment (1:1:1) at week 6 to one of three unblinded PrEP regimens for self-administered dosing over 24 weeks: daily; time-driven (twice a week plus a post-sex dose); or event-driven (one tablet both before and after sex). Primary outcomes were PrEP coverage (at least one dose within the 4 days before sex and one dose within 24 h after sex), pills needed or used to achieve regimen-specific adherence and coverage, and symptoms and side-effects. All analyses were by intention to treat. This trial is registered with ClinicalTrials.gov, number NCT01327651; the trial is completed and this report presents the final analysis. FINDINGS Between Sept 12, 2011, and Oct 3, 2012, 191 women were enrolled to the trial. 178 (93%) completed directly observed dosing and were randomly assigned one of the three PrEP regimens for the self-administered phase: 59 were allocated the daily regimen, 59 the time-driven regimen, and 60 the event-driven regimen. Median age of women was 26 years (IQR 21-37; range 18-52). In women allocated the daily regimen, 1459 (75%) of 1952 sex events were covered by PrEP, compared with 599 (56%) of 1074 sex events among those assigned the time-driven regimen (odds ratio [OR] 2·35, 95% CI 1·43-3·83; p=0·0007) and 798 (52%) of 1542 sex events among those allotted the event-driven regimen (2·76, 1·68-4·53; p<0·0001). Fewer pills were needed for complete adherence in women allocated non-daily regimens (vs daily regimen, relative mean 2·53 [95% CI 2·39-2·69] for the time-driven regimen and 4·16 [3·59-4·82] for the event-driven regimen; p<0·0001). Side-effects were uncommon. Eight HIV seroconversions occurred overall, with four documented during the self-administered phase (two with the time-driven regimen and two with the event-driven regimen). Adherence to the assigned regimen was 75% (7283 of 9652 doses taken) for women allocated the daily regimen compared with 65% for those assigned the time-driven regimen (2367 of 3616 doses taken; p=0·0028) and 53% for those allotted the event-driven regimen (1161 of 2203 doses taken; p<0·0001). When sex was reported in the previous week, PrEP drugs were detected (above the lower limits of quantification) more frequently in women assigned the daily regimen (73 [68%] of 107 samples) than in those allocated the time-driven regimen (42 [58%] of 72 samples) and the event-driven regimen (41 [41%] of 99 samples). INTERPRETATION Daily PrEP dosing resulted in higher coverage of sex events, increased adherence to the regimen, and augmented drug concentrations than did either time-driven or event-driven dosing. These findings support recommendations for daily use of PrEP with oral emtricitabine plus tenofovir disoproxil fumarate in women. FUNDING HIV Prevention Trials Network.
Collapse
Affiliation(s)
- Linda-Gail Bekker
- The Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa.
| | - Surita Roux
- Synexus Clinical Research SA, Somerset West, Cape Town, South Africa
| | - Elaine Sebastien
- The Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - Ntando Yola
- The Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - K Rivet Amico
- School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - James P Hughes
- Department of Biostatistics, University of Washington, Seattle, WA, USA; Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Mark A Marzinke
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Craig W Hendrix
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Peter L Anderson
- Department of Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Vanessa Elharrar
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Michael Stirratt
- Division of AIDS Research, National Institute of Mental Health, National Institutes of Health, Bethesda, MD USA
| | | | | | - Susan H Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Maoji Li
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Robert M Grant
- Gladstone Institutes, University of California, and San Francisco AIDS Foundation, San Francisco, CA, USA
| |
Collapse
|
130
|
Hypo-osmolar Formulation of Tenofovir (TFV) Enema Promotes Uptake and Metabolism of TFV in Tissues, Leading to Prevention of SHIV/SIV Infection. Antimicrob Agents Chemother 2017; 62:AAC.01644-17. [PMID: 29084755 DOI: 10.1128/aac.01644-17] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/19/2017] [Indexed: 12/31/2022] Open
Abstract
Oral preexposure prophylaxis (PrEP) has been approved for prophylaxis of HIV-1 transmission but is associated with high costs and issues of adherence. Protection from anal transmission of HIV using topical microbicides and methods congruent with sexual behavior offers the promise of improved adherence. We compared the pharmacokinetics (PK) and ex vivo efficacy of iso-osmolar (IOsm) and hypo-osmolar (HOsm) rectal enema formulations of tenofovir (TFV) in rhesus macaques. Single-dose PK of IOsm or HOsm high-dose (5.28 mg/ml) and low-dose (1.76 mg/ml) formulations of TFV enemas were evaluated for systemic uptake in blood, colorectal biopsy specimens, and rectal CD4+ T cells. Markedly higher TFV concentrations were observed in plasma and tissues after administration of the HOsm high-dose formulation than with all other formulations tested. TFV and TFV diphosphate (TFV-DP) concentrations in tissue correlated for the HOsm high-dose formulation, demonstrating rapid uptake and transformation of TFV to TFV-DP in tissues. TFV-DP amounts in tissues collected at 1 and 24 h were 7 times and 5 times higher, respectively (P < 0.01), than the ones collected in tissues with the IOsm formulation. The HOsm high-dose formulation prevented infection in ex vivo challenges of rectal tissues collected at 1, 24, and 72 h after the intrarectal dosing, whereas the same TFV dose formulated as an IOsm enema was less effective.
Collapse
|
131
|
Intracellular Tenofovir-Diphosphate and Emtricitabine-Triphosphate in Dried Blood Spots following Directly Observed Therapy. Antimicrob Agents Chemother 2017; 62:AAC.01710-17. [PMID: 29038282 DOI: 10.1128/aac.01710-17] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 10/10/2017] [Indexed: 12/11/2022] Open
Abstract
Studies of daily emtricitabine-tenofovir disoproxil fumarate (FTC-TDF) for HIV preexposure prophylaxis (PrEP) in men who have sex with men (MSM) modeled intracellular tenofovir-diphosphate (TFV-DP) in dried blood spots (DBS) to assess adherence and corresponding PrEP outcomes. We conducted a prospective, randomized, crossover pharmacokinetic study of TFV-DP in DBS during 33%, 67%, or 100% of daily dosing under directly observed therapy (DOT). Participants were assigned to two 12-week dosing regimens, separated by a 12-week washout. Forty-eight adults (25 women) from Denver and San Francisco were included. TFV-DP exhibited a median half-life of 17 days, reaching steady state in 8 weeks. TFV-DP was dose proportional with mean (SD) steady-state concentrations of 530 (159), 997 (267), and 1,605 (405) fmol/punch for the 33%, 67%, and 100% arms, respectively. Prior work in MSM demonstrated clinically meaningful TFV-DP thresholds of 350, 700, and 1,250 fmol/punch, which were estimated 25th percentiles for 2, 4, and 7 doses/week. In the present study, corresponding TFV-DP was within 3% of the prior estimates, and subgroups by site, race, and sex were within 14% of prior estimates, although males had 17.6% (95% confidence intervals [CIs], 6.5, 27.4%) lower TFV-DP than females. The thresholds of 350, 700, and 1,250 fmol/punch were achieved by 75% of men taking ≥1.2, 3.2, and 6 doses/week and 75% of women taking ≥0.6, 2.0, and 5.3 doses/week, indicating that lower dosing reached these thresholds for both sexes. In conclusion, TFV-DP arising from DOT was similar to previous estimates and is useful for interpreting PrEP adherence and study outcomes. (This study has been registered at ClinicalTrials.gov under identifier NCT02022657.).
