1
|
Singata-Madliki M, Smit J, Beksinska M, Balakrishna Y, Avenant C, Beesham I, Seocharan I, Batting J, Hapgood JP, Hofmeyr GJ. Effects of injectable contraception with depot medroxyprogesterone acetate or norethisterone enanthate on estradiol levels and menstrual, psychological and behavioral measures relevant to HIV risk: The WHICH randomized trial. PLoS One 2024; 19:e0295764. [PMID: 38530848 DOI: 10.1371/journal.pone.0295764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 11/14/2023] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Observational data suggest lower HIV risk with norethisterone enanthate (NET-EN) than with depo-medroxyprogesterone acetate intramuscular (DMPA-IM) injectable contraceptives. If confirmed, a switch between these similar injectable methods would be programmatically feasible and could impact the trajectory of the HIV epidemic. We aimed in this paper to investigate the effects of DMPA-IM and NET-EN on estradiol levels, measures of depression and sexual activity and menstrual effects, relevant to HIV risk; and to ascertain whether these measures are associated with estradiol levels. METHODS This open-label trial conducted at two sites in South Africa from 5 November 2018 to 30 November 2019, randomized HIV-negative women aged 18-40 to DMPA-IM 150 mg intramuscular 12-weekly (n = 262) or NET-EN 200 mg intramuscular 8-weekly (n = 259). Data were collected on hormonal, behavioral and menstrual effects at baseline and at 25 weeks (25W). RESULTS At 25W, median 17β estradiol levels were substantially lower than at baseline (p<0.001) for both methods: 76.5 pmol/L (interquartile range (IQR) 54.1 to 104.2) in the DMPA-IM group (n = 222), and 69.8 pmol/L (IQR: 55.1 to 89.3) in the NET-EN group (n = 225), with no statistical difference between the two methods (p = 0.450). Compared with DMPA-IM, NET-EN users reported significantly less amenorrhoea, fewer sexual acts, fewer users reporting at least one act of unprotected sex, more condom use with steady partner, more days with urge for sexual intercourse, more days feeling partner does not love her, and more days feeling sad for no reason. We did not find a clear association between estradiol levels and sexual behavior, depression and menstrual effects. Behavioral outcomes suggest less sexual exposure with NET-EN than DMPA-IM. The strength of this evidence is high due to the randomized study design and the consistency of results across the outcomes measured. CONCLUSIONS Estradiol levels were reduced to postmenopausal levels by both methods. Secondary outcomes suggesting less sexual exposure with NET-EN are consistent with reported observational evidence of less HIV risk with NET-EN. A randomized trial powered for HIV acquisition is feasible and needed to answer this important question. TRIAL REGISTRATION PACTR 202009758229976.
Collapse
Affiliation(s)
- Mandisa Singata-Madliki
- Effective Care Research Unit, Eastern Cape Department of Health/Universities of the Witwatersrand and Fort Hare, East London, South Africa
| | - Jenni Smit
- Wits MRU (MatCH Research Unit), Department of Obstetrics and Gynecology, Faculty of Health Sciences, University of the Witwatersrand, Durban, South Africa
| | - Mags Beksinska
- Wits MRU (MatCH Research Unit), Department of Obstetrics and Gynecology, Faculty of Health Sciences, University of the Witwatersrand, Durban, South Africa
| | - Yusentha Balakrishna
- Biostatistics Research Unit, South African Medical Research Council, Durban, South Africa
| | - Chanel Avenant
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| | - Ivana Beesham
- Wits MRU (MatCH Research Unit), Department of Obstetrics and Gynecology, Faculty of Health Sciences, University of the Witwatersrand, Durban, South Africa
| | - Ishen Seocharan
- Biostatistics Research Unit, South African Medical Research Council, Durban, South Africa
| | - Joanne Batting
- Effective Care Research Unit, Eastern Cape Department of Health/Universities of the Witwatersrand and Fort Hare, East London, South Africa
| | - Janet P Hapgood
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - G Justus Hofmeyr
- Effective Care Research Unit, Eastern Cape Department of Health/Universities of the Witwatersrand and Fort Hare, East London, South Africa
- Walter Sisulu University, East London, South Africa
- Department of Obstetrics and Gynecology, University of Botswana, Gabarone, Botswana
| |
Collapse
|
2
|
Gupta PM, Balle C, Tharp GK, Nelson SA, Gasper MA, Brown B, Alisoltani A, Onono M, Palanee-Phillips T, Nair G, Ayele H, Noel-Romas L, Passmore JAS, Burgener AD, Heffron R, Jaspan HB, Bosinger SE. Systems analysis reveals differential expression of endocervical genes in African women randomized to DMPA-IM, LNG implant or cu-IUD. Clin Immunol 2023; 255:109750. [PMID: 37660744 PMCID: PMC10570927 DOI: 10.1016/j.clim.2023.109750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 08/22/2023] [Indexed: 09/05/2023]
Abstract
Although effective contraceptives are crucial for preventing unintended pregnancies, evidence suggests that their use may perturb the female genital tract (FGT). A comparative analysis of the effects of the most common contraceptives on the FGT have not been evaluated in a randomized clinical trial setting. Here, we evaluated the effect of three long-acting contraceptive methods: depot medroxyprogesterone acetate(DMPA-IM), levonorgestrel(LNG) implant, and a copper intrauterine device (Cu-IUD), on the endocervical host transcriptome in 188 women from the Evidence for Contraceptive Options and HIV Outcomes Trial (ECHO) trial. Cu-IUD usage showed the most extensive transcriptomic changes, and was associated with inflammatory and anti-viral host responses. DMPA-IM usage was enriched for pathways associated with T cell responses. LNG implant had the mildest effect on endocervical gene expression, and was associated with growth factor signaling. These data provide a mechanistic basis for the diverse influence that varying contraceptives have on the FGT.
