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

Kathleen L Vincent, John A Moss, Mark A Marzinke, Craig W Hendrix, Peter A Anton, Richard B Pyles, Kate M Guthrie, Lauren Dawson, Trevelyn J Olive, Irina Butkyavichene, Scott A Churchman, John M Cortez Jr, Rob Fanter, Manjula Gunawardana, Christine S Miller, Flora Yang, Rochelle K Rosen, Sara E Vargas, Marc M Baum, Kathleen L Vincent, John A Moss, Mark A Marzinke, Craig W Hendrix, Peter A Anton, Richard B Pyles, Kate M Guthrie, Lauren Dawson, Trevelyn J Olive, Irina Butkyavichene, Scott A Churchman, John M Cortez Jr, Rob Fanter, Manjula Gunawardana, Christine S Miller, Flora Yang, Rochelle K Rosen, Sara E Vargas, Marc M Baum

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.

Conflict of interest statement

I have read the journal's policy and the authors of this manuscript have the following competing interests: During part of the time of the study, IB, SAC, and JMC were employees of Auritec and participated in manufacturing the vaginal rings; they were no longer employees at the time of sample collection, data analysis, and manuscript preparation. Prior to August 2016, MG was a 50% shared employee of Auritec and participated in the residual drug measurements and coordinating sample distribution. JAM and MMB are co-inventors on patent applications that include aspects of the intravaginal ring device that was used in the clinical trial described in this manuscript and have received funding for preclinical research relevant to HIV (but not vaginal rings) from ViiV/GSK. They also have received funding from the International Partnership for Microbicides, CONRAD, and the Population Council. JAM, MMB, and MG are co-inventors on the following patent applications relevant to HIV: https://patents.google.com/patent/WO2016149561A1/en and https://patents.google.com/patent/WO2017161136A1/en. KLV and RBP are paid consultants to ABL, Inc. CWH has received funding for clinical research from ViiV/GSK managed via Johns Hopkins University and was a consultant for 1-2 days in 2017 to ViiV/GSK.

Figures

Fig 1. CVF drug levels and washout…
Fig 1. CVF drug levels and washout following TDF pod-IVR use.
ARV drug levels in CVF fall off rapidly following IVR removal (grey arrow) but are quantifiable for a further 2 wk. Every circular datum represents an individual sample from one of the participants (n = 6), while triangles depict samples that were BLQ of the analytical method, and values were calculated as follows: [(assay BLQ)/2]/(median swab mass). (A) TDF. (B) TFV. (C) Box plots of ARV drug terminal half-lives of elimination from CVF. The box extends from the 25th to 75th percentiles, with the horizontal line in the box representing the median; whiskers represent the lowest and highest datum. Comparison of the TDF and TFV groups using an unpaired t test with Welch’s correction showed that the groups were significantly different (P = 0.0053). ARV, antiretroviral; BLQ, below the lower limit of quantitation; CVF, cervicovaginal fluid; IVR, intravaginal ring; TDF, tenofovir disoproxil fumarate; TFV, tenofovir.
Fig 2. CVF drug levels and washout…
Fig 2. CVF drug levels and washout following TDF-FTC pod-IVR use.
ARV drug levels in CVF fall off rapidly following IVR removal (grey arrow) but are quantifiable for a further 2 wk. Every circular datum represents an individual sample from one of the participants (n = 6), while triangles depict samples that were BLQ of the analytical method, and values were calculated as follows: [(assay BLQ)/2]/(median swab mass). (A) TDF. (B) TFV. (C) FTC. (D) Box plots of ARV drug terminal half-lives of elimination from CVF. The box extends from the 25th to 75th percentiles, with the horizontal line in the box representing the median; whiskers represent the lowest and highest datum. Comparison of the TDF and TFV groups using an unpaired t test with Welch’s correction showed that the groups were significantly different (P = 0.0131). ARV, antiretroviral; BLQ, below the lower limit of quantitation; CVF, cervicovaginal fluid; FTC, emtricitabine; IVR, intravaginal ring; TDF, tenofovir disoproxil fumarate; TFV, tenofovir.
