1
|
Young IC, Srinivasan P, Shrivastava R, Janusziewicz R, Thorson A, Cottrell ML, Sellers RS, Sykes C, Schauer A, Little D, Kelley K, Kashuba ADM, Katz D, Pyles RB, García-Lerma JG, Vincent KL, Smith J, Benhabbour SR. Next generation 3D-printed intravaginal ring for prevention of HIV and unintended pregnancy. Biomaterials 2023; 301:122260. [PMID: 37549505 PMCID: PMC11537264 DOI: 10.1016/j.biomaterials.2023.122260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/22/2023] [Accepted: 07/26/2023] [Indexed: 08/09/2023]
Abstract
Globally, there are 20 million adolescent girls and young women living with HIV who have limited access to long-acting, effective, women-controlled preventative methods. Additionally, although there are many contraceptive methods available, globally, half of all pregnancies remain unintended. Here we report the first 3D-printed multipurpose prevention technology (MPT) intravaginal ring (IVR) for HIV prevention and contraception. We utilized continuous liquid interface production (CLIP™) to fabricate MPT IVRs in a biocompatible silicone-based resin. Etonogestrel (ENG), ethinyl estradiol (EE), and islatravir (ISL) were loaded into the silicone poly(urethane) IVR in a controlled single step drug loading process driven by absorption. ENG/EE/ISL IVR promoted sustained release of drugs for 150 days in vitro and 14 days in sheep. There were no adverse MPT IVR-related findings of cervicovaginal toxicity or changes in vaginal biopsies or microbiome community profiles evaluated in sheep. Furthermore, ISL IVR in macaques promoted sustained release for 28 days with ISL-triphosphate levels above the established pharmacokinetic benchmark of 50-100 fmol/106 PBMCs. The ISL IVR was found to be safe and well tolerated in the macaques with no observed mucosal cytokine changes or alterations in peripheral CD4 T-cell populations. Collectively, the proposed MPT IVR has potential to expand preventative choices for young women and girls.
Collapse
Affiliation(s)
- Isabella C Young
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Priya Srinivasan
- Laboratory Branch, Division of HIV Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, 30329, USA
| | - Roopali Shrivastava
- Joint Department of Biomedical Engineering, North Carolina State University and the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Rima Janusziewicz
- Joint Department of Biomedical Engineering, North Carolina State University and the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Allison Thorson
- Joint Department of Biomedical Engineering, North Carolina State University and the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Mackenzie L Cottrell
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Rani S Sellers
- Pathology Services Core, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Craig Sykes
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Amanda Schauer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Dawn Little
- Katmai Government Services, Anchorage, AK, 99515, USA
| | | | - Angela D M Kashuba
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - David Katz
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Richard B Pyles
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - J Gerardo García-Lerma
- Laboratory Branch, Division of HIV Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, 30329, USA
| | - Kathleen L Vincent
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - James Smith
- Laboratory Branch, Division of HIV Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, 30329, USA
| | - S Rahima Benhabbour
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Joint Department of Biomedical Engineering, North Carolina State University and the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
2
|
Lantz AM, Nicol MR. Translational Models to Predict Target Concentrations for Pre-Exposure Prophylaxis in Women. AIDS Res Hum Retroviruses 2022; 38:909-923. [PMID: 36097755 PMCID: PMC9805887 DOI: 10.1089/aid.2022.0057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The HIV epidemic remains a significant public health burden. Women represent half of the global HIV epidemic, yet there is an urgent need for a variety of prevention options to meet the needs of more women. Pre-exposure prophylaxis (PrEP) is a valuable prevention tool that uses antiretrovirals before a potential HIV exposure to prevent virus transmission. Development of effective preventive drug regimens for women is dependent on convenient dosing schedules and routes of administration, and on identifying defined target concentrations in mucosal tissues that provide complete protection against HIV transmission. There is a critical need for a translational model that can accurately predict in vivo target concentrations that are completely protective against HIV infection. There is no gold-standard preclinical model to predict PrEP efficacy. In this study, we review the strengths and limitations of three different preclinical models and their utility in predicting target concentrations in the female genital tract: humanized mice, non-human primates, and the ex vivo tissue model.
Collapse
Affiliation(s)
- Alyssa M. Lantz
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Melanie R. Nicol
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
3
|
Bick AJ, Avenant C, Tomasicchio M, van der Spuy Z, Hapgood JP. Increased HIV-1 infection in PBMCs treated in vitro with menstrual cycle phase hormones or medroxyprogesterone acetate likely occurs via different mechanisms. Am J Reprod Immunol 2022; 88:e13643. [PMID: 36302121 PMCID: PMC9884997 DOI: 10.1111/aji.13643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 09/15/2022] [Accepted: 10/18/2022] [Indexed: 02/01/2023] Open
Abstract
PROBLEM Both luteal phase progesterone (P4) levels and use of the intramuscular (IM) injectable progestin-only contraceptive depo-medroxyprogesterone acetate (DMPA-IM) have been linked to increased S/HIV acquisition in animal, clinical and in vitro models. Several plausible mechanisms could explain MPA-induced HIV-1 acquisition while those for the luteal phase are underexplored. METHOD OF STUDY Peripheral blood mononuclear cells (PBMCs) were treated with P4 and estrogen at concentrations mimicking the luteal phase, follicular phase or with levels of MPA mimicking peak serum levels in DMPA-IM users. Cells were infected with an R5-tropic infectious molecular clone and HIV-1 infection was measured. A role for the glucocorticoid receptor (GR) was investigated using the GR/PR antagonist RU486. CCR5 protein levels and activation status, assessed by levels of the activation marker CD69, were measured by flow cytometry after treatment in vitro and in PBMCs from naturally-cycling women or DMPA-IM users. RESULTS Both MPA and luteal phase hormones significantly increased HIV-1 infection in vitro. However, MPA but not luteal phase hormones increased the CD4+/CD8+ T cell ratio, CCR5 protein expression on CD4+ T cells and increased expression of the activation marker CD69. The GR is involved in MPA-induced, but not luteal phase hormone-induced increased HIV-1 infection. In DMPA-IM users, the frequency of CCR5-expressing CD3+ and CD8+ cells was higher than for women in the luteal phase. CONCLUSIONS MPA increases HIV-1 infection in a manner different from that of luteal phase hormones, most likely involving the GR and at least in part changes in the frequency and/or expression of CCR5 and CD69.
Collapse
Affiliation(s)
- Alexis J. Bick
- Department of Molecular and Cell Biology, University of Cape, Cape Town, South Africa
| | - Chanel Avenant
- Department of Molecular and Cell Biology, University of Cape, Cape Town, South Africa
| | - Michele Tomasicchio
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, South Africa.,South African MRC Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa
| | - Zephne van der Spuy
- Department of Obstetrics and Gynaecology, University of Cape Town, Groote Schuur Hospital, Cape Town, South Africa
| | - Janet P. Hapgood
- Department of Molecular and Cell Biology, University of Cape, Cape Town, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town South Africa.,Corresponding author:
| |
Collapse
|
4
|
Dedeloudi A, Siamidi A, Pavlou P, Vlachou M. Recent Advances in the Excipients Used in Modified Release Vaginal Formulations. MATERIALS 2022; 15:ma15010327. [PMID: 35009472 PMCID: PMC8745980 DOI: 10.3390/ma15010327] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 12/10/2022]
Abstract
The formulation of an ideal vaginal drug delivery system (DDS), with the requisite properties, with respect to safety, efficacy, patient compliance, aesthetics, harmonization with the regulatory requirements, and cost, requires a meticulous selection of the active ingredients and the excipients used. Novel excipients defined by diversity and multifunctionality are used in order to ameliorate drug delivery attributes. Synthetic and natural polymers are broadly used in pharmaceutical vaginal formulations (solid, semi-solid dosage forms, implantable devices, and nanomedicines) with a promising perspective in improving stability and compatibility issues when administered topically or systemically. Moreover, the use of biopolymers is aiming towards formulating novel bioactive, biocompatible, and biodegradable DDSs with a controllable drug release rate. Overviewing vaginal microenvironment, which is described by variable and perplexed features, a perceptive choice of excipients is essential. This review summarizes the recent advances on the excipients used in modified vaginal drug delivery formulations, in an attempt to aid the formulation scientist in selecting the optimal excipients for the preparation of vaginal products.