Collapse
|
132
|
Technological methods to measure adherence to antiretroviral therapy and preexposure prophylaxis. Curr Opin HIV AIDS 2017; 12:467-474. [PMID: 28590335 DOI: 10.1097/coh.0000000000000393] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW The WHO's Consolidated Guidelines (2016) call for research on improved methods to proactively monitor adherence and identify those individuals who have the greatest needs for adherence support. This review aims to elucidate the latest technologies available to measure adherence to HIV antiretroviral therapy and preexposure prophylaxis against HIV infection and present their utility in various settings and populations. RECENT FINDINGS Within the last few years, advances have been made in the features of existing technology to measure adherence (real-time electronic adherence measurements), additional approaches have been developed (digital medicine systems) and improved (short message service surveys), and point of care testing for pharmacokinetic measures are under development. SUMMARY Technology advances in adherence measurement are promising for improved accuracy and, in some cases, the ability to intervene with adherence challenges in real time. This progress will greatly further our understanding of adherence behavior, as well as the ability to effectively link interventions with individuals who need them, thus maximizing the clinical and public health benefits of both antiretroviral therapy and preexposure prophylaxis.
Collapse
|
133
|
Brief Report: Context Matters: PrEP Adherence is Associated With Sexual Behavior Among HIV Serodiscordant Couples in East Africa. J Acquir Immune Defic Syndr 2017; 76:488-492. [PMID: 28961676 DOI: 10.1097/qai.0000000000001548] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Short message service (SMS) surveys are a promising tool for understanding whether preexposure prophylaxis (PrEP) adherence aligns with risk for HIV acquisition-a concept known as prevention-effective adherence. METHODS The Partners Demonstration Project was an open-label study of integrated PrEP and antiretroviral therapy (ART) delivery among high-risk HIV serodiscordant couples in East Africa. HIV-uninfected partners were offered PrEP until their HIV-infected partner had taken ART for ≥6 months. At 2 study sites, HIV-uninfected partners were offered enrollment into the Partners Mobile Adherence to PrEP (PMAP) substudy based on ongoing PrEP use, personal cell phone ownership, and ability to use SMS. SMS surveys asked about PrEP adherence and sexual activity in the previous 24 hours; these surveys were sent daily for the 7 days before and 7 days after routine study visits in the Partners Demonstration Project. RESULTS The PMAP substudy enrolled 373 HIV-uninfected partners; 69% were men and mean age was 31 years. Participants completed 17,030 of 23,056 SMS surveys sent (74%) with a mean of 47 surveys per participant over 9.8 months of follow-up. While HIV-infected partner use of ART was <6 months, mean reported PrEP adherence was 92% on surveys concurrently reporting sex within the serodiscordant partnership, and 84% on surveys reporting no sex (P < 0.001). DISCUSSION SMS surveys provided daily assessment of concurrent PrEP adherence and sexual behavior. Higher PrEP adherence was temporally associated with increased risk for HIV acquisition.
Collapse
|
134
|
Alignment of adherence and risk for HIV acquisition in a demonstration project of pre-exposure prophylaxis among HIV serodiscordant couples in Kenya and Uganda: a prospective analysis of prevention-effective adherence. J Int AIDS Soc 2017; 20:21842. [PMID: 28741331 PMCID: PMC5577705 DOI: 10.7448/ias.20.1.21842] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Introduction: Adherence is essential for pre-exposure prophylaxis (PrEP) to protect against HIV acquisition, but PrEP use need not be life-long. PrEP is most efficient when its use is aligned with periods of risk – a concept termed prevention-effective adherence. The objective of this paper is to describe prevention-effective adherence and predictors of adherence within an open-label delivery project of integrated PrEP and antiretroviral therapy (ART) among HIV serodiscordant couples in Kenya and Uganda (the Partners Demonstration Project). Methods: We offered PrEP to HIV-uninfected participants until the partner living with HIV had taken ART for ≥6 months (a strategy known as “PrEP as a bridge to ART”). The level of adherence sufficient to protect against HIV was estimated in two ways: ≥4 and ≥6 doses/week (per electronic monitoring). Risk for HIV acquisition was considered high if the couple reported sex with <100% condom use before six months of ART, low if they reported sex but had 100% condom use and/or six months of ART and very low if no sex was reported. We assessed prevention-effective adherence by cross-tabulating PrEP use with HIV risk and used multivariable regression models to assess predictors of ≥4 and ≥6 doses/week. Results: A total of 985 HIV-uninfected participants initiated PrEP; 67% were male, median age was twenty-nine years, and 67% reported condomless sex in the month before enrolment. An average of ≥4 doses and ≥6 doses/week were taken in 81% and 67% of participant-visits, respectively. Adherence sufficient to protect against HIV acquisition was achieved in 75–88% of participant-visits with high HIV risk. The strongest predictor of achieving sufficient adherence was reporting sex with the study partner who was living with HIV; other statistically significant predictors included no concerns about daily PrEP, pregnancy or pregnancy intention, females aged >25 years, older male partners and desire for relationship success. Predictors of not achieving sufficient adherence were no longer being a couple, delayed PrEP initiation, >6 months of follow-up, ART use >6 months by the partner living with HIV and problem alcohol use. Conclusions: Over three-quarters of participant-visits by HIV-uninfected partners in serodiscordant couples achieved prevention-effective adherence with PrEP. Greater adherence was observed during months with HIV risk and the strongest predictor of achieving sufficient adherence was sexual activity.
Collapse
|
135
|
Hoornenborg E, Krakower DS, Prins M, Mayer KH. Pre-exposure prophylaxis for MSM and transgender persons in early adopting countries. AIDS 2017; 31:2179-2191. [PMID: 28991023 PMCID: PMC5812254 DOI: 10.1097/qad.0000000000001627] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
: Pre-exposure prophylaxis (PrEP) is a potent and underutilized HIV prevention tool. In this paper we review the state of knowledge regarding PrEP implementation for men who have sex with men and transgender persons in early adopting countries. We focus on implementation of PrEP in demonstration projects and clinical care, and describe the status of PrEP availability and uptake. We report on approaches to identifying appropriate PrEP candidates in real-world settings and on best practices for clinical monitoring. This includes the exclusion of undiagnosed HIV infection prior to PrEP initiation and longitudinal measurement of renal function, in light of safety data. Since adherence is the primary factor moderating the effectiveness of PrEP, we discuss effective adherence support interventions. Additionally, we review the evidence for risk compensation with PrEP use and opportunities to provide PrEP as part of comprehensive and inclusive preventive health programs. We summarize cost-effectiveness studies, including their variable conclusions because of differing underlying assumptions, and discuss the importance of budgetary impact for public health programs and health care insurers. Further, we emphasize a need for greater engagement of health care providers in PrEP to increase access. We conclude with recommendations for ways to improve future efforts at implementing PrEP.