Collapse
Affiliation(s)
- Prachi Mehrotra Gupta
- Emory National Primate Research Center (ENPRC) Genomics Core Laboratory, Division of Microbiology & Immunology, Emory University, Atlanta, GA, USA
| | - Christina Balle
- Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
| | - Gregory K Tharp
- Emory National Primate Research Center (ENPRC) Genomics Core Laboratory, Division of Microbiology & Immunology, Emory University, Atlanta, GA, USA
| | - Sydney A Nelson
- Emory National Primate Research Center (ENPRC) Genomics Core Laboratory, Division of Microbiology & Immunology, Emory University, Atlanta, GA, USA
| | | | - Bryan Brown
- Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Arghavan Alisoltani
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Thesla Palanee-Phillips
- Wits RHI, University of the Witwatersrand, Faculty of Health Sciences, School of Public Health, Johannesburg, South Africa; Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | | | - Hosseana Ayele
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Laura Noel-Romas
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jo-Ann S Passmore
- Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa; National Health Laboratory Service, Cape Town, South Africa; CAPRISA DSI-NRF Centre of Excellence in HIV Prevention, University of Cape Town, South Africa
| | - Adam D Burgener
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Renee Heffron
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA
| | - Heather B Jaspan
- Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa; Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA
| | - Steven E Bosinger
- Emory University School of Medicine, Department of Pathology & Laboratory Medicine, GA, USA; Emory Vaccine Center, Emory University, GA, USA.
| |
Collapse
|
3
|
Bick AJ, Skosana SB, Avenant C, Hapgood JP. Variability and quantification of serum medroxyprogesterone acetate levels. Steroids 2022; 187:109100. [PMID: 35964796 PMCID: PMC9884996 DOI: 10.1016/j.steroids.2022.109100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 07/27/2022] [Accepted: 08/09/2022] [Indexed: 02/01/2023]
Abstract
Quantification of serum progestin levels in clinical contraceptive studies is now routinely performed to understand progestin pharmacokinetics and to correct for unreliable self-reporting of contraceptive use by study participants. Many such studies are focussed on the three-monthly progestin-only intramuscular (IM) injectable contraceptive depot medroxyprogesterone acetate (DMPA-IM). Methods commonly used to measure serum MPA levels include liquid chromatography coupled to mass spectrometry (LC/MS) and radioimmunoassay (RIA); however, RIA methods have not been used in recent years. We review the available literature and find that these methods vary widely in terms of use of organic solvent extraction, use of derivitization and choice of organic solvent and chromatography columns. There is a lack of standardization of LC/MS methodology, including a lack of detailed extraction protocols. Limited evidence suggests that RIA, without organic solvent extraction, likely over-estimates progestin levels. Maximum MPA concentrations in the first two weeks post-injection show wide inter-individual and inter-study variation, regardless of quantification method used. Standardization of quantification methods and sampling time post-injection is required to improve interpretation of clinical data, in particular the side effects arising at different times depending on the pharmacokinetic profile unique to injectable contraceptives.
Collapse
Affiliation(s)
- Alexis J Bick
- Department of Molecular and Cell Biology, University of Cape, Cape Town, South Africa
| | - Salndave B Skosana
- 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
| | - 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.
| |
Collapse
|
4
|
Balle C, Gupta PM, Tharp GK, Nelson SA, Konstantinus IN, Lennard K, Jaumdally SZ, Happel AU, Barnabas SL, Gill K, Bekker LG, Passmore JAS, Jaspan HB, Bosinger SE. Systems Analysis Reveals Contraceptive-Induced Alteration of Cervicovaginal Gene Expression in a Randomized Trial. FRONTIERS IN REPRODUCTIVE HEALTH 2022; 4:781687. [PMID: 36303659 PMCID: PMC9580795 DOI: 10.3389/frph.2022.781687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/31/2022] [Indexed: 12/15/2022] Open
Abstract
Hormonal contraceptives (HCs) are vital in managing the reproductive health of women. However, HC usage has been linked to perturbations in cervicovaginal immunity and increased risk of sexually transmitted infections. Here, we evaluated the impact of three HCs on the cervicovaginal environment using high-throughput transcriptomics. From 2015 to 2017, 130 adolescent females aged 15-19 years were enrolled into a substudy of UChoose, a single-site, open-label randomized, crossover trial (NCT02404038) and randomized to injectable norethisterone-enanthate (Net-En), combined oral contraceptives (COC), or etonorgesterol/ethinyl-estradiol-combined contraceptive vaginal ring (CCVR). Cervicovaginal samples were collected after 16 weeks of randomized HC use and analyzed by RNA-Seq, 16S rRNA gene sequencing, and Luminex analysis. Participants in the CCVR arm had a significant elevation of transcriptional networks driven by IL-6, IL-1, and NFKB, and lower expression of genes supporting epithelial barrier integrity. An integrated multivariate analysis demonstrated that networks of microbial dysbiosis and inflammation best discriminated the CCVR arm from the other contraceptive groups, while genes involved in epithelial cell differentiation were predictive of the Net-En and COC arms. Collectively, these data from a randomized trial represent the most comprehensive "omics" analyses of the cervicovaginal response to HCs and provide important mechanistic guidelines for the provision of HCs in sub-Saharan Africa.