Collapse
Affiliation(s)
- Aikaterini Dedeloudi
- Department of Pharmacy, Division of Pharmaceutical Technology, School of Health Sciences, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.D.); (A.S.)
| | - Angeliki Siamidi
- Department of Pharmacy, Division of Pharmaceutical Technology, School of Health Sciences, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.D.); (A.S.)
| | - Panagoula Pavlou
- Laboratory of Chemistry-Biochemistry-Cosmetic Science, Department of Biomedical Sciences, University of West Attica, 28 Ag. Spyridonos Str., 12243 Egaleo, Greece;
| | - Marilena Vlachou
- Department of Pharmacy, Division of Pharmaceutical Technology, School of Health Sciences, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.D.); (A.S.)
- Correspondence: ; Tel.: +30-2107274674
| |
Collapse
|
5
|
Keller MJ, Wood L, Billingsley JM, Ray LL, Goymer J, Sinclair S, McGinn AP, Marzinke MA, Frank B, Srinivasan S, Liu C, Atrio JM, Espinoza L, Mugo N, Spiegel HML, Anderson PL, Fredricks DN, Hendrix CW, Marrazzo J, Bosinger SE, Herold BC. Tenofovir disoproxil fumarate intravaginal ring for HIV pre-exposure prophylaxis in sexually active women: a phase 1, single-blind, randomised, controlled trial. Lancet HIV 2019; 6:e498-e508. [PMID: 31320290 PMCID: PMC6719300 DOI: 10.1016/s2352-3018(19)30145-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 03/28/2019] [Accepted: 04/18/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND An intravaginal ring that releases the tenofovir prodrug, tenofovir disoproxil fumarate, provided 100% protection in macaques against simian HIV and was safe in a 14-day clinical trial in sexually abstinent women. We aimed to assess the safety and pharmacokinetics of this intravaginal ring over 90 days in sexually active women. METHODS We did a phase 1, single-blind, randomised, placebo-controlled trial to assess safety, pharmacokinetics, and acceptability of a tenofovir disoproxil fumarate intravaginal ring used continuously with monthly ring changes for 3 months. Sexually active women who were HIV negative were randomly assigned (3:1) to a tenofovir disoproxil fumarate ring or placebo ring. Primary safety endpoint was the proportion of women who had grade 2 or higher genitourinary adverse events judged related to study product and any grade 2 or higher adverse event as defined by the Division of AIDS Table for Grading the Severity of Adult and Pediatric Adverse Events. We quantified tenofovir disoproxil fumarate and tenofovir concentrations in cervicovaginal fluid, tenofovir in plasma, and tenofovir diphosphate, the active metabolite, in cervical tissue and dried blood spots 1 month after each ring insertion. We compared changes over time in cervicovaginal fluid cytokine and chemokine concentrations and vaginal microbiota. The study was electively stopped early and is registered with ClinicalTrials.gov, number NCT02762617. FINDINGS Between Feb 24 and July 20, 2017, 17 women were enrolled before study termination. 12 were assigned to receive the tenofovir disoproxil fumarate ring and five were assigned to receive the placebo ring. Two participants in the tenofovir disoproxil fumarate ring group completed 3 months of continuous ring use; eight were asked to discontinue ring use early because of ulcerations (grade 1) near the ring; in the remaining two women, rings were electively removed by study staff on day 20 and day 23. Ulcers were detected a mean of 32 days after ring use (range 23-56). Four of eight participants with ulcers were symptomatic with vaginal discharge; four had ulcers identified when examined; three had two ulcers; all ulcers resolved after ring removal. No participants in the placebo group developed ulcers. No grade 2 product-related adverse events were reported in either group and four non-product-related grade 2 adverse events were reported in the tenofovir disoproxil fumarate ring group. Cervicovaginal fluid tenofovir concentrations did not differ at day 14 (p=0·14) comparing the eight patients who did (median 1·0 × 105 ng/mL [IQR 9·1 × 104-1·1 × 105]) with the four who did not (6·0 × 104 ng/mL [5·6 × 104-1·1 × 105]) develop ulcers. No significant changes in vaginal microbiota were detected in either group. Concentrations of multiple inflammatory cytokines and chemokines were significantly higher at days 14 and 28 compared with baseline in the tenofovir disoproxil fumarate ring group but not the placebo group. INTERPRETATION Future studies are needed to establish whether the unanticipated finding of ulcerations is specific to this tenofovir disoproxil fumarate ring or generalisable to other sustained topical release formulations of tenofovir or its prodrugs. FUNDING National Institutes of Health.
Collapse
Affiliation(s)
- Marla J Keller
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Lianna Wood
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Laurie L Ray
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jessica Goymer
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Shada Sinclair
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aileen P McGinn
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mark A Marzinke
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Sujatha Srinivasan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Congzhou Liu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jessica M Atrio
- Department of Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Lilia Espinoza
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Nelly Mugo
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Hans M L Spiegel
- Division of AIDS, Kelly Government Solutions, Contractor to National Institute of Allergy and Infectious Diseases, National Institutes of Health, Department of Health and Human Services, Rockville, MD, USA
| | - Peter L Anderson
- Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO, USA
| | - David N Fredricks
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Craig W Hendrix
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeanne Marrazzo
- Department of Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | | | - Betsy C Herold
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
6
|
Lowinger MB, Barrett SE, Zhang F, Williams RO. Sustained Release Drug Delivery Applications of Polyurethanes. Pharmaceutics 2018; 10:E55. [PMID: 29747409 PMCID: PMC6027189 DOI: 10.3390/pharmaceutics10020055] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/02/2018] [Accepted: 05/04/2018] [Indexed: 12/18/2022] Open
Abstract
Since their introduction over 50 years ago, polyurethanes have been applied to nearly every industry. This review describes applications of polyurethanes to the development of modified release drug delivery. Although drug delivery research leveraging polyurethanes has been ongoing for decades, there has been renewed and substantial interest in the field in recent years. The chemistry of polyurethanes and the mechanisms of drug release from sustained release dosage forms are briefly reviewed. Studies to assess the impact of intrinsic drug properties on release from polyurethane-based formulations are considered. The impact of hydrophilic water swelling polyurethanes on drug diffusivity and release rate is discussed. The role of pore formers in modulating drug release rate is examined. Finally, the value of assessing mechanical properties of the dosage form and approaches taken in the literature are described.
Collapse
Affiliation(s)
- Michael B Lowinger
- College of Pharmacy, The University of Texas at Austin, 2409 University Avenue, Austin, TX 78712, USA.