Fig 3. CVF drug levels and washout…
Fig 3. CVF drug levels and washout following TDF-FTC-MVC pod-IVR use.
ARV drug levels in CVFs fall off rapidly following IVR removal (grey arrows) but are quantifiable for a further 2 wk. Every circular datum represents an individual sample from one of the participants (n = 6), while triangles depict samples that were BLQ of the analytical method, and values were calculated as follows: [(assay BLQ)/2]/(median swab mass). (A) TDF. (B) TFV. (C) FTC. (D) MVC. (E) Box plots of ARV drug terminal half-lives of elimination from CVF. The box extends from the 25th to 75th percentiles, with the horizontal line in the box representing the median; whiskers represent the lowest and highest datum. Comparison of the TDF, FTC, and MVC groups using an ordinary 1-way ANOVA analysis (no matching or pairing of the data) showed that they were not significantly different (P = 0.8973). However, comparison of the TDF and TFV groups using an unpaired t test with Welch’s correction, showed that the groups were significantly different (P = 0.0327). ANOVA, analysis of variance; ARV, antiretroviral; BLQ, below the lower limit of quantitation; CVF, cervicovaginal fluid; FTC, emtricitabine; IVR, intravaginal ring; MVC, maraviroc; TDF, tenofovir disoproxil fumarate; TFV, tenofovir.
Fig 4. Drug and drug metabolite exposure…
Fig 4. Drug and drug metabolite exposure in VT at TDF and TDF-FTC pod-IVR removal (Day 7).
(A) ARV drug and TFV-DP levels in vaginal biopsies collected on Day 7 within 30 min of pod-IVR removal. Analyte concentrations are all expressed as fmol/mg of tissue homogenate to allow meaningful cross-comparison. Every circular datum represents an individual sample from one of the participants (n = 6); horizontal lines represent group medians; blue, TFV; green, TFV-DP; red, FTC. (B) Paired CVF:VT concentration ratios of TFV (molar sum of TDF and TFV concentrations) and FTC at Day 7; horizontal lines represent group means. The ratios provide a measure of the extent of tissue penetration for each analyte following vaginal delivery and, hence, vaginal bioavailability. The CVF TFV concentrations used in this calculation were the molar sum of the measured TFV and TDF concentrations as the prodrug hydrolyzes to TFV in the vaginal mucosa. ARV, antiretroviral; CVF, cervicovaginal fluid; FTC, emtricitabine; IVR, intravaginal ring; TDF, tenofovir disoproxil fumarate; TFV, tenofovir; TFV-DP, tenofovir diphosphate; VT, vaginal tissue.
Fig 5. Drug and drug metabolite exposure…
Fig 5. Drug and drug metabolite exposure in VTs at TDF-FTC-MVC pod-IVR removal (Day 7).
(A) ARV drug and TFV-DP levels in vaginal biopsies collected on Day 7 within 30 min of pod-IVR removal. Analyte concentrations are all expressed as fmol/mg of tissue homogenate to allow meaningful cross-comparison. Every circular datum represents an individual sample from one of the participants (n = 6), while inverted triangles depict samples that were ALQ of the analytical method, and values were calculated as follows: (assay ALQ)/(median biopsy mass); horizontal lines represent group medians. (B) Paired CVF:VT concentration ratios of total TFV (molar sum of TDF and TFV concentrations), FTC, and MVC at Day 7; horizontal lines represent group means. The ratios describe the extent of tissue penetration for each analyte following vaginal delivery and, hence, vaginal bioavailability. The CVF TFV concentrations used in this calculation were the molar sum of the measured TFV and TDF concentrations, as the prodrug hydrolyzes to TFV in the vaginal mucosa. The two MVC VT samples that were ALQ were omitted from the analysis. There was no statistically significant difference between the three groups according to an unpaired, nonparametric Kruskal–Wallis test (P = 0.1591). ALQ, above the upper limit of quantitation; ARV, antiretroviral; CVF, cervicovaginal fluid; FTC, emtricitabine; IVR, intravaginal ring; MVC, maraviroc; TDF, tenofovir disoproxil fumarate; TFV, tenofovir; TFV-DP, tenofovir diphosphate; VT, vaginal tissue.