Collapse
Affiliation(s)
- Christina Balle
- Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Prachi M. Gupta
- Yerkes Genomics Core Laboratory, Yerkes National Primate Research Center, Atlanta, GA, United States
| | - Gregory K. Tharp
- Yerkes Genomics Core Laboratory, Yerkes National Primate Research Center, Atlanta, GA, United States
| | - Sydney A. Nelson
- Yerkes Genomics Core Laboratory, Yerkes National Primate Research Center, Atlanta, GA, United States
| | - Iyaloo N. Konstantinus
- Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa,Namibia Institute of Pathology, Windhoek, Namibia
| | - Katie Lennard
- Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Shameem Z. Jaumdally
- Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Anna-Ursula Happel
- Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Shaun L. Barnabas
- Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa,Desmond Tutu Health Centre, University of Cape Town, Cape Town, South Africa,Family Clinical Research Center, Stellenbosch University, Tygerberg, South Africa
| | - Katherine Gill
- Desmond Tutu Health Centre, University of Cape Town, Cape Town, South Africa
| | - Linda-Gail Bekker
- Desmond Tutu Health Centre, University of Cape Town, Cape Town, South Africa
| | - Jo-Ann S. Passmore
- Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa,National Health Laboratory Service, Cape Town, South Africa
| | - Heather B. Jaspan
- Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa,Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States,Department of Pediatrics and Global Health, University of Washington, Seattle, WA, United States
| | - Steven E. Bosinger
- Yerkes Genomics Core Laboratory, Yerkes National Primate Research Center, Atlanta, GA, United States,Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States,Emory Vaccine Center, Emory University, Atlanta, GA, United States,*Correspondence: Steven E. Bosinger
| |
Collapse
|
5
|
VanBenschoten HM, Woodrow KA. Vaginal delivery of vaccines. Adv Drug Deliv Rev 2021; 178:113956. [PMID: 34481031 PMCID: PMC8722700 DOI: 10.1016/j.addr.2021.113956] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/06/2021] [Accepted: 08/28/2021] [Indexed: 11/22/2022]
Abstract
Recent estimates suggest that one in two sexually active individuals will acquire a sexually transmitted infection by age 25, an alarming statistic that amounts to over 1 million new infections per day worldwide. Vaccination against STIs is highly desirable for alleviating this global burden of disease. Vaginal immunization is a promising strategy to combat transmission via the vaginal mucosa. The vagina is typically considered a poor inductive site for common correlates of adaptive immunity. However, emerging evidence suggests that immune tolerance may be overcome by precisely engineered vaccination schemes that orchestrate cell-mediated immunity and establish tissue resident memory immune cells. In this review, we will discuss the unique immunological milieu of the vaginal mucosa and our current understanding of correlates of pathogenesis and protection for several common STIs. We then present a summary of recent vaginal vaccine studies and explore the role that mucosal adjuvants and delivery systems play in enhancing protection according to requisite features of immunity. Finally, we offer perspectives on the challenges and future directions of vaginal vaccine delivery, discussing remaining physiological barriers and innovative vaccine formulations that may overcome them.
Collapse
Affiliation(s)
- Hannah M VanBenschoten
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States
| | - Kim A Woodrow
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States.
| |
Collapse
|
6
|
Vincent KL, Frost PA, Motamedi M, Dick EJ, Wei J, Yang J, White R, Gauduin MC. High-Resolution Quantitative Mapping of Macaque Cervicovaginal Epithelial Thickness: Implications for Mucosal Vaccine Delivery. Front Immunol 2021; 12:660524. [PMID: 34262561 PMCID: PMC8273733 DOI: 10.3389/fimmu.2021.660524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/28/2021] [Indexed: 11/13/2022] Open
Abstract
Vaginal mucosal surfaces naturally offer some protection against sexually transmitted infections (STIs) including Human Immunodeficiency Virus-1, however topical preventative medications or vaccine designed to boost local immune responses can further enhance this protection. We previously developed a novel mucosal vaccine strategy using viral vectors integrated into mouse dermal epithelium to induce virus-specific humoral and cellular immune responses at the site of exposure. Since vaccine integration occurs at the site of cell replication (basal layer 100-400 micrometers below the surface), temporal epithelial thinning during vaccine application, confirmed with high resolution imaging, is desirable. In this study, strategies for vaginal mucosal thinning were evaluated noninvasively using optical coherence tomography (OCT) to map reproductive tract epithelial thickness (ET) in macaques to optimize basal layer access in preparation for future effective intravaginal mucosal vaccination studies. Twelve adolescent female rhesus macaques (5-7kg) were randomly assigned to interventions to induce vaginal mucosal thinning, including cytobrush mechanical abrasion, the chemical surfactant spermicide nonoxynol-9 (N9), the hormonal contraceptive depomedroxyprogesterone acetate (DMPA), or no intervention. Macaques were evaluated at baseline and after interventions using colposcopy, vaginal biopsies, and OCT imaging, which allowed for real-time in vivo visualization and measurement of ET of the mid-vagina, fornices, and cervix. P value ≤0.05 was considered significant. Colposcopy findings included pink, rugated tissue with variable degrees of white-tipped, thickened epithelium. Baseline ET of the fornices was thinner than the cervix and vagina (p<0.05), and mensing macaques had thinner ET at all sites (p<0.001). ET was decreased 1 month after DMPA (p<0.05) in all sites, immediately after mechanical abrasion (p<0.05) in the fornix and cervix, and after two doses of 4% N9 (1.25ml) applied over 14 hrs in the fornix only (p<0.001). Histological assessment of biopsied samples confirmed OCT findings. In summary, OCT imaging allowed for real time assessment of macaque vaginal ET. While varying degrees of thinning were observed after the interventions, limitations with each were noted. ET decreased naturally during menses, which may provide an ideal opportunity for accessing the targeted vaginal mucosal basal layers to achieve the optimum epithelial thickness for intravaginal mucosal vaccination.