- MRL, Merck & Co., Inc., 126 E. Lincoln Ave, Rahway, NJ 07065, USA.
| | | | - Feng Zhang
- College of Pharmacy, The University of Texas at Austin, 2409 University Avenue, Austin, TX 78712, USA.
| | - Robert O Williams
- College of Pharmacy, The University of Texas at Austin, 2409 University Avenue, Austin, TX 78712, USA.
| |
Collapse
|
7
|
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
|
8
|
Derby N, Aravantinou M, Kenney J, Ugaonkar SR, Wesenberg A, Wilk J, Kizima L, Rodriguez A, Zhang S, Mizenina O, Levendosky K, Cooney ML, Seidor S, Gettie A, Grasperge B, Blanchard J, Piatak M, Lifson JD, Fernández-Romero J, Zydowsky TM, Robbiani M. An intravaginal ring that releases three antiviral agents and a contraceptive blocks SHIV-RT infection, reduces HSV-2 shedding, and suppresses hormonal cycling in rhesus macaques. Drug Deliv Transl Res 2017; 7:840-858. [PMID: 28600625 PMCID: PMC5656733 DOI: 10.1007/s13346-017-0389-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Women globally need access to multipurpose prevention technologies (MPTs) that prevent human immunodeficiency virus (HIV), sexually transmitted infections that increase HIV acquisition/transmission risk, and unintended pregnancy. Seeking an MPT with activity against HIV, herpes simplex virus-2 (HSV-2), and human papillomavirus (HPV), we developed a prototype intravaginal ring (IVR), the MZCL IVR, which released the antiviral agents MIV-150, zinc acetate, and carrageenan (MZC for short) and the contraceptive levonorgestrel (LNG). Previously, we showed that an MZC gel has potent activity against immunodeficiency viruses, HSV-2, and HPV and that the MZCL (MZC with LNG) IVR releases all four components in macaques in vivo at levels associated with efficacy. Vaginal fluid from treated macaques has in vitro activity against HIV, HSV-2, and HPV. Herein, we assessed the ability of the MZCL IVR to protect macaques against repeated co-challenge with HSV-2 and SHIV-RT (simian immunodeficiency virus [SIV] containing the reverse transcriptase gene from HIV) and prevent hormonal cycling. We evaluated in vivo drug release in co-challenged macaques by measuring drug levels in blood and vaginal fluid and residual drug levels in used IVRs. The MZCL IVR significantly prevented SHIV-RT infection, reduced HSV-2 vaginal shedding, and prevented cycling. No non-nucleoside HIV reverse transcriptase inhibitor (NNRTI)-resistant SHIV was detected in macaques that became infected after continuous exposure to MZC from the IVR. Macaques wearing the MZCL IVR also had carrageenan levels in vaginal fluid expected to protect from HPV (extrapolated from mice) and LNG levels in blood associated with contraceptive efficacy. The MZCL IVR is a promising MPT candidate that warrants further development.
Collapse
MESH Headings
- Alphapapillomavirus/drug effects
- Alphapapillomavirus/physiology
- Animals
- Antiviral Agents/administration & dosage
- Antiviral Agents/pharmacology
- Carrageenan/administration & dosage
- Carrageenan/pharmacology
- Contraceptive Agents, Female/administration & dosage
- Contraceptive Agents, Female/pharmacology
- Contraceptive Devices, Female
- Disease Models, Animal
- Drug Therapy, Combination/methods
- Female
- Herpes Simplex/prevention & control
- Herpesvirus 2, Human/drug effects
- Herpesvirus 2, Human/physiology
- Humans
- Macaca mulatta
- Menstrual Cycle
- Pyridines/administration & dosage
- Pyridines/pharmacology
- Simian Acquired Immunodeficiency Syndrome/prevention & control
- Urea/administration & dosage
- Urea/analogs & derivatives
- Urea/pharmacology
- Vaginal Creams, Foams, and Jellies/administration & dosage
- Vaginal Creams, Foams, and Jellies/pharmacology
- Virus Shedding/drug effects
- Zinc Acetate/administration & dosage
- Zinc Acetate/pharmacology
Collapse
Affiliation(s)
- Nina Derby
- Population Council, 1230 York Avenue, New York, NY, 10065, USA.
| | | | - Jessica Kenney
- Population Council, 1230 York Avenue, New York, NY, 10065, USA
| | | | - Asa Wesenberg
- Population Council, 1230 York Avenue, New York, NY, 10065, USA
| | - Jolanta Wilk
- Population Council, 1230 York Avenue, New York, NY, 10065, USA
| | - Larisa Kizima
- Population Council, 1230 York Avenue, New York, NY, 10065, USA
| | - Aixa Rodriguez
- Population Council, 1230 York Avenue, New York, NY, 10065, USA
| | - Shimin Zhang
- Population Council, 1230 York Avenue, New York, NY, 10065, USA
| | - Olga Mizenina
- Population Council, 1230 York Avenue, New York, NY, 10065, USA
| | | | | | - Samantha Seidor
- Population Council, 1230 York Avenue, New York, NY, 10065, USA
| | - Agegnehu Gettie
- Aaron Diamond AIDS Research Center, 455 First Avenue, 7th Floor, New York, NY, 10016, USA
| | - Brooke Grasperge
- Tulane Primate Research Center, 18703 Three Rivers Road, Covington, LA, 70433-8915, USA
| | - James Blanchard
- Tulane Primate Research Center, 18703 Three Rivers Road, Covington, LA, 70433-8915, USA
| | - Michael Piatak
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702-1201, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702-1201, USA
| | - José Fernández-Romero
- Population Council, 1230 York Avenue, New York, NY, 10065, USA
- Science Department, Borough of Manhattan Community College, The City University of New York, 199 Chambers Street, New York, NY, 10007, USA
| | | | | |
Collapse
|
9
|
Pharmacokinetics of the Protein Microbicide 5P12-RANTES in Sheep following Single-Dose Vaginal Gel Administration. Antimicrob Agents Chemother 2017; 61:AAC.00965-17. [PMID: 28784672 DOI: 10.1128/aac.00965-17] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/28/2017] [Indexed: 11/20/2022] Open
Abstract
5P12-RANTES, a chemokine analogue that potently blocks the HIV CCR5 coreceptor, is being developed as both a vaginal and rectal microbicide for prevention of sexual transmission of HIV. Here, we report the first pharmacokinetic data for 5P12-RANTES following single-dose vaginal gel administration in sheep. Aqueous gel formulations containing low (1.24-mg/ml), intermediate (6.18-mg/ml), and high (32.0-mg/ml; suspension-type gel) concentrations of 5P12-RANTES were assessed via rheology, syringeability, and in vitro release testing. Following vaginal gel administration to sheep, 5P12-RANTES concentrations were measured in vaginal fluid, vaginal tissue, and serum over a 96-h period. All gels showed non-Newtonian pseudoplastic behavior, with the high-concentration gels exhibiting a greater viscosity and cohesive structure than the intermediate- and low-concentration gels. In in vitro release testing, >90% 5P12-RANTES was released from the low- and intermediate-concentration gels after 72 h. For the high-concentration gel, ∼50% 5P12-RANTES was detected, attributed to protein denaturation during lyophilization and/or subsequent solvation of the protein within the gel matrix. In sheep, 5P12-RANTES concentrations in vaginal fluid, vaginal tissue, and serum increased in a dose-dependent manner. The highest concentrations were measured in vaginal fluid (105 to 107 ng/ml), followed by vaginal tissue (104 to 106 ng/ml). Both of these concentration ranges are several orders of magnitude above the reported half-maximal inhibitory concentrations. The lowest concentration was measured in serum (<102 ng/ml). The 5P12-RANTES pharmacokinetic data are similar to those reported previously for other candidate microbicides. These data, coupled with 5P12-RANTES's potency at picomolar concentrations, its strong barrier to resistance, and the full protection that it was observed to provide in a rhesus macaque vaginal challenge model, support the continued development of 5P12-RANTES as a microbicide.