References

    1. Birx DL. Delivering an AIDS-free generation [Presentation]: The Henry J. Kaiser Family Foundation; 2014.
    1. Fast-Track: Ending the AIDS Epidemic by 2030. Geneva, Switzerland: UNAIDS, 2014.
    1. Prevention Gap Report. Geneva, Switzerland: UNAIDS, 2016.
    1. Abdool Karim Q, Abdool Karim SS, Frohlich JA, Grobler AC, Baxter C, Mansoor LE, et al. Effectiveness and Safety of Tenofovir Gel, an Antiretroviral Microbicide, for the Prevention of HIV Infection in Women. Science. 2010;329(5996):1168–74. 10.1126/science.1193748
    1. Grant RM, Lama JR, Anderson PL, McMahan V, Liu AY, Vargas L, et al. Preexposure Chemoprophylaxis for HIV Prevention in Men Who Have Sex with Men. N Engl J Med. 2010;363(27):2587–99. PubMed PMID: WOS:000285763700004. 10.1056/NEJMoa1011205
    1. Baeten JM, Donnell D, Ndase P, Mugo NR, Campbell JD, Wangisi J, et al. Antiretroviral prophylaxis for HIV prevention in heterosexual men and women. N Engl J Med. 2012;367(5):399–410. 10.1056/NEJMoa1108524 ; PubMed Central PMCID: PMC3770474.
    1. Thigpen MC, Kebaabetswe PM, Paxton LA, Smith DK, Rose CE, Segolodi TM, et al. Antiretroviral Preexposure Prophylaxis for Heterosexual HIV Transmission in Botswana. N Engl J Med. 2012;367(5):423–34. 10.1056/NEJMoa1110711 PubMed PMID: WOS:000307001900007.
    1. Choopanya K, Martin M, Suntharasamai P, Sangkum U, Mock PA, Leethochawalit M, et al. Antiretroviral Prophylaxis for HIV Infection in Injecting Drug Users in Bangkok, Thailand (the Bangkok Tenofovir Study): a Randomised, Double-blind, Placebo-controlled Phase 3 Trial. Lancet. 2013;381(9883):2083–90. 10.1016/S0140-6736(13)61127-7
    1. Molina JM, Capitant C, Spire B, Pialoux G, Cotte L, Charreau I, et al. On-demand Preexposure Prophylaxis in Men at High Risk for HIV-1 Infection. N Engl J Med. 2015;373(23):2237–46. 10.1056/NEJMoa1506273 PubMed PMID: WOS:000365743100007.
    1. Marcus JL, Hurley LB, Hare CB, Nguyen DP, Phengrasamy T, Silverberg MJ, et al. Preexposure Prophylaxis for HIV Prevention in a Large Integrated Health Care System: Adherence, Renal Safety, and Discontinuation. J Acquir Immune Defic Syndr. 2016;73(5):540–6. PubMed PMID: WOS:000389045300011. 10.1097/QAI.0000000000001129
    1. McCormack S, Dunn DT, Desai M, Dolling DI, Gafos M, Gilson R, et al. Pre-exposure Prophylaxis to Prevent the Acquisition of HIV-1 Infection (PROUD): Effectiveness Results from the Pilot Phase of a Pragmatic Open-label Randomised Trial. Lancet. 2016;387(10013):53–60. 10.1016/S0140-6736(15)00056-2 PubMed PMID: WOS:000367457300027.