Collapse
Affiliation(s)
- Kathleen L. Vincent
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Patrice A. Frost
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, United States
- Southwest National Primate Research Center, San Antonio, TX, United States
| | - Massoud Motamedi
- Department of Ophthalmology and Visual Sciences, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Edward J. Dick
- Southwest National Primate Research Center, San Antonio, TX, United States
- Disease Intervention and Prevention Program, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Jingna Wei
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Jinping Yang
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Robert White
- Disease Intervention and Prevention Program, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Marie-Claire Gauduin
- Southwest National Primate Research Center, San Antonio, TX, United States
- Disease Intervention and Prevention Program, Texas Biomedical Research Institute, San Antonio, TX, United States
| |
Collapse
|
7
|
Ayele H, Perner M, McKinnon LR, Birse K, Farr Zuend C, Burgener A. An updated review on the effects of depot medroxyprogesterone acetate on the mucosal biology of the female genital tract. Am J Reprod Immunol 2021; 86:e13455. [PMID: 33991137 PMCID: PMC8459266 DOI: 10.1111/aji.13455] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/28/2021] [Accepted: 05/10/2021] [Indexed: 11/29/2022] Open
Abstract
Background Access to safe, effective, and affordable contraception is important for women’s health and essential to mitigate maternal and fetal mortality rates. The progestin‐based contraceptive depot medroxyprogesterone acetate (DMPA) is a popular contraceptive choice with a low failure rate and convenient administration schedule. Aim In this review, we compiled observational data from human cohorts that examine how DMPA influences the mucosal biology of the female genital tract (FGT) that are essential in maintaining vaginal health, including resident immune cells, pro‐inflammatory cytokines, epithelial barrier function, and the vaginal microbiome Materials and Methods This review focused on the recent published literature published in 2019 and 2020. Results Recent longitudinal studies show that DMPA use associates with an immunosuppressive phenotype, increase in CD4+CCR5+ T cells, and alterations to growth factors. In agreement with previous meta‐analyses, DMPA use is associated with minimal effects of the composition of the vaginal microbiome. Cross‐sectional studies associate a more pro‐inflammatory relationship with DMPA, but these studies are confounded by inherent weaknesses of cross‐sectional studies, including differences in study group sizes, behaviors, and other variables that may affect genital inflammation. Discussion & Conclusion These recent results indicate that the interactions between DMPA and the vaginal mucosa are complex emphasizing the need for comprehensive longitudinal studies that take into consideration the measurement of multiple biological parameters.
Collapse
Affiliation(s)
- Hossaena Ayele
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada.,Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA
| | - Michelle Perner
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lyle R McKinnon
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Kenzie Birse
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Obstetrics & Gynecology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Christina Farr Zuend
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA
| | - Adam Burgener
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada.,Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Obstetrics & Gynecology, University of Manitoba, Winnipeg, Manitoba, Canada.,Unit of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Solna, Sweden
| |
Collapse
|
8
|
Böttinger P, Schreiber K, Hyjek E, Krausz T, Spiotto MT, Steiner M, Idel C, Booras H, Beck-Engeser G, Riederer J, Willimsky G, Wolf SP, Karrison T, Jensen E, Weichselbaum RR, Nakamura Y, Yew PY, Lambert PF, Kurita T, Kiyotani K, Leisegang M, Schreiber H. Cooperation of genes in HPV16 E6/E7-dependent cervicovaginal carcinogenesis trackable by endoscopy and independent of exogenous estrogens or carcinogens. Carcinogenesis 2021; 41:1605-1615. [PMID: 32221533 DOI: 10.1093/carcin/bgaa027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 03/16/2020] [Accepted: 03/26/2020] [Indexed: 02/07/2023] Open
Abstract
Human papillomavirus (HPV) infection is necessary but insufficient for progression of epithelial cells from dysplasia to carcinoma-in situ (CIS) to invasive cancer. The combination of mutant cellular and viral oncogenes that regulate progression of cervical cancer (CC) remains unclear. Using combinations of HPV16 E6/E7 (E+), mutant Kras (mKras) (K+) and/or loss of Pten (P-/-), we generated autochthonous models of CC without exogenous estrogen, carcinogen or promoters. Furthermore, intravaginal instillation of adenoCre virus enabled focal activation of the oncogenes/inactivation of the tumor suppressor gene. In P+/+ mice, E6/E7 alone (P+/+E+K-) failed to cause premalignant changes, while mKras alone (P+/+E-K+) caused persistent mucosal abnormalities in about one-third of mice, but no cancers. To develop cancer, P+/+ mice needed both E6/E7 and mKras expression. Longitudinal endoscopies of P+/+E+K+ mice predicted carcinoma development by detection of mucosal lesions, found on an average of 23 weeks prior to death, unlike longitudinal quantitative PCRs of vaginal lavage samples from the same mice. Endoscopy revealed that individual mice differed widely in the time required for mucosal lesions to appear after adenoCre and in the time required for these lesions to progress to cancer. These cancers developed in the transition zone that extends, unlike in women, from the murine cervix to the distal vagina. The P-/-E+K+ genotype led to precipitous cancer development within a few weeks and E6/E7-independent cancer development occurred in the P-/-E-K+ genotype. In the P-/-E+K- genotype, mice only developed CIS. Thus, distinct combinations of viral and cellular oncogenes are involved in distinct steps in cervical carcinogenesis.