Collapse
Affiliation(s)
- Elske Hoornenborg
- aPublic Health Service of Amsterdam, Department of Infectious Diseases bAcademic Medical Center, Department of Infectious Diseases, Center for Immunity and Infection Amsterdam (CINIMA), University of Amsterdam, the Netherlands cBeth Israel Deaconess Medical Center, Department of Medicine, Division of Infectious Diseases, Harvard Medical School dThe Fenway Institute, Fenway Health, Boston, Massachusetts, USA
| | | | | | | |
Collapse
|
136
|
Gulick RM, Wilkin TJ, Chen YQ, Landovitz RJ, Amico KR, Young AM, Richardson P, Marzinke MA, Hendrix CW, Eshleman SH, McGowan I, Cottle LM, Andrade A, Marcus C, Klingman KL, Chege W, Rinehart AR, Rooney JF, Andrew P, Salata RA, Siegel M, Manabe YC, Frank I, Ho K, Santana J, Stekler JD, Swaminathan S, McCauley M, Hodder S, Mayer KH. Safety and Tolerability of Maraviroc-Containing Regimens to Prevent HIV Infection in Women: A Phase 2 Randomized Trial. Ann Intern Med 2017; 167:384-393. [PMID: 28828489 PMCID: PMC5667908 DOI: 10.7326/m17-0520] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Maraviroc (MVC) is a candidate drug for HIV preexposure prophylaxis (PrEP). OBJECTIVE To assess the safety and tolerability of MVC-containing PrEP over 48 weeks in U.S. women at risk for HIV infection. DESIGN Phase 2 randomized, controlled, double-blinded study of 4 antiretroviral regimens used as PrEP. (ClinicalTrials.gov: NCT01505114). SETTING 12 clinical research sites of the HIV Prevention Trials Network and AIDS Clinical Trials Group. PARTICIPANTS HIV-uninfected women reporting condomless vaginal or anal intercourse with at least 1 man with HIV infection or unknown serostatus within 90 days. INTERVENTION MVC only, MVC-emtricitabine (FTC), MVC-tenofovir disoproxil fumarate (TDF), and TDF-FTC (control). MEASUREMENTS At each visit, clinical and laboratory (including HIV) assessments were done. Primary outcomes were grade 3 and 4 adverse events and time to permanent discontinuation of the study regimen. All randomly assigned participants were analyzed according to their original assignment. RESULTS Among 188 participants, 85% completed follow-up, 11% withdrew early, and 4% were lost to follow-up; 19% discontinued their regimen prematurely. The number discontinuing and the time to discontinuation did not differ among regimens. Grade 3 or 4 adverse events occurred in 5 (MVC), 13 (MVC-FTC), 9 (MVC-TDF), and 8 (TDF-FTC) participants; rates did not differ among regimens. One death (by suicide) occurred in the MVC-TDF group but was judged not to be related to study drugs. Of available plasma samples at week 48 (n = 126), 60% showed detectable drug concentrations. No new HIV infections occurred. LIMITATIONS Participants were not necessarily at high risk for HIV infection. The regimen comprised 3 pills taken daily. The study was not powered for efficacy. CONCLUSION Maraviroc-containing PrEP regimens were safe and well-tolerated compared with TDF-FTC in U.S. women. No new HIV infections occurred, although whether this was due to study drugs or low risk in the population is uncertain. Maraviroc-containing PrEP for women may warrant further study. PRIMARY FUNDING SOURCE National Institutes of Health.
Collapse
Affiliation(s)
- Roy M Gulick
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Timothy J Wilkin
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Ying Q Chen
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Raphael J Landovitz
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - K Rivet Amico
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Alicia M Young
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Paul Richardson
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Mark A Marzinke
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Craig W Hendrix
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Susan H Eshleman
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Ian McGowan
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Leslie M Cottle
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Adriana Andrade
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Cheryl Marcus
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Karin L Klingman
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Wairimu Chege
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Alex R Rinehart
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - James F Rooney
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Philip Andrew
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Robert A Salata
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Marc Siegel
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Yukari C Manabe
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Ian Frank
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Ken Ho
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Jorge Santana
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Joanne D Stekler
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Shobha Swaminathan
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Marybeth McCauley
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Sally Hodder
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Kenneth H Mayer
- From Weill Cornell Medicine, New York, New York; Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington; University of California, Los Angeles, Los Angeles, California; University of Michigan, Ann Arbor, Michigan; Johns Hopkins University School of Medicine, Baltimore, Maryland; University of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; University of North Carolina, Chapel Hill, North Carolina; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland; ViiV Healthcare, Durham, North Carolina; Gilead Sciences, Foster City, California; Case Western Reserve University, Cleveland, Ohio; The George Washington University and FHI 360, Washington, DC; Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Rutgers New Jersey Medical School, Newark, New Jersey; West Virginia University, Morgantown, West Virginia; and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
137
|
Characterization of HIV Seroconverters in a TDF/FTC PrEP Study: HPTN 067/ADAPT. J Acquir Immune Defic Syndr 2017; 75:271-279. [PMID: 28328548 DOI: 10.1097/qai.0000000000001374] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND HIV Prevention Trials Network (HPTN) 067/ADAPT evaluated tenofovir disoproxil fumarate/emtricitabine (TDF/FTC) pre-exposure prophylaxis (PrEP) in women (South Africa) and men who have sex with men (Thailand, US). Participants received once-weekly directly observed therapy (DOT) of TDF/FTC, and were then randomized to daily, time-driven, or event-driven PrEP. This report describes characterization of 12 HIV seroconversion events in this trial. METHODS HIV rapid testing was performed at study sites. Retrospective testing included fourth generation assays, HIV RNA testing, Western blot, an HIV-1/2 discriminatory assay, resistance testing, and antiretroviral drug testing. RESULTS Six of the 12 seroconverters received TDF/FTC in the DOT phase, but were not randomized (3 were acutely infected at enrollment; 2 were infected during the DOT phase; 1 was not randomized because of pregnancy). One of the 6 randomized participants had acute infection at randomization but was not diagnosed for 3-4 months because HIV rapid tests were nonreactive; continued daily PrEP use was associated with false-negative antibody tests and low HIV RNA levels. The 5 participants infected after randomization included 4 with low adherence to the PrEP regimen, and one who reported a 7-day period without dosing before infection. Three participants had TDF/FTC resistance (M184I, K65R), including 2 who received only 4 once-weekly TDF/FTC doses; most TDF/FTC mutations were detected by next generation sequencing only. CONCLUSIONS In HPTN 067/ADAPT, participants who acquired HIV infection had infrequent PrEP dosing or low/suboptimal adherence. Sensitive assays improved detection of HIV infection and drug resistance. Drug resistance was observed with limited PrEP exposure.