    1. Roehr B. FDA Approves First Drug to Prevent HIV Infection. B M J. 2012;345:e4879.
    1. Sheth AN, Rolle CP, Gandhi M. HIV Pre-exposure Prophylaxis for Women. J Virus Erad. 2(3):149–55.
    1. Amico KR, Mansoor LE, Corneli A, Torjesen K, van der Straten A. Adherence Support Approaches in Biomedical HIV Prevention Trials: Experiences, Insights and Future Directions from Four Multisite Prevention Trials. AIDS Behav. 2013;17(6):2143–55. 10.1007/s10461-013-0429-9
    1. Kruse W, Eggertkruse W, Rampmaier J, Runnebaum B, Weber E. Dosage Frequency and Drug Compliance Behavior—a Comparative Study on Compliance with a Medication to Be Taken Twice or 4 Times Daily. Eur J Clin Pharmacol. 1991;41(6):589–92. 10.1007/BF00314990 PubMed PMID: WOS:A1991GU48100016.
    1. Sershen S, West J. Implantable, Polymeric Systems for Modulated Drug Delivery. Adv Drug Deliv Rev. 2002;54(9):1225–35. 10.1016/s0169-409x(02)00090-x PubMed PMID: WOS:000179294000006.
    1. Kutilek VD, Sheeter DA, Elder JH, Torbett BE. Is Resistance Futile? Curr Drug Targets Infect Disord. 2003;3(4)::295–309.
    1. Yeaw J, Benner JS, Walt JG, Sian S, Smith DB. Comparing Adherence and Persistence Across 6 Chronic Medication Classes. J Manag Care Pharm. 2009;15(9):728–40. PubMed PMID: WOS:000272878200001. doi:
    1. Moss JA, Baum MM. Microbicide Vaginal Rings In: das Neves J, Sarmento B, editors. Drug Delivery and Development of Anti-HIV Microbicides. Singapore: Pan Stanford Publishing; 2014. p. 221–90.
    1. McGowan I. Injectable and Implantable Antiretroviral Strategies for HIV Prevention. Future Virol. 2015;10(10):1163–76. PubMed PMID: WOS:000364917600006.
    1. Baeten JM, Palanee-Phillips T, Brown ER, Schwartz K, Soto-Torres LE, Govender V, et al. Use of a Vaginal Ring Containing Dapivirine for HIV-1 Prevention in Women. N Engl J Med. 2016;375:2121–32. 10.1056/NEJMoa1506110
    1. Nel A, van Niekerk N, Kapiga S, Bekker LG, Gama C, Gill K, et al. Safety and Efficacy of a Dapivirine Vaginal Ring for HIV Prevention in Women. N Engl J Med. 2016;375(22):2133–43. 10.1056/NEJMoa1602046 PubMed PMID: WOS:000389030400005.
    1. Guthrie KM, Vargas S, Shaw JG, Rosen RK, van den Berg JJ, Kiser PF, et al. The Promise of Intravaginal Rings for Prevention: User Perceptions of Biomechanical Properties and Implications for Prevention Product Development. PLoS ONE. 2015;10(12):e0145642 10.1371/journal.pone.0145642
    1. Rosen RK, van den Berg JJ, Vargas SE, Senocak N, Shaw JG, Buckheit RW, et al. Meaning-making Matters in Product Design: Users' Sensory Perceptions and Experience Evaluations of Long-acting Vaginal Gels and Intravaginal Rings. Contraception. 2015;92(6):596–601. 10.1016/j.contraception.2015.08.007 PubMed PMID: WOS:000365556500015.