Collapse
Affiliation(s)
- Paula Böttinger
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Karin Schreiber
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Elizabeth Hyjek
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
| | - Thomas Krausz
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
| | - Michael T Spiotto
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
| | - Madeline Steiner
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Christian Idel
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Heather Booras
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | | | - Jessie Riederer
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Gerald Willimsky
- Institute of Immunology, Charité-Universitätsmedizin Berlin, Campus Buch, Berlin, Germany.,German Cancer Research Center, Heidelberg, Germany.,German Cancer Consortium, Partner site Berlin, Berlin, Germany
| | - Steven P Wolf
- Department of Pathology, The University of Chicago, Chicago, IL, USA.,Institute of Immunology, Charité-Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| | - Theodore Karrison
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA
| | - Elizabeth Jensen
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
| | - Yusuke Nakamura
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Poh Yin Yew
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Paul F Lambert
- McArdle Laboratory for Cancer Research/Department of Oncology, University of Wisconsin, Madison, WI, USA
| | - Takeshi Kurita
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH, USA
| | - Kazuma Kiyotani
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Matthias Leisegang
- Institute of Immunology, Charité-Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| | - Hans Schreiber
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
9
|
Daly MB, Sterling M, Holder A, Dinh C, Nishiura K, Khalil G, García-Lerma JG, Dobard C. The effect of depot medroxyprogesterone acetate on tenofovir alafenamide in rhesus macaques. Antiviral Res 2020; 186:105001. [PMID: 33385420 PMCID: PMC8480307 DOI: 10.1016/j.antiviral.2020.105001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 12/17/2020] [Accepted: 12/23/2020] [Indexed: 11/28/2022]
Abstract
Prevention of HIV infection and unintended pregnancies are public health priorities. In sub-Saharan Africa, where HIV prevalence is highest, depot medroxyprogesterone acetate (DMPA) is widely used as contraception. Therefore, understanding potential interactions between DMPA and antiretrovirals is critical. Here, we use a macaque model to investigate the effect of DMPA on the pharmacology of the antiretroviral tenofovir alafenamide (TAF). Female rhesus macaques received 30 mg of DMPA (n = 9) or were untreated (n = 9). Macaques received a human equivalent dose of TAF (1.5 mg/kg) orally by gavage. Tenofovir (TFV) and TFV-diphosphate (TFV-DP) were measured in blood, secretions, and tissues over 72 h. The median area under the curve (AUC0-72h) values for TFV-DP in peripheral blood mononuclear cells were similar in DMPA-treated (6991 fmol*h/106 cells) and untreated controls (5256 fmol*h/106 cells) (P = 0.174). Rectal tissue TFV-DP concentrations from DMPA+ animals [median: 20.23 fmol/mg of tissue (range: 4.94-107.95)] were higher than the DMPA- group [median: below the limit of quantification (BLOQ-11.92)], (P = 0.019). TFV-DP was not detectable in vaginal tissue from either group. A high-dose DMPA treatment in macaques was associated with increased rectal TFV-DP levels, indicating a potential tissue-specific drug-drug interaction. The lack of detectable TFV-DP in the vaginal tissue warrants further investigation of PrEP efficacy with single-agent TAF products. DMPA did not affect systemic TAF metabolism, with similar PBMC TFV-DP in both groups, suggesting that DMPA use should not alter the antiviral activity of TAF.
Collapse
Affiliation(s)
- Michele B Daly
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV, Hepatitis, STD, and Prevention, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA.
| | - Mara Sterling
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV, Hepatitis, STD, and Prevention, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - Angela Holder
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV, Hepatitis, STD, and Prevention, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA.
| | - Chuong Dinh
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV, Hepatitis, STD, and Prevention, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA.
| | - Kenji Nishiura
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV, Hepatitis, STD, and Prevention, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA.
| | - George Khalil
- Quantitative Sciences and Data Management Branch, Division of HIV/AIDS Prevention, National Center for HIV, Hepatitis, STD, and Prevention, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA.
| | - J Gerardo García-Lerma
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV, Hepatitis, STD, and Prevention, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA.
| | - Charles Dobard
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV, Hepatitis, STD, and Prevention, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA, 30329, USA.
| |
Collapse
|
10
|
Noël-Romas L, Perner M, Molatlhegi R, Farr Zuend C, Mabhula A, Hoger S, Lamont A, Birse K, Berard A, McCorrister S, Westmacott G, Leslie A, Poliquin V, Heffron R, McKinnon LR, Burgener AD. Vaginal microbiome-hormonal contraceptive interactions associate with the mucosal proteome and HIV acquisition. PLoS Pathog 2020; 16:e1009097. [PMID: 33362285 PMCID: PMC7790405 DOI: 10.1371/journal.ppat.1009097] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 01/07/2021] [Accepted: 10/26/2020] [Indexed: 01/02/2023] Open
Abstract
Alterations to the mucosal environment of the female genital tract, such as genital inflammation, have been associated with increased HIV acquisition in women. As the microbiome and hormonal contraceptives can affect vaginal mucosal immunity, we hypothesized these components may interact in the context of HIV susceptibility. Using previously published microbiome data from 685 women in the CAPRISA-004 trial, we compared relative risk of HIV acquisition in this cohort who were using injectable depot medroxyprogesterone acetate (DMPA), norethisterone enanthate (NET-EN), and combined oral contraceptives (COC). In women who were Lactobacillus-dominant, HIV acquisition was 3-fold higher in women using DMPA relative to women using NET-EN or COC (OR: 3.27; 95% CI: 1.24–11.24, P = 0.0305). This was not observed in non-Lactobacillus-dominant women (OR: 0.95, 95% CI: 0.44–2.15, P = 0.895) (interaction P = 0.0686). Higher serum MPA levels associated with increased molecular pathways of inflammation in the vaginal mucosal fluid of Lactobacillus-dominant women, but no differences were seen in non-Lactobacillus dominant women. This study provides data suggesting an interaction between the microbiome, hormonal contraceptives, and HIV susceptibility. Alterations to the mucosal environment of the female genital tract have been associated with increased HIV acquisition in women. As both the vaginal microbiome and hormonal contraceptives affect mucosal immunity, we investigated their interaction with HIV susceptibility. We characterized the vaginal microbiomes in 685 women from the CAPRISA-004 trial, who utilized three major types of hormonal contraceptives including injectable depot medroxyprogesterone acetate (DMPA), norethisterone enanthate (NET-EN), and combined oral contraceptives (COC). In the 40% of women with Lactobacillus-depleted microbiomes, HIV acquisition was not different between contraceptive groups. However, in the 60% of women with Lactobacillus as the dominant bacterial taxa, HIV acquisition risk was 3-fold higher (in women using DMPA relative to women using NET-EN and COC). Higher serum medroxyprogesterone acetate levels in Lactobacillus dominant women associated with increased cervicovaginal inflammation pathways in the mucosal proteome, biomarkers of which associated with HIV susceptibility. This study provides data suggesting an interaction between the microbiome, hormonal contraceptives, and HIV susceptibility.