Collapse
|
138
|
Donnell D, Ramos E, Celum C, Baeten J, Dragavon J, Tappero J, Lingappa JR, Ronald A, Fife K, Coombs RW. The effect of oral preexposure prophylaxis on the progression of HIV-1 seroconversion. AIDS 2017; 31:2007-2016. [PMID: 28692542 PMCID: PMC5578893 DOI: 10.1097/qad.0000000000001577] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/08/2017] [Accepted: 06/15/2017] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To investigate whether oral preexposure prophylaxis (PrEP) alters timing and patterns of seroconversion when PrEP use continues after HIV-1 infection. DESIGN Retrospective testing of the timing of Fiebig stage HIV-1 seroconversion in the Partners PrEP Study, a randomized placebo-controlled clinical trial of PrEP conducted in Kenya and Uganda. METHODS Specimens from 138 seroconverters were collected every 3 months and when HIV-1 infection was suspected based on monthly rapid HIV-1 tests. Progression of seroconversion was compared between randomized groups (PrEP versus placebo) and per-protocol groups (placebo versus PrEP participants with detectable tenofovir during the seroconversion period) using laboratory assessment of Fiebig stage. Delay in site-detection of seroconversion and association with PrEP drug-regimen resistant virus were assessed using logistic regression. Analysis of time to each Fiebig stage used maximum likelihood estimation with a parametric model to accommodate the varying lengths of HIV-infection intervals. RESULTS There was a significant increase in delayed site detection of infection associated with PrEP (odds ratio = 3.49, P = 0.044). Delay in detection was not associated with increased risk of resistance in the PrEP arm (odds ratio = 0.93, P = 0.95). Estimated time to each Fiebig stage was elongated in seroconverters with evidence of ongoing PrEP use, significantly for only Stage 5 (28 versus 17 days, P = 0.05). Adjusted for Fiebig stage, viral RNA was ∼2/3 log lower in those assigned to PrEP compared with placebo; no differences were found in Architect signal to cut-off at any stage. CONCLUSION Ongoing PrEP use in seroconverters may delay detection of infection and elongate seroconversion, although the delay does not increase risk of resistance.
Collapse
Affiliation(s)
- Deborah Donnell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
- Department of Global Health
| | | | - Connie Celum
- Department of Global Health
- Department of Epidemiology
- Department of Medicine
| | - Jared Baeten
- Department of Global Health
- Department of Epidemiology
- Department of Medicine
| | | | - Jordan Tappero
- Division of Global Health Protection, Center for Global Health, CDC, Atlanta, Georgia, USA
| | - Jairam R. Lingappa
- Department of Global Health
- Department of Medicine
- Department of Pediatrics, University of Washington, Seattle, Washington
| | - Allan Ronald
- Departments of Medical Microbiology and Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Kenneth Fife
- Department of Microbiology and immunology, Indiana University, Indianapolis, Indiana, USA
| | | |
Collapse
|
139
|
Gallay PA, Chatterji U, Kirchhoff A, Gandarilla A, Gunawardana M, Pyles RB, Marzinke MA, Moss JA, Baum MM. Prevention of vaginal and rectal HIV transmission by antiretroviral combinations in humanized mice. PLoS One 2017; 12:e0184303. [PMID: 28880948 PMCID: PMC5589224 DOI: 10.1371/journal.pone.0184303] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/21/2017] [Indexed: 12/24/2022] Open
Abstract
With more than 7,000 new HIV infections daily worldwide, there is an urgent need for non-vaccine biomedical prevention (nBP) strategies that are safe, effective, and acceptable. Clinical trials have demonstrated that pre-exposure prophylaxis (PrEP) with antiretrovirals (ARVs) can be effective at preventing HIV infection. In contrast, other trials using the same ARVs failed to show consistent efficacy. Topical (vaginal and rectal) dosing is a promising regimen for HIV PrEP as it leads to low systematic drug exposure. A series of titration studies were carried out in bone marrow/liver/thymus (BLT) mice aimed at determining the adequate drug concentrations applied vaginally or rectally that offer protection against rectal or vaginal HIV challenge. The dose-response relationship of these agents was measured and showed that topical tenofovir disoproxil fumarate (TDF) and emtricitabine (FTC) can offer 100% protection against rectal or vaginal HIV challenges. From the challenge data, EC50 values of 4.6 μM for TDF and 0.6 μM for FTC for HIV vaginal administration and 6.1 μM TDF and 0.18 μM for FTC for rectal administration were obtained. These findings suggest that the BLT mouse model is highly suitable for studying the dose-response relationship in single and combination ARV studies of vaginal or rectal HIV exposure. Application of this sensitive HIV infection model to more complex binary and ternary ARV combinations, particularly where agents have different mechanisms of action, should allow selection of optimal ARV combinations to be advanced into pre-clinical and clinical development as nBP products.
Collapse
Affiliation(s)
- Philippe A. Gallay
- Department of Immunology & Microbiology, The Scripps Research Institute; La Jolla, California, United States of America
| | - Udayan Chatterji
- Department of Immunology & Microbiology, The Scripps Research Institute; La Jolla, California, United States of America
| | - Aaron Kirchhoff
- Department of Immunology & Microbiology, The Scripps Research Institute; La Jolla, California, United States of America
| | - Angel Gandarilla
- Department of Immunology & Microbiology, The Scripps Research Institute; La Jolla, California, United States of America
| | - Manjula Gunawardana
- Department of Chemistry, Oak Crest Institute of Science; Monrovia, California, United States of America
| | - Richard B. Pyles
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Mark A. Marzinke
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - John A. Moss
- Department of Chemistry, Oak Crest Institute of Science; Monrovia, California, United States of America
| | - Marc M. Baum
- Department of Chemistry, Oak Crest Institute of Science; Monrovia, California, United States of America
| |
Collapse
|
140
|
High levels of adherence to a rectal microbicide gel and to oral Pre-Exposure Prophylaxis (PrEP) achieved in MTN-017 among men who have sex with men (MSM) and transgender women. PLoS One 2017; 12:e0181607. [PMID: 28750059 PMCID: PMC5531503 DOI: 10.1371/journal.pone.0181607] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 07/04/2017] [Indexed: 11/19/2022] Open
Abstract
Trials to assess microbicide safety require strict adherence to prescribed regimens. If adherence is suboptimal, safety cannot be adequately assessed. MTN-017 was a phase 2, randomized sequence, open-label, expanded safety and acceptability crossover study comparing 1) daily oral emtricitabine/tenofovir disoproxil fumarate (FTC/TDF), 2) daily use of reduced-glycerin 1% tenofovir (RG-TFV) gel applied rectally, and 3) RG-TFV gel applied before and after receptive anal intercourse (RAI)—if participants had no RAI in a week, they were asked to use two doses of gel within 24 hours. Product use was assessed by mixed methods including unused product return count, text messaging reports, and qualitative plasma TFV pharmacokinetic (PK) results. Convergence interviews engaged participants in determining the most accurate number of doses used based on product count and text messaging reports. Client-centered adherence counseling was also used. Participants (N = 187) were men who have sex with men and transgender women enrolled in the United States (42%), Thailand (29%), Peru (19%) and South Africa (10%). Mean age was 31.4 years (range 18–64 years). Based on convergence interviews, over an 8-week period, 94% of participants had ≥80% adherence to daily tablet, 41% having perfect adherence; 83% had ≥80% adherence to daily gel, 29% having perfect adherence; and 93% had ≥80% adherence to twice-weekly use during the RAI-associated gel regimen, 75% having perfect adherence and 77% having ≥80% adherence to gel use before and after RAI. Only 4.4% of all daily product PK results were undetectable and unexpected (TFV concentrations <0.31 ng/mL) given self-reported product use near sampling date. The mixed methods adherence measurement indicated high adherence to product use in all three regimens. Adherence to RAI-associated rectal gel use was as high as adherence to daily oral PrEP. A rectal microbicide gel, if efficacious, could be an alternative for individuals uninterested in daily oral PrEP.