Collapse
|
10
|
A Depot Medroxyprogesterone Acetate Dose That Models Human Use and Its Effect on Vaginal SHIV Acquisition Risk. J Acquir Immune Defic Syndr 2017; 72:363-71. [PMID: 27355414 DOI: 10.1097/qai.0000000000000975] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Hormonal contraception with depot medroxyprogesterone acetate (DMPA) may increase HIV acquisition risk, but observational human studies are inconclusive, and animal models can help investigate this risk. In this study, we test the impact of a low DMPA dose, designed to resemble human contraceptive use, on Simian-Human Immunodeficiency Virus (SHIV) acquisition risk in pigtail macaques (Macaca nemestrina). METHODS Macaques metabolize DMPA faster than humans. We previously identified a per-weight DMPA dose and administration frequency that achieves long-lasting suppression of ovulation in macaques. Eight macaques were given 1.5-mg/kg DMPA monthly, whereas 11 were untreated controls. For comparison, women receive 150 mg (approximately 2 mg/kg) every 3 months. We exposed monkeys to 20 suboptimal SHIV challenges, designed to slowly infect half of controls and allow increased infection in the DMPA group. RESULTS It took a median 5.5 viral challenges to infect DMPA-treated macaques and 9 challenges for controls (P = 0.27; exact conditional logistic regression). The exact odds ratio was 2.2 (CI: 0.6 to 8.3). Ovulation was suppressed, and the vaginal epithelium was thinned after DMPA treatment in all animals (mean, 30 and 219 mm in DMPA-treated and control macaques, respectively, P = 0.03, t test using the Satterthwaite degrees-of-freedom approximation). CONCLUSIONS SHIV infections in DMPA-treated macaques were 2.2 times those of controls, but this was not statistically significant. The result is remarkably similar to studies of human DMPA use, which have shown HIV risk increases of a similar magnitude and of variable significance. Taken together with previous studies of higher DMPA doses in macaques, the results suggest a dose-dependent effect of DMPA on Simian Immunodeficiency Virus (SIV) or SHIV acquisition.
Collapse
|
11
|
Srinivasan P, Zhang J, Dinh CT, Teller RS, McNicholl JM, Kiser PF, Herold BC, Smith JM. Repeated administration of high-dose depot medroxyprogesterone acetate does not alter SHIV SF162p3 viral kinetics and tenofovir pharmacokinetics when delivered via intravaginal rings. J Med Primatol 2017; 46:129-136. [PMID: 28748662 PMCID: PMC7192064 DOI: 10.1111/jmp.12299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2017] [Indexed: 12/24/2022]
Abstract
BACKGROUND Intravaginal rings (IVR) for HIV prevention will likely be used by women on depot medroxyprogesterone acetate (DMPA) hormonal contraception. We used pigtailed macaques to evaluate the effects of DMPA on tenofovir disoproxil fumarate (TDF) IVR pharmacokinetics and viral shedding. METHODS Mucosal tenofovir (TFV) levels were compared in SHIVSF162p3 -negative DMPA-treated (n=4) and normally cycling (n=6) macaques receiving TDF IVRs. Plasma viremia and vaginal shedding were determined in groups of SHIVSF162p3 -positive DMPA-treated (n=6) and normally cycling (n=5) macaques. RESULTS Similar median vaginal fluid TFV concentrations were observed in the DMPA-treated and cycling macaques over 4 weeks (1.2×105 and 1.1.×105 ng/mL, respectively). Median plasma viremia and vaginal shedding AUC of the DMPA-treated (2.73×107 and 8.15×104 copies/mL, respectively) and cycling macaques (3.98×107 and 1.47×103 copies/mL, respectively) were statistically similar. CONCLUSIONS DMPA does not affect TDF IVR pharmacokinetics or SHIV shedding.
Collapse
Affiliation(s)
| | | | | | - Ryan S. Teller
- Department of Biomedical Engineering, Northwestern University, Evanston, IL
| | | | - Patrick F. Kiser
- Department of Biomedical Engineering, Northwestern University, Evanston, IL
| | - Betsy C. Herold
- Department of Pediatrics and Microbiology-Immunology, Albert Einstein College of Medicine, Bronx, NY
| | | |
Collapse
|
12
|
A multiplex assay for detection of SHIV plasma and mucosal IgG and IgA. J Immunol Methods 2017; 450:34-40. [PMID: 28750871 DOI: 10.1016/j.jim.2017.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/13/2017] [Accepted: 07/24/2017] [Indexed: 11/24/2022]
Abstract
Evaluating antibody maturation provides valuable data to characterize immune responses to HIV infection and can provide insight into biomedical intervention efficacy. It is important to develop assays that evaluate antibody maturation in both plasma and mucosal compartments. The nonhuman primate model provides a controlled system to collect temporal data that are integral to assessing intervention strategies. We report the development of a novel multiplex assay, based on the Bio-Plex platform, to evaluate plasma and mucosal IgG and IgA avidity and maturation against simian/human immunodeficiency virus (SHIV) in this controlled system. Vaginal mucosa and plasma samples were collected from a prior study evaluating the efficacy of a tenofovir disoproxil fumarate (TDF) intravaginal ring (IVR) against SHIVSF162P3 challenge in female pigtailed macaques. For validation of the multiplex assay, specimens from six SHIV-infected placebo animals and one TDF breakthrough animal were evaluated. For SHIV and HIV envelope analytes, antibody levels and avidity in both compartments continued to mature post-infection. Maturation of IgG and IgA levels was similar in each compartment, however, mucosal antibody levels tended to be more variable. This SHIV assay elucidates IgG/IgA antibody kinetics in the plasma and vaginal mucosa and will be a valuable tool in vaccine and other biomedical intervention studies in the nonhuman primate model.
Collapse
|
13
|
Notario-Pérez F, Ruiz-Caro R, Veiga-Ochoa MD. Historical development of vaginal microbicides to prevent sexual transmission of HIV in women: from past failures to future hopes. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:1767-1787. [PMID: 28670111 PMCID: PMC5479294 DOI: 10.2147/dddt.s133170] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Infection with human immunodeficiency virus (HIV) remains a global public health concern and is particularly serious in low- and middle-income countries. Widespread sexual violence and poverty, among other factors, increase the risk of infection in women, while currently available prevention methods are outside the control of most. This has driven the study of vaginal microbicides to prevent sexual transmission of HIV from men to women in recent decades. The first microbicides evaluated were formulated as gels for daily use and contained different substances such as surfactants, acidifiers and monoclonal antibodies, which failed to demonstrate efficacy in clinical trials. A gel containing the reverse transcriptase inhibitor tenofovir showed protective efficacy in women. However, the lack of adherence by patients led to the search for dosage forms capable of releasing the active principle for longer periods, and hence to the emergence of the vaginal ring loaded with dapivirine, which requires a monthly application and is able to reduce the sexual transmission of HIV. The future of vaginal microbicides will feature the use of alternative dosage forms, nanosystems for drug release and probiotics, which have emerged as potential microbicides but are still in the early stages of development. Protecting women with vaginal microbicide formulations would, therefore, be a valuable tool for avoiding sexual transmission of HIV.