    1. Baum MM, Butkyavichene I, Gilman J, Kennedy S, Kopin E, Malone AM, et al. An Intravaginal Ring for the Simultaneous Delivery of Multiple Drugs. J Pharm Sci. 2012;101(8):2833–43. PubMed Central PMCID: PMC3857731. 10.1002/jps.23208
    1. Moss JA, Srinivasan P, Smith TJ, Butkyavichene I, Lopez G, Brooks AA, et al. Pharmacokinetics and Preliminary Safety Study of Pod-Intravaginal Rings Delivering Antiretroviral Combinations for HIV Prophylaxis in a Macaque Model. Antimicrob Agents Chemother. 2014;58(9):5125–35. PubMed Central PMCID: PMC4135875. 10.1128/AAC.02871-14
    1. Smith JM, Moss JA, Srinivasan P, Butkyavichene I, Gunawardana M, Fanter R, et al. Novel Multipurpose Pod-intravaginal Ring for the Prevention of HIV, HSV, and Unintended Pregnancy: Pharmacokinetic Evaluation in a Macaque Model. PLoS ONE. 2017;12(10):e0185946 10.1371/journal.pone.0185946
    1. Baum MM, Butkyavichene I, Churchman SA, Lopez G, Miller CS, Smith TJ, et al. An Intravaginal Ring for the Sustained Delivery of Tenofovir Disoproxil Fumarate. Int J Pharm. 2015;495(1):579–87. PubMed Central PMCID: PMC4609628. 10.1016/j.ijpharm.2015.09.028
    1. Moss JA, Butkyavichene I, Churchman SA, Gunawardana M, Fanter R, Miller CS, et al. Combination Pod-intravaginal Ring Delivers Antiretroviral Agents for HIV Prophylaxis: Pharmacokinetic Evaluation in an Ovine Model. Antimicrob Agents Chemother. 2016;60(6):3759–66. 10.1128/AAC.00391-16
    1. Nugent RP, Krohn MA, Hillier SL. Reliability of Diagnosing Bacterial Vaginosis Is Improved by a Standardized Method of Gram Stain Interpretation. J Clin Microbiol. 1991;29(2):297–301. PubMed PMID: WOS:A1991ET57800014.
    1. Pyles RB, Vincent KL, Baum MM, Elsom B, Miller AL, Maxwell C, et al. Cultivated Vaginal Microbiomes Alter HIV-1 Infection and Antiretroviral Efficacy in Colonized Epithelial Multilayer Cultures. PLoS ONE. 2014;9(3):e93419 PubMed Central PMCID: PMC3968159. 10.1371/journal.pone.0093419
    1. Hendrix CW, Chen BA, Guddera V, Hoesley C, Justman J, Nakabiito C, et al. MTN-001: Randomized Pharmacokinetic Cross-over Study Comparing Tenofovir Vaginal Gel and Oral Tablets in Vaginal Tissue and Other Compartments. PLoS ONE. 2013;8(1):e55013 10.1371/journal.pone.0055013
    1. Parsons TL, Emory JF, Seserko LA, Aung WS, Marzinke MA. Dual Quantification of Dapivirine and Maraviroc in Cervicovaginal Secretions from Ophthalmic Tear Strips and Polyester-based Swabs via Liquid Chromatographic-tandem Mass Spectrometric (LC-MS/MS) Analysis. J Pharm Biomed Anal. 2014;98:407–16. 10.1016/j.jpba.2014.06.018 PubMed PMID: WOS:000339859800055.
    1. Hendrix CW, Andrade A, Bumpus NN, Kashuba AD, Marzinke MA, Moore A, et al. Dose Frequency Ranging Pharmacokinetic Study of Tenofovir-Emtricitabine After Directly Observed Dosing in Healthy Volunteers to Establish Adherence Benchmarks (HPTN 066). AIDS Res Hum Retrovir. 2016;32(1):32–43. 10.1089/AID.2015.0182 PubMed PMID: WOS:000367335100006.
    1. Parsons TL, Marzinke MA. Development and Validation of a Liquid Chromatographic-tandem Mass Spectrometric Method for the Multiplexed Quantification of Etravirine, Maraviroc, Raltegravir, and Rilpivirine in Human Plasma and Tissue. J Pharm Biomed Anal. 2016;131:333–44. 10.1016/j.jpba.2016.08.016 PubMed PMID: WOS:000387628400044.