Collapse
|
141
|
Evaluation of a Multidrug Assay for Monitoring Adherence to a Regimen for HIV Preexposure Prophylaxis in a Clinical Study, HIV Prevention Trials Network 073. Antimicrob Agents Chemother 2017; 61:AAC.02743-16. [PMID: 28438932 DOI: 10.1128/aac.02743-16] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 04/17/2017] [Indexed: 02/06/2023] Open
Abstract
Daily oral tenofovir disoproxil fumarate (TDF)-emtricitabine (FTC) is a safe and effective intervention for HIV preexposure prophylaxis (PrEP). We evaluated the performance of a qualitative assay that detects 20 antiretroviral (ARV) drugs (multidrug assay) in assessing recent PrEP exposure (detection limit, 2 to 20 ng/ml). Samples were obtained from 216 Black men who have sex with men (208 HIV-uninfected men and 8 seroconverters) who were enrolled in a study in the United States evaluating the acceptability of TDF-FTC PrEP (165 of the uninfected men and 5 of the seroconverters accepted PrEP). Samples from 163 of the 165 HIV-uninfected men who accepted PrEP and samples from all 8 seroconverters were also tested for tenofovir (TFV) and FTC using a quantitative assay (detection limit for both drugs, 0.31 ng/ml). HIV drug resistance was assessed in seroconverter samples. The multidrug assay detected TFV and/or FTC in 3 (1.4%) of the 208 uninfected men at enrollment, 84 (40.4%) of the 208 uninfected men at the last study visit, and 1 (12.5%) of the 8 seroconverters. No other ARV drugs were detected. The quantitative assay confirmed all positive results from the multidrug assay and detected TFV and/or FTC in 9 additional samples (TFV range, 0.65 to 16.5 ng/ml; FTC range, 0.33 to 14.6 ng/ml). Resistance mutations were detected in 4 of the 8 seroconverter samples. The multidrug assay had 100% sensitivity and specificity for detecting TFV and FTC at drug concentrations consistent with daily PrEP use. The quantitative assay detected TFV and FTC at lower levels, which also might have provided protection against HIV infection.
Collapse
|
142
|
Brief Report: Medication Sharing Is Rare Among African HIV-1 Serodiscordant Couples Enrolled in an Efficacy Trial of Oral Pre-exposure Prophylaxis (PrEP) for HIV-1 Prevention. J Acquir Immune Defic Syndr 2017; 75:184-189. [PMID: 28291050 DOI: 10.1097/qai.0000000000001356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Sharing of pre-exposure prophylaxis (PrEP) medications is a concern for PrEP implementation. For HIV-1 serodiscordant couples, sharing may undermine the HIV-1 prevention benefit and also cause antiretroviral resistance if taken by HIV-1 infected partners. Within a PrEP efficacy trial among HIV-1 serodiscordant couples, we assessed the occurrence of PrEP sharing by self-report and plasma tenofovir concentrations in HIV-1 infected partners. PrEP sharing was self-reported at <0.01% of visits, and 0%-1.6% of randomly selected and 0% of purposively selected specimens from HIV-1 infected participants had detectable tenofovir concentrations (median: 66.5 ng/mL, range: 1.3-292 ng/mL). PrEP sharing within HIV-1 serodiscordant couples was extremely rare.
Collapse
|
143
|
Dolutegravir with tenofovir disoproxil fumarate-emtricitabine as HIV postexposure prophylaxis in gay and bisexual men. AIDS 2017; 31:1291-1295. [PMID: 28301425 DOI: 10.1097/qad.0000000000001447] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Completion rates for HIV postexposure prophylaxis (PEP) are often low. We investigated the adherence and safety of dolutegravir (DTG; 50 mg daily) with tenofovir disoproxil fumarate-emtricitabine (TDF-FTC; 300/200 mg, respectively) as three-drug PEP in gay and bisexual men. DESIGN Open-label, single-arm study at three sexual health clinics and two emergency departments in Australia. METHODS In total, 100 HIV-uninfected gay and bisexual men requiring PEP received DTG and TDF-FTC for 28 days. The primary end point was PEP failure (premature PEP cessation or primary HIV infection through week 12). Additional end points were adherence by self-report (n = 98) and pill count (n = 55), safety, and plasma drug levels at day 28. RESULTS PEP completion was 90% (95% confidence interval 84-96%). Failures (occurring at a median 9 days, interquartile range 3-16) comprised loss to follow-up (9%) and adverse event resulting in study drug discontinuation (headache, 1%). No participant was found to acquire HIV through week 12. Adherence to PEP was 98% by self-report and in the 55 participants with corresponding pill count data. The most common clinical adverse events were fatigue (26%), nausea (25%), diarrhoea (21%), and headache (10%). There were only four grade 3-4 subjective adverse events. The most common laboratory adverse event was raised alanine aminotransferase (22%), but there was no case of clinical hepatitis. At day 28, the mean estimated glomerular filtration rate decrease was 14 ml/min/1.73m (SD 17, P = 0.001); an estimated glomerular filtration rate of less than 60 ml/min/1.73m occurred in 3%. CONCLUSIONS DTG with TDF-FTC is a well tolerated option for once-daily PEP.
Collapse
|
144
|
Mayer KH, Safren SA, Elsesser SA, Psaros C, Tinsley JP, Marzinke M, Clarke W, Hendrix C, Wade Taylor S, Haberer J, Mimiaga MJ. Optimizing Pre-Exposure Antiretroviral Prophylaxis Adherence in Men Who Have Sex with Men: Results of a Pilot Randomized Controlled Trial of "Life-Steps for PrEP". AIDS Behav 2017; 21:1350-1360. [PMID: 27848089 DOI: 10.1007/s1046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Antiretroviral pre-exposure prophylaxis (PrEP) has been demonstrated to decrease HIV acquisition in multiple efficacy trials, but medication adherence is critical, and was suboptimal in several studies. Fifty HIV-uninfected at risk men who have sex with men (MSM) were randomized to a cognitive behavioral intervention condition or a time and session-matched comparison counseling intervention. The experimental intervention entailed four nurse-delivered initial and two booster sessions based on Life-Steps, an ART treatment adherence intervention. The comparison condition provided information and supportive counseling. The primary analyses compared adherence (Wisepill and tenofovir plasma levels) at 3 and 6 months. Fifty-eight MSM were screened to enroll 50 participants. Median age was 38.2 years old, 86% were white; 64% had completed college. Wisepill adherence was high in both groups, and not statistically different. Plasma tenofovir levels were significantly higher in the intervention group at 6 months using mean substitution analysis (i.e., computing missing variables) (p = 0.037), however, in the completer analyses (i.e., using only those completing all study visits), there were no statistically significant differences between randomization conditions. Medication adherence was high across a cognitive-behavioral (Life-Steps) and time-matched counseling intervention for PrEP adherence, with some evidence suggesting superiority of Life-Steps in this pilot RCT. Further evaluation in a fully powered efficacy trial is warranted to assess the robustness of this intervention.