Collapse
Affiliation(s)
- Fernando Notario-Pérez
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidad Complutense de Madrid, Madrid, Spain
| | - Roberto Ruiz-Caro
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidad Complutense de Madrid, Madrid, Spain
| | - María-Dolores Veiga-Ochoa
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
14
|
Makarova N, Henning T, Taylor A, Dinh C, Lipscomb J, Aubert R, Hanson D, Phillips C, Papp J, Mitchell J, McNicholl J, Garcia-Lerma GJ, Heneine W, Kersh E, Dobard C. Topical tenofovir protects against vaginal simian HIV infection in macaques coinfected with Chlamydia trachomatis and Trichomonas vaginalis. AIDS 2017; 31:745-752. [PMID: 28060011 PMCID: PMC11513895 DOI: 10.1097/qad.0000000000001389] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Chlamydia trachomatis and Trichomonas vaginalis, two prevalent sexually transmitted infections, are known to increase HIV risk in women and could potentially diminish preexposure prophylaxis efficacy, particularly for topical interventions that rely on local protection. We investigated in macaques whether coinfection with Chlamydia trachomatis/Trichomonas vaginalis reduces protection by vaginal tenofovir (TFV) gel. METHODS Vaginal TFV gel dosing previously shown to provide 100 or 74% protection when applied either 30 min or 3 days before simian HIV(SHIV) challenge was assessed in pigtailed macaques coinfected with Chlamydia trachomatis/Trichomonas vaginalis and challenged twice weekly with SHIV162p3 for up to 10 weeks (two menstrual cycles). Three groups of six macaques received either placebo or 1% TFV gel 30 min or 3 days before each SHIV challenge. We additionally assessed TFV and TFV diphosphate concentrations in plasma and vaginal tissues in Chlamydia trachomatis/Trichomonas vaginalis coinfected (n = 4) and uninfected (n = 4) macaques. RESULTS Chlamydia trachomatis/Trichomonas vaginalis coinfections were maintained during the SHIV challenge period. All macaques that received placebo gel were SHIV infected after a median of seven challenges (one menstrual cycle). In contrast, no infections were observed in macaques treated with TFV gel 30 min before SHIV challenge (P < 0.001). Efficacy was reduced to 60% when TFV gel was applied 3 days before SHIV challenge (P = 0.07). Plasma TFV and TFV diphosphate concentrations in tissues and vaginal lymphocytes were significantly higher in Chlamydia trachomatis/Trichomonas vaginalis coinfected compared with Chlamydia trachomatis/Trichomonas vaginalis uninfected macaques. CONCLUSION Our findings in this model suggest that Chlamydia trachomatis/Trichomonas vaginalis coinfection may have little or no impact on the efficacy of highly effective topical TFV modalities and highlight a significant modulation of TFV pharmacokinetics.
Collapse
Affiliation(s)
- Natalia Makarova
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers of Disease Control and Prevention (CDC) Atlanta, Georgia
| | - Tara Henning
- Division of Sexually Transmitted Disease Prevention, Centers of Disease Control and Prevention (CDC) Atlanta, Georgia
| | | | - Chuong Dinh
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers of Disease Control and Prevention (CDC) Atlanta, Georgia
| | - Jonathan Lipscomb
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers of Disease Control and Prevention (CDC) Atlanta, Georgia
| | - Rachael Aubert
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers of Disease Control and Prevention (CDC) Atlanta, Georgia
| | - Debra Hanson
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers of Disease Control and Prevention (CDC) Atlanta, Georgia
| | - Christi Phillips
- Division of Sexually Transmitted Disease Prevention, Centers of Disease Control and Prevention (CDC) Atlanta, Georgia
| | - John Papp
- Division of Sexually Transmitted Disease Prevention, Centers of Disease Control and Prevention (CDC) Atlanta, Georgia
| | - James Mitchell
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers of Disease Control and Prevention (CDC) Atlanta, Georgia
| | - Janet McNicholl
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers of Disease Control and Prevention (CDC) Atlanta, Georgia
| | - Gerardo J. Garcia-Lerma
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers of Disease Control and Prevention (CDC) Atlanta, Georgia
| | - Walid Heneine
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers of Disease Control and Prevention (CDC) Atlanta, Georgia
| | - Ellen Kersh
- Division of Sexually Transmitted Disease Prevention, Centers of Disease Control and Prevention (CDC) Atlanta, Georgia
| | - Charles Dobard
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers of Disease Control and Prevention (CDC) Atlanta, Georgia
| |
Collapse
|
15
|
Notario-Pérez F, Martín-Illana A, Cazorla-Luna R, Ruiz-Caro R, Bedoya LM, Tamayo A, Rubio J, Veiga MD. Influence of Chitosan Swelling Behaviour on Controlled Release of Tenofovir from Mucoadhesive Vaginal Systems for Prevention of Sexual Transmission of HIV. Mar Drugs 2017; 15:md15020050. [PMID: 28230790 PMCID: PMC5334630 DOI: 10.3390/md15020050] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/13/2017] [Accepted: 02/16/2017] [Indexed: 12/27/2022] Open
Abstract
The main challenges facing efforts to prevent the transmission of human immunodeficiency virus (HIV) are the lack of access to sexual education services and sexual violence against young women and girls. Vaginal formulations for the prevention of sexually transmitted infections are currently gaining importance in drug development. Vaginal mucoadhesive tablets can be developed by including natural polymers that have good binding capacity with mucosal tissues, such as chitosan or guar gum, semisynthetic polymers such as hydroxypropylmethyl cellulose, or synthetic polymers such as Eudragit® RS. This paper assesses the potential of chitosan for the development of sustained-release vaginal tablets of Tenofovir and compares it with different polymers. The parameters assessed were the permanence time of the bioadhesion—determined ex vivo using bovine vaginal mucosa as substrate—the drug release profiles from the formulation to the medium (simulated vaginal fluid), and swelling profiles in the same medium. Chitosan can be said to allow the manufacture of tablets that remain adhered to the vaginal mucosa and release the drug in a sustained way, with low toxicity and moderate swelling that ensures the comfort of the patient and may be useful for the prevention of sexual transmission of HIV.
Collapse
Affiliation(s)
- Fernando Notario-Pérez
- Departamento Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Araceli Martín-Illana
- Departamento Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Raúl Cazorla-Luna
- Departamento Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Roberto Ruiz-Caro
- Departamento Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Luis-Miguel Bedoya
- Departamento Farmacología, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Aitana Tamayo
- Instituto de Cerámica y Vidrio, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain.
| | - Juan Rubio
- Instituto de Cerámica y Vidrio, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain.
| | - María-Dolores Veiga
- Departamento Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| |
Collapse
|
16
|
Carias AM, Allen SA, Fought AJ, Kotnik Halavaty K, Anderson MR, Jimenez ML, McRaven MD, Gioia CJ, Henning TR, Kersh EN, Smith JM, Pereira LE, Butler K, McNicholl SJM, Hendry RM, Kiser PF, Veazey RS, Hope TJ. Increases in Endogenous or Exogenous Progestins Promote Virus-Target Cell Interactions within the Non-human Primate Female Reproductive Tract. PLoS Pathog 2016; 12:e1005885. [PMID: 27658293 PMCID: PMC5033389 DOI: 10.1371/journal.ppat.1005885] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/22/2016] [Indexed: 12/22/2022] Open
Abstract
Currently, there are mounting data suggesting that HIV-1 acquisition in women can be affected by the use of certain hormonal contraceptives. However, in non-human primate models, endogenous or exogenous progestin-dominant states are shown to increase acquisition. To gain mechanistic insights into this increased acquisition, we studied how mucosal barrier function and CD4+ T-cell and CD68+ macrophage density and localization changed in the presence of natural progestins or after injection with high-dose DMPA. The presence of natural or injected progestins increased virus penetration of the columnar epithelium and the infiltration of susceptible cells into a thinned squamous epithelium of the vaginal vault, increasing the likelihood of potential virus interactions with target cells. These data suggest that increasing either endogenous or exogenous progestin can alter female reproductive tract barrier properties and provide plausible mechanisms for increased HIV-1 acquisition risk in the presence of increased progestin levels.