Collapse
Affiliation(s)
- Laura Noël-Romas
- Departments of Obstetrics & Gynecology, University of Manitoba, Winnipeg, Canada
- Center for Global Health and Diseases, Case Western Reserve University, Ohio, United States of America
| | - Michelle Perner
- Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | | | - Christina Farr Zuend
- Center for Global Health and Diseases, Case Western Reserve University, Ohio, United States of America
| | | | - Sarah Hoger
- Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | - Alana Lamont
- Departments of Obstetrics & Gynecology, University of Manitoba, Winnipeg, Canada
| | - Kenzie Birse
- Departments of Obstetrics & Gynecology, University of Manitoba, Winnipeg, Canada
- Center for Global Health and Diseases, Case Western Reserve University, Ohio, United States of America
| | - Alicia Berard
- Departments of Obstetrics & Gynecology, University of Manitoba, Winnipeg, Canada
| | - Stuart McCorrister
- Mass Spectrometry and Proteomics Core Facility, National Microbiology Lab, Public Health Agency of Canada, Winnipeg, Canada
| | - Garett Westmacott
- Mass Spectrometry and Proteomics Core Facility, National Microbiology Lab, Public Health Agency of Canada, Winnipeg, Canada
| | - Al Leslie
- Africa Health Research Institute, Durban, South Africa
- Department of Infection and Immunity, University College London, London, United Kingdom
| | - Vanessa Poliquin
- Departments of Obstetrics & Gynecology, University of Manitoba, Winnipeg, Canada
| | - Renee Heffron
- Department of Global Health and Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
| | | | - Adam D. Burgener
- Departments of Obstetrics & Gynecology, University of Manitoba, Winnipeg, Canada
- Center for Global Health and Diseases, Case Western Reserve University, Ohio, United States of America
- Medical Microbiology, University of Manitoba, Winnipeg, Canada
- Unit of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
11
|
Edfeldt G, Lajoie J, Röhl M, Oyugi J, Åhlberg A, Khalilzadeh-Binicy B, Bradley F, Mack M, Kimani J, Omollo K, Wählby C, Fowke KR, Broliden K, Tjernlund A. Regular use of depot medroxyprogesterone acetate causes thinning of the superficial lining and apical distribution of HIV target cells in the human ectocervix. J Infect Dis 2020; 225:1151-1161. [PMID: 32780807 PMCID: PMC8974825 DOI: 10.1093/infdis/jiaa514] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/08/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The hormonal contraceptive depot medroxyprogesterone acetate (DMPA) may be associated with an increased risk of acquiring human immunodeficiency virus (HIV). We hypothesize that DMPA use influences the ectocervical tissue architecture and HIV target cell localization. METHODS Quantitative image analysis workflows were developed to assess ectocervical tissue samples collected from DMPA users and control subjects not using hormonal contraception. RESULTS Compared to controls, the DMPA group exhibited a significantly thinner apical ectocervical epithelial layer and a higher proportion of CD4+CCR5+ cells with a more superficial location. This localization corresponded to an area with a non-intact E-cadherin net structure. CD4+Langerin+ cells were also more superficially located in the DMPA group, while fewer in number compared to the controls. Natural plasma progesterone levels did not correlate with any of these parameters, whereas estradiol levels were positively correlated with E-cadherin expression and a more basal location for HIV target cells of the control group. CONCLUSIONS DMPA users have a less robust epithelial layer and a more apical distribution of HIV target cells in the human ectocervix, which could confer a higher risk of HIV infection. Our results highlight the importance of assessing intact genital tissue samples to gain insights into HIV susceptibility factors.