Collapse
Affiliation(s)
- Kenneth H Mayer
- The Fenway Institute, Fenway Health, 1340 Boylston St, Boston, MA, 02215, USA.
- Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard T.C. Chan School of Public Health, Boston, MA, USA.
| | - Steven A Safren
- The Fenway Institute, Fenway Health, 1340 Boylston St, Boston, MA, 02215, USA
- University of Miami, Miami, FL, USA
| | - Steven A Elsesser
- The Fenway Institute, Fenway Health, 1340 Boylston St, Boston, MA, 02215, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Christina Psaros
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jake P Tinsley
- The Fenway Institute, Fenway Health, 1340 Boylston St, Boston, MA, 02215, USA
| | | | | | | | - S Wade Taylor
- The Fenway Institute, Fenway Health, 1340 Boylston St, Boston, MA, 02215, USA
- Wheelock College, Boston, MA, USA
| | - Jessica Haberer
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew J Mimiaga
- The Fenway Institute, Fenway Health, 1340 Boylston St, Boston, MA, 02215, USA
- Brown University, Providence, RI, USA
| |
Collapse
|
145
|
Mayer KH, Safren SA, Elsesser SA, Psaros C, Tinsley JP, Marzinke M, Clarke W, Hendrix C, Wade Taylor S, Haberer J, Mimiaga MJ. Optimizing Pre-Exposure Antiretroviral Prophylaxis Adherence in Men Who Have Sex with Men: Results of a Pilot Randomized Controlled Trial of "Life-Steps for PrEP". AIDS Behav 2017; 21:1350-1360. [PMID: 27848089 DOI: 10.1007/s10461-016-1606-4] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Antiretroviral pre-exposure prophylaxis (PrEP) has been demonstrated to decrease HIV acquisition in multiple efficacy trials, but medication adherence is critical, and was suboptimal in several studies. Fifty HIV-uninfected at risk men who have sex with men (MSM) were randomized to a cognitive behavioral intervention condition or a time and session-matched comparison counseling intervention. The experimental intervention entailed four nurse-delivered initial and two booster sessions based on Life-Steps, an ART treatment adherence intervention. The comparison condition provided information and supportive counseling. The primary analyses compared adherence (Wisepill and tenofovir plasma levels) at 3 and 6 months. Fifty-eight MSM were screened to enroll 50 participants. Median age was 38.2 years old, 86% were white; 64% had completed college. Wisepill adherence was high in both groups, and not statistically different. Plasma tenofovir levels were significantly higher in the intervention group at 6 months using mean substitution analysis (i.e., computing missing variables) (p = 0.037), however, in the completer analyses (i.e., using only those completing all study visits), there were no statistically significant differences between randomization conditions. Medication adherence was high across a cognitive-behavioral (Life-Steps) and time-matched counseling intervention for PrEP adherence, with some evidence suggesting superiority of Life-Steps in this pilot RCT. Further evaluation in a fully powered efficacy trial is warranted to assess the robustness of this intervention.
Collapse
Affiliation(s)
- Kenneth H Mayer
- The Fenway Institute, Fenway Health, 1340 Boylston St, Boston, MA, 02215, USA.
- Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard T.C. Chan School of Public Health, Boston, MA, USA.
| | - Steven A Safren
- The Fenway Institute, Fenway Health, 1340 Boylston St, Boston, MA, 02215, USA
- University of Miami, Miami, FL, USA
| | - Steven A Elsesser
- The Fenway Institute, Fenway Health, 1340 Boylston St, Boston, MA, 02215, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Christina Psaros
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jake P Tinsley
- The Fenway Institute, Fenway Health, 1340 Boylston St, Boston, MA, 02215, USA
| | | | | | | | - S Wade Taylor
- The Fenway Institute, Fenway Health, 1340 Boylston St, Boston, MA, 02215, USA
- Wheelock College, Boston, MA, USA
| | - Jessica Haberer
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew J Mimiaga
- The Fenway Institute, Fenway Health, 1340 Boylston St, Boston, MA, 02215, USA
- Brown University, Providence, RI, USA
| |
Collapse
|
146
|
Haberer JE, Sabin L, Amico KR, Orrell C, Galárraga O, Tsai AC, Vreeman RC, Wilson I, Sam‐Agudu NA, Blaschke TF, Vrijens B, Mellins CA, Remien RH, Weiser SD, Lowenthal E, Stirratt MJ, Sow PS, Thomas B, Ford N, Mills E, Lester R, Nachega JB, Bwana BM, Ssewamala F, Mbuagbaw L, Munderi P, Geng E, Bangsberg DR. Improving antiretroviral therapy adherence in resource-limited settings at scale: a discussion of interventions and recommendations. J Int AIDS Soc 2017; 20:21371. [PMID: 28630651 PMCID: PMC5467606 DOI: 10.7448/ias.20.1.21371] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 02/24/2017] [Indexed: 01/01/2023] Open
Abstract
Introduction: Successful population-level antiretroviral therapy (ART) adherence will be necessary to realize both the clinical and prevention benefits of antiretroviral scale-up and, ultimately, the end of AIDS. Although many people living with HIV are adhering well, others struggle and most are likely to experience challenges in adherence that may threaten virologic suppression at some point during lifelong therapy. Despite the importance of ART adherence, supportive interventions have generally not been implemented at scale. The objective of this review is to summarize the recommendations of clinical, research, and public health experts for scalable ART adherence interventions in resource-limited settings. Methods: In July 2015, the Bill and Melinda Gates Foundation convened a meeting to discuss the most promising ART adherence interventions for use at scale in resource-limited settings. This article summarizes that discussion with recent updates. It is not a systematic review, but rather provides practical considerations for programme implementation based on evidence from individual studies, systematic reviews, meta-analyses, and the World Health Organization Consolidated Guidelines for HIV, which include evidence from randomized controlled trials in low- and middle-income countries. Interventions are categorized broadly as education and counselling; information and communication technology-enhanced solutions; healthcare delivery restructuring; and economic incentives and social protection interventions. Each category is discussed, including descriptions of interventions, current evidence for effectiveness, and what appears promising for the near future. Approaches to intervention implementation and impact assessment are then described. Results and discussion: The evidence base is promising for currently available, effective, and scalable ART adherence interventions for resource-limited settings. Numerous interventions build on existing health care infrastructure and leverage available resources. Those most widely studied and implemented to date involve peer counselling, adherence clubs, and short message service (SMS). Many additional interventions could have an important impact on ART adherence with further development, including standardized counselling through multi-media technology, electronic dose monitoring, decentralized and differentiated models of care, and livelihood interventions. Optimal targeting and tailoring of interventions will require improved adherence measurement. Conclusions: The opportunity exists today to address and resolve many of the challenges to effective ART adherence, so that they do not limit the potential of ART to help bring about the end of AIDS.