    1. US FDA. Guidance for Industry: Bioanalytical Method Validation Rockville, MD: U.S. Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER), Center for Veterinary Medicine (CVM); 2001. p. 22.
    1. Churchman SA, Moss JA, Baum MM. Accurate Measurement of Female Genital Tract Fluid Dilution in Cervicovaginal Lavage Samples. J Chromatogr B. 2016;1017:75–81.
    1. Guest G, MacQueen KM, Namey EE. Applied Thematic Analysis Thousand Oaks: SAGE Publications; 2012. 360 p.
    1. Guthrie KM, Rosen RK, Vargas SE, Getz ML, Dawson L, Guillen M, et al. User Evaluations Offer Promise for Pod-intravaginal Ring as a Drug Delivery Platform: A Mixed Methods Study of Acceptability and Use Experiences. PLoS ONE. 2018;13(5):e0197269 10.1371/journal.pone.0197269
    1. Moss JA, Baum MM, Malone AM, Kennedy S, Kopin E, Nguyen C, et al. Tenofovir and Tenofovir Disoproxil Pharmacokinetics from Intravaginal Rings. AIDS. 2012;26(6):707–10. PubMed Central PMCID: PMC3855348. 10.1097/QAD.0b013e3283509abb
    1. Belec L, Meillet D, Levy M, Georges A, Tevibenissan C, Pillot J. Dilution Assessment of Cervicovaginal Secretions Obtained by Vaginal Washing for Immunological Assays. Clin Diagn Lab Immunol. 1995;2(1):57–61. PubMed PMID: WOS:A1995QA64900011.
    1. Baeten JM, Donnell D, Ndase P, Mugo NR, Campbell JD, Wangisi J, et al. Antiretroviral Prophylaxis for HIV Prevention in Heterosexual Men and Women. N Engl J Med. 2012;367(5):399–410. 10.1056/NEJMoa1108524 PubMed PMID: WOS:000307001900005.
    1. Keller MJ, Mesquita PM, Marzinke MA, Teller R, Espinoza L, Atrio JM, et al. A Phase 1 Randomized Placebo-controlled Safety and Pharmacokinetic Trial of a Tenofovir Disoproxil Fumarate Vaginal Ring. AIDS. 2016;30(5):743–51. 10.1097/QAD.0000000000000979 PubMed PMID: WOS:000371309700001.
    1. Louissaint NA, Cao YJ, Skipper PL, Liberman RG, Tannenbaum SR, Nimmagadda S, et al. Single Dose Pharmacokinetics of Oral Tenofovir in Plasma, Peripheral Blood Mononuclear Cells, Colonic Tissue, and Vaginal Tissue. AIDS Res Hum Retroviruses. 2013;29(11):1443–50. 10.1089/AID.2013.0044 PubMed PMID: WOS:000326037500505.
    1. Srinivasan P, Moss JA, Gunawardana M, Churchman SA, Yang F, Dinh CT, et al. Topical Delivery of Tenofovir Disoproxil Fumarate and Emtricitabine from Pod-intravaginal Rings Protect Macaques from Multiple SHIV Exposures. PLoS ONE. 2016;11(6):e0157061 PubMed Central PMCID: PMC4898685. 10.1371/journal.pone.0157061
    1. Smith JM, Rastogi R, Teller RS, Srinivasan P, Mesquita PM, Nagaraja U, et al. Intravaginal Ring Eluting Tenofovir Disoproxil Fumarate Completely Protects Macaques from Multiple Vaginal Simian-HIV Challenges. Proc Natl Acad Sci U S A. 2013;110(40):16145–50. 10.1073/pnas.1311355110
    1. Dorr P, Westby M, Dobbs S, Griffin P, Irvine B, Macartney M, et al. Maraviroc (UK-427,857), a Potent, Orally Bioavailable, and Selective Small-molecule Inhibitor of Chemokine Receptor CCR5 with Broad-spectrum Anti-human Immunodeficiency Virus Type 1 Activity. Antimicrob Agents Chemother. 2005;49(11):4721–32. 10.1128/AAC.49.11.4721-4732.2005 PubMed PMID: ISI:000233020900039.