Collapse
Affiliation(s)
- Ann M. Carias
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Shannon A. Allen
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Angela J. Fought
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Katarina Kotnik Halavaty
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Meegan R. Anderson
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Maria L. Jimenez
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Michael D. McRaven
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Casey J. Gioia
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Tara R. Henning
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Ellen N. Kersh
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - James M. Smith
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Lara E. Pereira
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Katherine Butler
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - S. Janet M. McNicholl
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - R. Michael Hendry
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Patrick F. Kiser
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Ronald S. Veazey
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
| | - Thomas J. Hope
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| |
Collapse
|
17
|
Malcolm RK, Boyd PJ, McCoy CF, Murphy DJ. Microbicide vaginal rings: Technological challenges and clinical development. Adv Drug Deliv Rev 2016; 103:33-56. [PMID: 26829289 DOI: 10.1016/j.addr.2016.01.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 01/13/2016] [Accepted: 01/20/2016] [Indexed: 10/22/2022]
Abstract
Vaginal rings (VRs) are flexible, torus-shaped, polymeric devices designed to sustain delivery of pharmaceutical drugs to the vagina for clinical benefit. Following first report in a 1970 patent application, several steroid-releasing VR products have since been marketed for use in hormone replacement therapy and contraception. Since 2002, there has been growing interest in the use of VR technology for delivery of drugs that can reduce the risk of sexual acquisition of human immunodeficiency virus type 1 (HIV-1), the causative agent of acquired immunodeficiency syndrome (AIDS). Although no vaginally-administered product has yet been approved for HIV reduction/prevention, extensive research efforts are continuing and a number of VR devices offering sustained release of so-called 'HIV microbicide' compounds are currently being evaluated in late-stage clinical studies. This review article provides an overview of the published scientific literature within this important field of research, focusing primarily on articles published within peer-reviewed journal publications. Many important aspects of microbicide-releasing VR technology are discussed, with a particular emphasis on the technological, manufacturing and clinical challenges that have emerged in recent years.
Collapse
|
18
|
Safety and Pharmacokinetics of Quick-Dissolving Polymeric Vaginal Films Delivering the Antiretroviral IQP-0528 for Preexposure Prophylaxis. Antimicrob Agents Chemother 2016; 60:4140-50. [PMID: 27139475 DOI: 10.1128/aac.00082-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 04/24/2016] [Indexed: 12/22/2022] Open
Abstract
For human immunodeficiency virus (HIV) prevention, microbicides or drugs delivered as quick-dissolving films may be more acceptable to women than gels because of their compact size, minimal waste, lack of an applicator, and easier storage and transport. This has the potential to improve adherence to promising products for preexposure prophylaxis. Vaginal films containing IQP-0528, a nonnucleoside reverse transcriptase inhibitor, were evaluated for their pharmacokinetics in pigtailed macaques. Polymeric films (22 by 44 by 0.1 mm; providing 75% of a human dose) containing IQP-0528 (1.5%, wt/wt) with and without poly(lactic-co-glycolic acid) (PLGA) nanoparticle encapsulation were inserted vaginally into pigtailed macaques in a crossover study design (n = 6). With unencapsulated drug, the median (range) vaginal fluid concentrations of IQP-0528 were 160.97 (2.73 to 2,104), 181.79 (1.86 to 15,800), and 484.50 (8.26 to 4,045) μg/ml at 1, 4, and 24 h after film application, respectively. Median vaginal tissue IQP-0528 concentrations at 24 h were 3.10 (0.03 to 222.58) μg/g. The values were similar at locations proximal, medial, and distal to the cervix. The IQP-0528 nanoparticle-formulated films delivered IQP-0528 in vaginal tissue and secretions at levels similar to those obtained with the unencapsulated formulation. A single application of either formulation did not disturb the vaginal microflora or the pH (7.24 ± 0.84 [mean ± standard deviation]). The high mucosal IQP-0528 levels delivered by both vaginal film formulations were between 1 and 5 log higher than the in vitro 90% inhibitory concentration (IC90) of 0.146 μg/ml. The excellent coverage and high mucosal levels of IQP-0528, well above the IC90, suggest that the films may be protective and warrant further evaluation in a vaginal repeated low dose simian-human immunodeficiency virus (SHIV) transmission study in macaques and clinically in women.
Collapse
|
19
|
Samizadeh M, Zhang X, Gunaseelan S, Nelson AG, Palombo MS, Myers DR, Singh Y, Ganapathi U, Szekely Z, Sinko PJ. Colorectal delivery and retention of PEG-Amprenavir-Bac7 nanoconjugates--proof of concept for HIV mucosal pre-exposure prophylaxis. Drug Deliv Transl Res 2016; 6:1-16. [PMID: 26712122 DOI: 10.1007/s13346-015-0269-4] [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: 12/01/2022]
Abstract
Local delivery of anti-HIV drugs to the colorectal mucosa, a major site of HIV replication, and their retention within mucosal tissue would allow for a reduction in dose administered, reduced dosing frequency and minimal systemic exposure. The current report describes a mucosal pre-exposure prophylaxis (mPrEP) strategy that utilizes nanocarrier conjugates (NC) consisting of poly(ethylene glycol) (PEG), amprenavir (APV), and a cell-penetrating peptide (CPP; namely Bac7, a fragment derived from bactenecin 7). APV-PEG NCs with linear PEGs (2, 5, 10, and 30 kDa) exhibited reduced (52-21%) anti-HIV-1 protease (PR) activity as compared to free APV in an enzyme-based FRET assay. In MT-2 T cells, APV-PEG3.4 kDa-FITC (APF) anti-HIV-1 activity was significantly reduced (160-fold, IC50 = 8064 nM) due to poor cell uptake, whereas it was restored (IC50 = 78.29 nM) and similar to APV (IC50 = 50.29 nM) with the addition of Bac7 to the NC (i.e., APV-PEG3.4 kDa-Bac7, APB). Flow cytometry and confocal microscopy demonstrated Bac7-PEG3.4 kDa-FITC (BPF) uptake was two- and fourfold higher than APF in MT-2 T cells and Caco-2 intestinal epithelial cells, respectively. There was no detectable punctate fluorescence in either cell line suggesting that BPF directly enters the cytosol thus avoiding endosomal entrapment. After colorectal administration in mice, BPF mucosal concentrations were 21-fold higher than APF concentrations. BPF concentrations also remained constant for the 5 days of the study suggesting that (1) the NC's structural characteristics (i.e., the size of the PEG carrier and the presence of a CPP) significantly influenced tissue persistence, and (2) the NCs were probably lodged in the lamina propria since the average rodent colon mucosal cell turnover time is 2-3 days. These encouraging results suggest that Bac7 functionalized NCs delivered locally to the colorectal mucosa may form drug delivery depots that are capable of sustaining colorectal drug concentrations. Although the exact mechanisms for tissue persistence are unclear and will require further study, these results provide proof-of-concept feasibility for mPrEP.