Collapse
Affiliation(s)
- Gabriella Edfeldt
- Department of Medicine Solna, Division of Infectious Diseases, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Julie Lajoie
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
| | - Maria Röhl
- Department of Medicine Solna, Division of Infectious Diseases, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Julius Oyugi
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
| | - Alexandra Åhlberg
- Department of Medicine Solna, Division of Infectious Diseases, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Behnaz Khalilzadeh-Binicy
- Department of Medicine Solna, Division of Infectious Diseases, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Frideborg Bradley
- Department of Medicine Solna, Division of Infectious Diseases, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Mathias Mack
- Department of Internal Medicine - Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Joshua Kimani
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya.,Partners for Health and Development in Africa, Nairobi, Kenya
| | - Kenneth Omollo
- Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
| | - Carolina Wählby
- Department of Information Technology, Uppsala University, Uppsala, Sweden.,SciLifeLab BioImage Informatics Facility, Uppsala, Sweden
| | - Keith R Fowke
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya.,Partners for Health and Development in Africa, Nairobi, Kenya.,Department of Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Kristina Broliden
- Department of Medicine Solna, Division of Infectious Diseases, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Annelie Tjernlund
- Department of Medicine Solna, Division of Infectious Diseases, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| |
Collapse
|
12
|
Plasma concentration of injectable contraceptive correlates with reduced cervicovaginal growth factor expression in South African women. Mucosal Immunol 2020; 13:449-459. [PMID: 31896762 DOI: 10.1038/s41385-019-0249-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/10/2019] [Accepted: 12/15/2019] [Indexed: 02/06/2023]
Abstract
Long-acting injectable contraceptives have been associated with mucosal immune changes and increased HIV acquisition, but studies have often been hampered by the inaccuracy of self-reported data, unknown timing of injection, and interactions with mucosal transmission co-factors. We used mass spectrometry to quantify the plasma concentrations of injectable contraceptives in women from the CAPRISA004 study (n = 664), with parallel quantification of 48 cytokines and >500 host proteins in cervicovaginal lavage. Higher DMPA levels were associated with reduced CVL concentrations of GCSF, MCSF, IL-16, CTACK, LIF, IL-1α, and SCGF-β in adjusted linear mixed models. Dose-dependent relationships between DMPA concentration and genital cytokines were frequently observed. Unsupervised clustering of host proteins by DMPA concentration suggest that women with low DMPA had increases in proteins associated with mucosal fluid function, growth factors, and keratinization. Although DMPA was not broadly pro-inflammatory, DMPA was associated with increased IP-10 in HSV-2 seropositive and older women. DMPA-cytokine associations frequently differed by vaginal microbiome; in non-Lactobacillus-dominant women, DMPA was associated with elevated IL-8, MCP-1, and IP-10 concentrations. These data confirm a direct, concentration-dependant effect of DMPA on functionally important immune factors within the vaginal compartment. The biological effects of DMPA may vary depending on age, HSV-2 status, and vaginal microbiome composition.
Collapse
|
13
|
Depot medroxyprogesterone acetate administration increases cervical CCR5+CD4+ T cells and induces immunosuppressive milieu at the cervicovaginal mucosa. AIDS 2020; 34:729-735. [PMID: 31972606 DOI: 10.1097/qad.0000000000002475] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVES Depot medroxyprogesterone acetate (Depo-Provera) is the most commonly used injectable hormone contraceptive in Sub-Saharan Africa where HIV incidence is high. We determined the impact of Depo-Provera on cervical immune cells and mediators in healthy women. METHODS In this longitudinal study, vaginal, endocervical, and rectal swabs were collected at baseline (visit 1), 1 month (visit 2), and 3 months (visit 3) after Depo-Provera injection. Cervical cells were collected by cytobrush and immune markers on cervical CD4 T cells were analyzed by multicolor flow cytometry at three different visits. The levels of immune mediators in cytobrush supernatants as well as vaginal, cervical, and rectal secretions from swabs were analyzed by multiplex assays and ELISA. RESULTS Compared with baseline levels, we found a significant increase in the frequency of cervical CCR5CD4 T cells and a significant decrease in the frequency of cervical central memory CD4 T cells. Depo-Provera treatment had little effect on expression of immune mediators in rectal mucosa but significantly suppressed numerous immune mediators at cervicovaginal mucosa. Levels of MCP-1, G-CSF, IL-6, IL-10, GM-CSF, and IP-10 were significantly decreased in both vaginal and cervical secretions after Depo-Provera injection. In cervical samples collected by cytobrush, we found reduced levels of 22 of 25 immune mediators after Depo-Provera injection. Changes in immune mediators differed between vaginal and cervical mucosa, demonstrating compartment-specific responses. CONCLUSION Depo-Provera altered immune profiles of cervical CD4 T cells and suppressed host immune response at cervicovaginal mucosa, suggesting its likely effect on transmission of sexually transmitted infections including HIV.
Collapse
|
14
|
Thurman A, Chandra N, Schwartz JL, Brache V, Chen BA, Asin S, Rollenhagen C, Herold BC, Fichorova RN, Hillier SL, Weiner DH, Mauck C, Doncel GF. The Effect of Hormonal Contraception on Cervicovaginal Mucosal End Points Associated with HIV Acquisition. AIDS Res Hum Retroviruses 2019; 35:853-864. [PMID: 30997816 DOI: 10.1089/aid.2018.0298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Reproductive age women may choose to concurrently use topical antiretrovirals and hormonal contraceptives (HCs) to simultaneously prevent HIV-1 infection and unintended/mistimed pregnancy. There are conflicting data on the effect of HCs on mucosal susceptibility to HIV-1. The objective of this study was to evaluate cervicovaginal (CV) mucosal data from healthy women before and after initiation of either oral contraceptive pills (OCPs) or depot medroxyprogesterone acetate (DMPA) injection. CONRAD A10-114 was a prospective, open-label, parallel cohort study. We enrolled 74 women and 62 completed the visits (32 and 30 who selected OCPs and DMPA, respectively). Participants provided CV lavage, vaginal biopsies, and CV swabs at baseline in the luteal phase and then ∼6 weeks after initiating HCs. After contraceptive initiation, there were significant increases in vaginal immune cell density among both DMPA and OCP users. Changes for OCP users were concentrated in the subepithelial lamina propria, whereas for DMPA users, they were distributed throughout the vaginal tissue, including the epithelium (CD45+, CD3+, CD4+, and CD1a+). Contraceptive use altered concentrations of soluble CV inflammatory and immune mediators, with significant reductions in some proinflammatory cytokines and secretory leukoprotease inhibitor. Compared with baseline, p24 antigen production after ex vivo HIV-1 infection of vaginal biopsies doubled after DMPA use, but all p-values were >.05. HIV-1 replication was significantly higher in DMPA-exposed tissues compared with those from the OCP group at the end of the tissue culture (p = .01). Although not statistically significant, median in vitro inhibition of HIV-1 by CV fluid (innate antiviral activity), was reduced by ∼50% with HCs (p > .21). Exposure to exogenous contraceptive hormones significantly increased vaginal immune cells and reduced CV proinflammatory cytokines and antimicrobial peptides. DMPA users showed higher susceptibility to HIV-1 ex vivo infection.