Collapse
Affiliation(s)
- Jessica E. Haberer
- Massachusetts General Hospital Global Health, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Lora Sabin
- Department of Global Health, Center for Global Health and Department, Boston University School of Public Health, Boston, MA, USA
| | - K. Rivet Amico
- Department of Health Behavior and Health Education, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Catherine Orrell
- Desmond Tutu HIV Centre, Institute of Infectious Disease and Molecular Medicine, and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Omar Galárraga
- Department of Health Services, Policy and Practice, Brown University School of Public Health, Providence, RI, USA
| | - Alexander C. Tsai
- Chester M. Pierce, MD Division of Global Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Rachel C. Vreeman
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Academic Model Providing Access to Healthcare (AMPATH), Eldoret, Kenya
| | - Ira Wilson
- Department of Health Services, Policy and Practice, Brown University School of Public Health, Providence, RI, USA
| | - Nadia A. Sam‐Agudu
- Clinical Department, Institute of Human Virology Nigeria, Abuja, Nigeria
- Institute of Human Virology and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Terrence F. Blaschke
- Department of Medicine and Clinical Pharmacology, Stanford University School of Medicine, Stanford, CA, USA
| | - Bernard Vrijens
- Department of Biostatistics, University of Liège, Liège, Wallonia, Belgium
- WestRock Healthcare, Sion, Switzerland
| | - Claude A. Mellins
- HIV Center for Clinical and Behavioral Studies, NYSPI and Department of Psychiatry, Columbia; University, New York, NY, USA
| | - Robert H. Remien
- HIV Center for Clinical and Behavioral Studies, NYSPI and Department of Psychiatry, Columbia; University, New York, NY, USA
| | - Sheri D. Weiser
- Division of HIV, ID and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Elizabeth Lowenthal
- Departments of Pediatrics and Epidemiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael J. Stirratt
- Division of AIDS Research, National Institute of Mental Health, Bethesda, MD, USA
| | - Papa Salif Sow
- Bill and Melinda Gates Foundation, Seattle, WA, USA
- Department of Infectious diseases, University of Dakar, Dakar, Sénégal
| | | | - Nathan Ford
- Department of HIV/AIDS, World Health Organization, Geneva, Switzerland
| | - Edward Mills
- Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Richard Lester
- Division of Infectious Diseases, Department of Medicine, University of British Columbia
| | - Jean B. Nachega
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Bosco Mwebesa Bwana
- Department of Medicine, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Fred Ssewamala
- Columbia University School of Social Work & School of International and Public Affairs, New York, NY, USA
| | - Lawrence Mbuagbaw
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Paula Munderi
- HIV Care Research Program, Medical Research Council, Uganda Virus Research Institute, Entebbe, Uganda
| | - Elvin Geng
- Division of HIV, Infectious Disease and Global Medicine, San Francisco General Hospital, Department of Medicine, University of California, San Francisco, CA, USA
| | - David R. Bangsberg
- Oregon Health & Sciences University‐Portland State University School of Public Health, Portland, OR, USA
| |
Collapse
|
147
|
Does Adherence Change When No One is Looking? Comparing Announced and Unannounced Tenofovir Levels in a PrEP Trial. AIDS Behav 2016; 20:2639-2643. [PMID: 26781865 DOI: 10.1007/s10461-016-1292-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Differences between unannounced and announced tenofovir levels as measures of PrEP adherence are not well understood. In an ancillary adherence study involving one urban site (Kampala) and two rural sites (Kabwohe and Tororo) from the Partners PrEP study, 268 specimen pairs from chronologically proximal clinic and home visits were tested for plasma tenofovir levels. Comparing clinic and home specimens, 89 versus 89 % were classified as detectable (>0.31 ng/ml; p = 0.77), 87 versus 86 % as recent dosing (>10 ng/ml; p = 0.80), and 82 versus 80 % as steady-state (>40 ng/ml; p = 0.44). Mean difference between announced and unannounced drug levels, adjusted for specimen collection time was 3.2 ng/ml (p = 0.50) for Kabwohe, 23.2 ng/ml (p = 0.003) for Kampala and -3.3 ng/ml p = 0.69) for Tororo. In the setting of high adherence, plasma tenofovir levels tested at the clinic were categorically similar as levels tested at home; however, differences were seen between urban and rural settings.
Collapse
|
148
|
Emtricitabine-Triphosphate in Dried Blood Spots as a Marker of Recent Dosing. Antimicrob Agents Chemother 2016; 60:6692-6697. [PMID: 27572401 DOI: 10.1128/aac.01017-16] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/20/2016] [Indexed: 11/20/2022] Open
Abstract
New objective measures of antiretroviral adherence are needed. We determined if emtricitabine triphosphate (FTC-TP) in dried blood spots (DBS) can be used as a marker of recent dosing with tenofovir disoproxil fumarate-emtricitabine (TDF-FTC). The half-life of FTC-TP was estimated in DBS samples obtained from an intensive pharmacokinetic (PK) study of coformulated TDF-FTC in HIV-negative and HIV-infected participants. The concordance of quantifiable FTC-TP in DBS with tenofovir (TFV)/FTC in plasma was evaluated by utilizing paired plasma-DBS samples from participants enrolled in 2 large preexposure prophylaxis (PrEP) open-label trials. The time to FTC-TP nondetectability after TDF-FTC dosing was evaluated utilizing DBS from HIV-negative participants enrolled in a directly observed therapy study of variable adherence to TDF-FTC. The mean (95% confidence interval [CI]) terminal half-life of FTC-TP in the PK study was 35 (23 to 47) h. A total of 143/163 (88%) samples obtained 0 to 48 h post-TDF-FTC dose had quantifiable FTC-TP in DBS, compared with 2/93 (2%) and 0/87 (0%) obtained >48 and >96 h postdose. In 746 paired plasma-DBS samples from 445 participants enrolled in PrEP trials, when both TFV/FTC in plasma were below the limit of quantification, FTC-TP was as well in 98.9% of the samples, and when either TFV or FTC in plasma was quantifiable, FTC-TP was as well in 90.5% of the samples. The half-life of FTC-TP in DBS is short relative to that of TFV-diphosphate (TFV-DP), making it a surrogate for TFV-FTC detection in plasma. FTC-TP can be quantified in DBS simultaneously with TFV-DP, which quantifies cumulative adherence to TDF-FTC. (The clinical trials discussed in this article have been registered at ClinicalTrials.gov under identifiers NCT01040091, NCT02022657, NCT00458393, NCT01772823, and NCT02012621.).