Collapse
Affiliation(s)
- Mahta Samizadeh
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Xiaoping Zhang
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Simi Gunaseelan
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Antoinette G Nelson
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Matthew S Palombo
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Daniel R Myers
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Yashveer Singh
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Usha Ganapathi
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Zoltan Szekely
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Patrick J Sinko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA.
| |
Collapse
|
20
|
Srinivasan P, Moss JA, Gunawardana M, Churchman SA, Yang F, Dinh CT, Mitchell JM, Zhang J, Fanter R, Miller CS, Butkyavichene I, McNicholl JM, Smith TJ, Baum MM, Smith JM. Topical Delivery of Tenofovir Disoproxil Fumarate and Emtricitabine from Pod-Intravaginal Rings Protects Macaques from Multiple SHIV Exposures. PLoS One 2016; 11:e0157061. [PMID: 27275923 PMCID: PMC4898685 DOI: 10.1371/journal.pone.0157061] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/23/2016] [Indexed: 11/19/2022] Open
Abstract
Topical preexposure prophylaxis (PrEP) against HIV has been marginally successful in recent clinical trials with low adherence rates being a primary factor for failure. Controlled, sustained release of antiretroviral (ARV) drugs may help overcome these low adherence rates if the product is protective for extended periods of time. The oral combination of tenofovir disoproxil fumarate (TDF) and emtricitabine (FTC) is currently the only FDA-approved ARV drug for HIV PrEP. A novel pod-intravaginal ring (IVR) delivering TDF and FTC at independently controlled rates was evaluated for efficacy at preventing SHIV162p3 infection in a rigorous, repeat low-dose vaginal exposure model using normally cycling female pigtailed macaques. Six macaques received pod-IVRs containing TDF (65 mg) and FTC (68 mg) every two weeks, and weekly vaginal exposures to 50 TCID50 of SHIV162p3 began one week after the first pod-IVR insertion. All pod-IVR-treated macaques were fully protected throughout the study (P = 0.0002, Log-rank test), whereas all control animals became infected with a median of 4 exposures to infection. The topical, sustained release of TDF and FTC from the pod-IVR maintained protective drug levels in macaques over four months of virus exposures. This novel and versatile delivery system has the capacity to deliver and maintain protective levels of multiple drugs and the protection observed here warrants clinical evaluation of this pod-IVR design.
Collapse
Affiliation(s)
- Priya Srinivasan
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - John A. Moss
- 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
| | - Scott A. Churchman
- Auritec Pharmaceuticals, Inc., Pasadena, California, United States of America
| | - Flora Yang
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| | - Chuong T. Dinh
- Total Solutions, Inc., Atlanta, Georgia, United States of America
| | - James M. Mitchell
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jining Zhang
- Total Solutions, Inc., Atlanta, Georgia, United States of America
| | - Rob Fanter
- 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
| | - Irina Butkyavichene
- Auritec Pharmaceuticals, Inc., Pasadena, California, United States of America
| | - Janet M. McNicholl
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Thomas J. Smith
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
- Auritec Pharmaceuticals, Inc., Pasadena, California, United States of America
| | - Marc M. Baum
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| | - James M. Smith
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
21
|
A phase 1 randomized placebo-controlled safety and pharmacokinetic trial of a tenofovir disoproxil fumarate vaginal ring. AIDS 2016; 30:743-51. [PMID: 26605514 DOI: 10.1097/qad.0000000000000979] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Tenofovir disoproxil fumarate (TDF), a prodrug of tenofovir (TFV), may be ideal for topical HIV preexposure prophylaxis because it has higher tissue and cell permeability than TFV; is not adversely impacted by seminal proteins; and its active metabolite, TFV-diphosphate (TFV-DP), has a long intracellular half-life. We engineered a TDF eluting polyurethane reservoir intravaginal ring (IVR) to provide near constant mucosal antiretroviral concentrations. METHODS A first-in-human randomized placebo-controlled trial was conducted to assess the safety and pharmacokinetics of the TDF IVR in healthy, sexually abstinent women (15 TDF and 15 placebo). Drug concentrations were measured in cervicovaginal fluid (CVF) obtained by swab, cervical tissue, plasma, and dried blood spots (DBS) over 14 days of continuous ring use. RESULTS There were 43 total, 23 reproductive tract, and eight product-related grade 1 adverse events. Steady-state CVF TFV concentrations were achieved proximal (vagina, ectocervix) and distal (introitus) to the TDF IVR 1 day after ring insertion. Median tissue TFV-DP concentrations 14 days after TDF IVR placement were 120 fmol/mg (interquartile range 90, 550). CVF collected from the cervix 1 week and 2 weeks after TDF IVR insertion provided significant protection against ex-vivo HIV challenge. Eleven of 14 (78%) participants had detectable TFV-DP DBS concentrations 14 days after TDF IVR placement, suggesting that DBS may provide a surrogate marker of adherence in future clinical trials. CONCLUSION A TDF IVR is safe, well tolerated, and results in mucosal TFV concentrations that exceed those associated with HIV protection. The findings support further clinical evaluation of this TDF IVR.
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW In response to the need for strategies women can use to protect themselves from HIV infection, a new class of product commonly referred to as vaginal 'microbicides' has been under development for the past few decades. Several leading products currently in development contain antiviral agents delivered in a vaginal ring. RECENT FINDINGS Research published over the past year reports advances in identification and continued formulation of specific antiviral agents that have potential for delivery in vaginal rings, including drug combinations for HIV, other sexually transmitted infections and contraception. Most products are antiretroviral reverse transcriptase inhibitors. Advances in vaginal ring design have also been reported; some of these are designed to release specific antiviral agents, while other designs could be used for multiple drugs. This review focuses both on antiviral agents and vaginal ring designs. SUMMARY Over the past year, advances continued to be made in the development of vaginal rings to deliver antiviral agents for prevention of HIV. An array of antiviral agents and vaginal ring designs to deliver these products are at various stages in the product pipeline process. Results from the first efficacy trials of an antiretroviral-containing vaginal ring are expected soon and will inform the continued development of this important product class.
Collapse
|
23
|
Ariën KK, Venkatraj M, Michiels J, Joossens J, Vereecken K, Van der Veken P, Heeres J, De Winter H, Heyndrickx L, Augustyns K, Vanham G. Resistance and cross-resistance profile of the diaryltriazine NNRTI and candidate microbicide UAMC01398. J Antimicrob Chemother 2016; 71:1159-68. [PMID: 26850721 DOI: 10.1093/jac/dkv501] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 12/26/2015] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVES The resistance development, cross-resistance to other NNRTIs and the impact of resistance on viral replicative fitness were studied for the new and potent NNRTI UAMC01398. METHODS Resistance was selected by dose escalation and by single high-dose selection against a comprehensive panel of NNRTIs used as therapeutics and NNRTIs under investigation for pre-exposure prophylaxis of sexual HIV transmission. A panel of 27 site-directed mutants with single mutations or combinations of mutations involved in reverse transcriptase (RT) inhibitor-mediated resistance was developed and used to confirm resistance to UAMC01398. Cross-resistance to other NNRTIs was assessed, as well as susceptibility of UAMC01398-resistant HIV to diarylpyrimidine-resistant viruses. Finally, the impact of UAMC01398 resistance on HIV replicative fitness was studied. RESULTS We showed that UAMC01398 has potent activity against dapivirine-resistant HIV, that at least four mutations in the RT are required in concert for resistance and that the resistance profile is similar to rilpivirine, both genotypically and phenotypically. Resistance development to UAMC01398 is associated with a severe fitness cost. CONCLUSIONS These data, together with the enhanced safety profile and good solubility in aqueous gels, make UAMC01398 an excellent candidate for HIV topical prevention.