    1. Chen BA, Panther L, Marzinke MA, Hendrix CW, Hoesley CJ, van der Straten A, et al. Phase 1 Safety, Pharmacokinetics, and Pharmacodynamics of Dapivirine and Maraviroc Vaginal Rings: A Double-blind Randomized Trial. J Acquir Immune Defic Syndr. 2015;70(3):242–9. 10.1097/QAI.0000000000000702
    1. Baeten JM, Palanee-Phillips T, Brown ER, Schwartz K, Soto-Torres LE, Govender V, et al. Use of a Vaginal Ring Containing Dapivirine for HIV-1 Prevention in Women. N Engl J Med. 2016;375(22):2121–32. 10.1056/NEJMoa1506110 ; PubMed Central PMCID: PMC4993693.
    1. Nel A, van Niekerk N, Kapiga S, Bekker LG, Gama C, Gill K, et al. Safety and Efficacy of a Dapivirine Vaginal Ring for HIV Prevention in Women. N Engl J Med. 2016;375(22):2133–43. 10.1056/NEJMoa1602046 .
    1. Brady M, Manning J. Lessons from Reproductive Health to Inform Multipurpose Prevention Technologies: Don't Reinvent the Wheel. Antiviral Res. 2013;100:S25–S31. 10.1016/j.antiviral.2013.09.019 PubMed PMID: WOS:000328923200005.
    1. Boonstra H, Barot S, Lusti-Narasimhan M. Making the Case for Multipurpose Prevention Technologies: the Socio-epidemiological Rationale. BJOG. 2014;121:23–6. 10.1111/1471-0528.12851 PubMed PMID: WOS:000344399300007.
    1. Woodsong C, Holt JDS. Acceptability and Preferences for Vaginal Dosage Forms Intended for Prevention of HIV or HIV and Pregnancy. Adv Drug Deliv Rev. 2015;92:146–54. 10.1016/j.addr.2015.02.004 PubMed PMID: WOS:000364269500011.
    1. Friend DR. An Update on Multipurpose Prevention Technologies for the Prevention of HIV Transmission and Pregnancy. Expert Opin Drug Deliv. 2016;13(4):533–45. PubMed PMID: WOS:000372075500004. 10.1517/17425247.2016.1134485
    1. Quaife M, Terris-Prestholt F, Vickerman P. The Promise of Multipurpose Pregnancy, STI, and HIV Prevention. Lancet Infect Dis. 2017;17(1):21–2. PubMed PMID: WOS:000389864700021.
    1. Baeten J, Heffron R, Kidoguchi L, Mugo N, Katabira E, Bukusi E, et al., editors. Near Elimination of HIV Transmission in a Demonstration Project of PrEP and ART. 2015 Conference on Retroviruses and Opportunistic Infections (CROI); 2015 Feb. 23–26, 2015; Seattle, WA: CROI, Alexandria, VA.
    1. Amico KR, Wallace M, Bekker LG, Roux S, Atujuna M, Sebastian E, et al. Experiences with HPTN 067/ADAPT Study Provided Open-Label PrEP Among Women in Cape Town: Facilitators and Barriers Within a Mutuality Framework. AIDS Behav. 2017;5:1361–75.
    1. Molina J-M, Charreau I, Spire B, Cotte L, Pialoux, Capitant C, et al., editors. On Demand PrEP With Oral TDF-FTC in the Open-Label Phase of the ANRS IPERGAY Trial. 2016 Conference on Retroviruses and Opportunistic Infections (CROI); 2016 Feb. 22–25, 2016; Boston, MA: CROI, Alexandria, VA.

Source: PubMed

3
Tilaa