Collapse
|
149
|
Fonsart J, Saragosti S, Taouk M, Peytavin G, Bushman L, Charreau I, Hance A, Goldwirt L, Morel S, Mammano F, Loze B, Capitant C, Clavel F, Mahjoub N, Meyer L, Anderson PL, Delaugerre C, Molina JM. Single-dose pharmacokinetics and pharmacodynamics of oral tenofovir and emtricitabine in blood, saliva and rectal tissue: a sub-study of the ANRS IPERGAY trial. J Antimicrob Chemother 2016; 72:478-485. [PMID: 28073964 DOI: 10.1093/jac/dkw412] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 08/20/2016] [Accepted: 08/28/2016] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVES In the ANRS IPERGAY pre-exposure prophylaxis (PrEP) trial, a single dose of tenofovir disoproxil fumarate and emtricitabine was taken orally 2-24 h before sexual intercourse. A sub-study was conducted to assess the pharmacokinetics of tenofovir and emtricitabine in blood, saliva and rectal tissue following this initial oral intake. METHODS Plasma, PBMC, saliva and rectal tissue sampling was performed over 24 h in 12 seronegative men before enrolment in the ANRS IPERGAY trial, following a single dose of 600 mg tenofovir disoproxil fumarate/400 mg emtricitabine. Ex vivo HIV infectibility of rectal biopsies was also assessed. RESULTS The median plasma Tmax of tenofovir (median Cmax: 401 μg/L) and emtricitabine (median Cmax: 2868 μg/L) was obtained 1 h (range: 0.5-4) and 2 h (range: 1-4) after dosing, respectively. The median C24 of tenofovir and emtricitabine was 40 and 63 μg/L, respectively. The median PBMC tenofovir diphosphate and emtricitabine triphosphate levels were 12.2 and 16.7 fmol/106 cells and 2800 and 2000 fmol/106 cells at 2 and 24 h after dosing, respectively. Saliva/plasma AUC0-24 ratios were 2% and 17% for tenofovir and emtricitabine, respectively. Emtricitabine was detected in rectal tissue 30 min after dosing, whereas tenofovir was only detectable at 24 h. Ex vivo HIV infectibility assays of rectal biopsies showed partial protection after dosing (P < 0.07). DISCUSSION A single high dose of oral tenofovir disoproxil fumarate/emtricitabine provides rapid and high blood levels of tenofovir and emtricitabine, with rapid diffusion of emtricitabine in saliva and rectal tissue.
Collapse
Affiliation(s)
- Julien Fonsart
- Department of Biochemistry, AP-HP, Hôpital Saint-Louis, Paris, France
| | | | - Milad Taouk
- Department of Infectious Diseases, Hôpital Saint-Louis, AP-HP and University of Paris Diderot, Paris Sorbonne Cité, France
| | - Gilles Peytavin
- Department of Pharmacology, Hôpital Bichat Claude Bernard, Paris, France
| | | | | | | | - Lauriane Goldwirt
- Department of Pharmacology, AP-HP, Hôpital Saint-Louis, Paris, France
| | | | | | - Bénédicte Loze
- Department of Infectious Diseases, Hôpital Saint-Louis, AP-HP and University of Paris Diderot, Paris Sorbonne Cité, France
| | | | | | - Nadia Mahjoub
- Department of Virology, AP-HP, Hôpital Saint-Louis, University of Paris Diderot, Paris, France
| | | | | | - Constance Delaugerre
- INSERM U941, Paris, France.,Department of Virology, AP-HP, Hôpital Saint-Louis, University of Paris Diderot, Paris, France
| | - Jean-Michel Molina
- INSERM U941, Paris, France .,Department of Infectious Diseases, Hôpital Saint-Louis, AP-HP and University of Paris Diderot, Paris Sorbonne Cité, France
| |
Collapse
|
150
|
Seifert SM, Chen X, Meditz AL, Castillo-Mancilla JR, Gardner EM, Predhomme JA, Clayton C, Austin G, Palmer BE, Zheng JH, Klein B, Kerr BJ, Guida LA, Rower C, Rower JE, Kiser JJ, Bushman LR, MaWhinney S, Anderson PL. Intracellular Tenofovir and Emtricitabine Anabolites in Genital, Rectal, and Blood Compartments from First Dose to Steady State. AIDS Res Hum Retroviruses 2016; 32:981-991. [PMID: 27526873 DOI: 10.1089/aid.2016.0008] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The pharmacokinetics (PK) of tenofovir-diphosphate (TFV-DP) and emtricitabine-triphosphate (FTC-TP), the active anabolites of tenofovir disoproxil fumarate (TDF), and emtricitabine (FTC) in blood, genital, and rectal compartments was determined in HIV-positive and seronegative adults who undertook a 60-day intensive PK study of daily TDF/FTC (plus efavirenz in HIV positives). Lymphocyte cell sorting, genital, and rectal sampling occurred once per subject, at staggered visits. Among 19 HIV-positive (3 female) and 21 seronegative (10 female) adults, TFV-DP in peripheral blood mononuclear cells (PBMC) accumulated 8.6-fold [95% confidence interval (CI): 7.2-10] from first-dose to steady-state concentration (Css) versus 1.7-fold (95% CI: 1.5-1.9) for FTC-TP. Css was reached in ∼11 and 3 days, respectively. Css values were similar between HIV-negative and HIV-positive individuals. Css TFV-DP in rectal mononuclear cells (1,450 fmol/106 cells, 898-2,340) was achieved in 5 days and was >10 times higher than PBMC (95 fmol/106 cells, 85-106), seminal cells (22 fmol/106 cells, 6-79), and cervical cells (111 fmol/106 cells, 64-194). FTC-TP Css was highest in PBMC (5.7 pmol/106 cells, 5.2-6.1) and cervical cells (7 pmol/106 cells, 2-19) versus rectal (0.8 pmol/106 cells, 0.6-1.1) and seminal cells (0.3 pmol/106 cells, 0.2-0.5). Genital drug concentrations on days 1-7 overlapped with estimated Css, but accumulation characteristics were based on limited data. TFV-DP and FTC-TP in cell sorted samples were highest and achieved most rapidly in CD14+ compared with CD4+, CD8+, and CD19+ cells. Together, these findings demonstrate cell-type and tissue-dependent cellular pharmacology, preferential accumulation of TFV-DP in rectal mononuclear cells, and rapid distribution into rectal and genital compartments.
Collapse
Affiliation(s)
- Sharon M. Seifert
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Xinhui Chen
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Amie L. Meditz
- Boulder Community Hospital, Beacon Center for Infectious Diseases, Boulder, Colorado
| | - Jose R. Castillo-Mancilla
- Division of Infectious Diseases, School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | | | - Julie A. Predhomme
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Carolyn Clayton
- Department of Biostatistics and Informatics, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Gregory Austin
- School of Medicine, Internal Medicine, Gastroenterology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Brent E. Palmer
- Division of Allergy and Clinical Immunology, School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Jia-Hua Zheng
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Brandon Klein
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Becky J. Kerr
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - L. Anthony Guida
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Caitlin Rower
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Joseph E. Rower
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Jennifer J. Kiser
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Lane R. Bushman
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Samantha MaWhinney
- Department of Biostatistics and Informatics, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Peter L. Anderson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|