Collapse
Affiliation(s)
- Kevin K Ariën
- Department of Biomedical Sciences, Institute of Tropical Medicine, B-2000 Antwerp, Belgium
| | - Muthusamy Venkatraj
- Laboratory of Medicinal Chemistry, University of Antwerp, B-2000 Antwerp, Belgium
| | - Johan Michiels
- Department of Biomedical Sciences, Institute of Tropical Medicine, B-2000 Antwerp, Belgium
| | - Jurgen Joossens
- Laboratory of Medicinal Chemistry, University of Antwerp, B-2000 Antwerp, Belgium
| | - Katleen Vereecken
- Department of Biomedical Sciences, Institute of Tropical Medicine, B-2000 Antwerp, Belgium
| | - Pieter Van der Veken
- Laboratory of Medicinal Chemistry, University of Antwerp, B-2000 Antwerp, Belgium
| | - Jan Heeres
- Laboratory of Medicinal Chemistry, University of Antwerp, B-2000 Antwerp, Belgium
| | - Hans De Winter
- Laboratory of Medicinal Chemistry, University of Antwerp, B-2000 Antwerp, Belgium
| | - Leo Heyndrickx
- Department of Biomedical Sciences, Institute of Tropical Medicine, B-2000 Antwerp, Belgium
| | - Koen Augustyns
- Laboratory of Medicinal Chemistry, University of Antwerp, B-2000 Antwerp, Belgium
| | - Guido Vanham
- Department of Biomedical Sciences, Institute of Tropical Medicine, B-2000 Antwerp, Belgium Department of Biomedical Sciences, University of Antwerp, B-2000 Antwerp, Belgium
| |
Collapse
|
24
|
Differential Mechanisms of Tenofovir and Tenofovir Disoproxil Fumarate Cellular Transport and Implications for Topical Preexposure Prophylaxis. Antimicrob Agents Chemother 2015; 60:1667-75. [PMID: 26711762 DOI: 10.1128/aac.02793-15] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 12/16/2015] [Indexed: 12/14/2022] Open
Abstract
Intravaginal rings releasing tenofovir (TFV) or its prodrug, tenofovir disoproxil fumarate (TDF), are being evaluated for HIV and herpes simplex virus (HSV) prevention. The current studies were designed to determine the mechanisms of drug accumulation in human vaginal and immune cells. The exposure of vaginal epithelial or T cells to equimolar concentrations of radiolabeled TDF resulted in over 10-fold higher intracellular drug levels than exposure to TFV. Permeability studies demonstrated that TDF, but not TFV, entered cells by passive diffusion. TDF uptake was energy independent but its accumulation followed nonlinear kinetics, and excess unlabeled TDF inhibited radiolabeled TDF uptake in competition studies. The carboxylesterase inhibitor bis-nitrophenyl phosphate reduced TDF uptake, suggesting saturability of intracellular carboxylesterases. In contrast, although TFV uptake was energy dependent, no competition between unlabeled and radiolabeled TFV was observed, and the previously identified transporters, organic anion transporters (OATs) 1 and 3, were not expressed in human vaginal or T cells. The intracellular accumulation of TFV was reduced by the addition of endocytosis inhibitors, and this resulted in the loss of TFV antiviral activity. Kinetics of drug transport and metabolism were monitored by quantifying the parent drugs and their metabolites by high-performance liquid chromatography tandem mass spectrometry (HPLC-MS/MS). Results were consistent with the identified mechanisms of transport, and the exposure of vaginal epithelial cells to equimolar concentrations of TDF compared to TFV resulted in ∼40-fold higher levels of the active metabolite, tenofovir diphosphate. Together, these findings indicate that substantially lower concentrations of TDF than TFV are needed to protect cells from HIV and HSV-2.
Collapse
|
25
|
Herold BC, Chen BA, Salata RA, Marzinke MA, Kelly CW, Dezzutti CS, McGowan I, Galaska B, Levy L, Piper JM, Hillier S, Hendrix CW. Impact of Sex on the Pharmacokinetics and Pharmacodynamics of 1% Tenofovir Gel. Clin Infect Dis 2015; 62:375-382. [PMID: 26508513 DOI: 10.1093/cid/civ913] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 10/16/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Tenofovir (TFV) gel partially protected against human immunodeficiency virus (HIV) in one but not subsequent trials. The disappointing results were attributed largely to poor adherence. However, timing of gel application relative to sex may impact pharmacokinetics and contribute to outcomes. Thus, we conducted a single-dose pharmacokinetic study of TFV gel applied 1 or 24 hours before or 1 hour before and 1 hour after (BAT) sex and compared results with dosing without sex. METHODS Twenty-four couples were enrolled; cervicovaginal lavage (CVL) and tissue were collected 2 hours after sex with matching timed collections at no sex visits and assayed for drug concentrations and CVL anti-HIV activity. RESULTS Compared with dosing without sex, median TFV concentrations after sex decreased 72% and 78% (P < .001) in CVL, 75% and 71% (P < .001) in vaginal tissue, and 75% (P = .06) and 55% (P < .001) in cervical tissue with -1 hour and -24 hour dosing, respectively. Median concentration of TFV-diphosphate also decreased significantly in cervical tissue with -1 hour, dosing. BAT dosing resulted in drug levels at least as great as those in the absence of sex. Percent inhibition of HIV infection by post-coital CVL increased significantly from median (interquartile range) of 55% (54%) in the absence of gel to 99% (7%), 77% (57%), and 100% (0.4%) with -1 hour, -24 hour, or BAT dosing, respectively, and correlated significantly with drug concentration. CONCLUSIONS Timing of TFV gel application relative to sex significantly impacts drug levels. BAT dosing or sustained delivery may be optimal for preexposure prophylaxis.
Collapse
Affiliation(s)
- Betsy C Herold
- Departments of Pediatrics and Microbiology-Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Beatrice A Chen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, School of Medicine, Pennsylvania
| | - Robert A Salata
- Department of Medicine, Case Western Reserve, Cleveland, Ohio
| | - Mark A Marzinke
- Department of Medicine (Clinical Pharmacology), Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Clifton W Kelly
- Statistical Center for HIV/AIDS Research & Prevention, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Charlene S Dezzutti
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, School of Medicine, Pennsylvania
| | - Ian McGowan
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, School of Medicine, Pennsylvania
| | - Beth Galaska
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, School of Medicine, Pennsylvania
| | - Lisa Levy
- Family Health International 360, Durham, North Carolina
| | - Jeanna M Piper
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Sharon Hillier
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, School of Medicine, Pennsylvania
| | - Craig W Hendrix
- Department of Medicine (Clinical Pharmacology), Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|