Collapse
Affiliation(s)
- Andrea Thurman
- Eastern Virginia Medical School, CONRAD, Norfolk, Virginia
| | | | | | | | - Beatrice A. Chen
- Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Susana Asin
- V.A. Medical Center, White River Junction, VT and Geisel School of Medicine, Dartmouth, New Hampshire
| | - Christiane Rollenhagen
- V.A. Medical Center, White River Junction, VT and Geisel School of Medicine, Dartmouth, New Hampshire
| | - Betsy C. Herold
- Department of Pediatric Infectious Diseases, Albert Einstein College of Medicine, Bronx, New York
| | - Raina N. Fichorova
- Laboratory of Genital Tract Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Sharon L. Hillier
- Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | | | | |
Collapse
|
15
|
Zalenskaya IA, Chandra N, Yousefieh N, Fang X, Adedipe OE, Jackson SS, Anderson SM, Mauck CK, Schwartz JL, Thurman AR, Doncel GF. Use of contraceptive depot medroxyprogesterone acetate is associated with impaired cervicovaginal mucosal integrity. J Clin Invest 2018; 128:4622-4638. [PMID: 30222141 DOI: 10.1172/jci120583] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 07/31/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Injectable depot medroxyprogesterone acetate (DMPA) is one of the most popular contraception methods in areas of high HIV seroprevalence. Evidence is accumulating that use of DMPA might be associated with an increased risk of HIV-1 acquisition by women; however, mechanisms of this association are not completely understood. The goal of this study was to gain insight into mechanisms underlying the possible link between use of DMPA and risk of HIV-1 acquisition, exploring transcription profiling of ectocervical tissues. METHODS Healthy women received either DMPA (n = 31) or combined oral contraceptive (COC), which has not been linked to an increased risk of HIV acquisition (n = 32). We conducted a comparative microarray-based whole-genome transcriptome profiling of human ectocervical tissues before and after 6 weeks of hormonal contraception use. RESULTS The analysis identified that expression of 235 and 76 genes was significantly altered after DMPA and COC use, respectively. The most striking effect of DMPA, but not COC, was significantly altered expression (mostly downregulation) of many genes strategically involved in the maintenance of mucosal barrier function; the alterations, as indicated by Ingenuity Pathway Analysis (IPA), were most likely due to the DMPA-induced estrogen deficiency. Furthermore, IPA predicted that transcriptome alterations related to ectocervical immune responses were in general compatible with an immunosuppressive effect of DMPA, but, in some women, also with an inflammatory-like response. CONCLUSION Our results suggest that impairment of cervicovaginal mucosal integrity in response to DMPA administration is an important mechanism contributing to the potential increased risk of HIV-1 acquisition in DMPA users. TRIAL REGISTRATION ClinicalTrials.gov NCT01421368. FUNDING This study was supported by the United States Agency for International Development (USAID) under Cooperative Agreement GPO-A-00-08-00005-00.
Collapse
Affiliation(s)
| | - Neelima Chandra
- CONRAD, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | | | - Xi Fang
- CONRAD, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | | | | | | | | | - Jill L Schwartz
- CONRAD, Eastern Virginia Medical School, Arlington, Virginia, USA
| | | | - Gustavo F Doncel
- CONRAD, Eastern Virginia Medical School, Norfolk, Virginia, USA.,CONRAD, Eastern Virginia Medical School, Arlington, Virginia, USA
| |
Collapse
|
16
|
Abstract
Biological sex is a determinant of both susceptibility to and pathogenesis of multiple infections, including HIV. These differences have effects on the spectrum of HIV disease from acquisition to eradication, with diverse mechanisms including distinct chromosomal complements, variation in microbiota composition, hormonal effects on transcriptional profiles, and expression of different immunoregulatory elements. With a comparative biology approach, these sex differences can be used to highlight protective and detrimental immune activation pathways, to identify strategies for effective prevention, treatment, and curative interventions.
Collapse
Affiliation(s)
- Eileen P Scully
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW This review will outline the multilevel effects of biological sex on HIV acquisition, pathogenesis, treatment response, and prospects for cure. Potential mechanisms will be discussed along with future research directions. RECENT FINDINGS HIV acquisition risk is modified by sex hormones and the vaginal microbiome, with the latter acting through both inflammation and local metabolism of pre-exposure prophylaxis drugs. Female sex associates with enhanced risk for non-AIDS morbidities including cardiovascular and cerebrovascular disease, suggesting different inflammatory profiles in men and women. Data from research on HIV cure points to sex differences in viral reservoir dynamics and a direct role for sex hormones in latency maintenance. Biological sex remains an important variable in determining the risk of HIV infection and subsequent viral pathogenesis, and emerging data suggest sex differences relevant to curative interventions. Recruitment of women in HIV clinical research is a pathway to both optimize care for women and to identify novel therapeutics for use in both men and women.
Collapse
Affiliation(s)
- Eileen P Scully
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Pre-Clinical Teaching Building, Suite 211, 725 N Wolfe Street, Baltimore, MD, 21205, USA.
| |
Collapse
|
18
|
Hapgood JP, Kaushic C, Hel Z. Hormonal Contraception and HIV-1 Acquisition: Biological Mechanisms. Endocr Rev 2018; 39:36-78. [PMID: 29309550 PMCID: PMC5807094 DOI: 10.1210/er.2017-00103] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 11/27/2017] [Indexed: 12/12/2022]
Abstract
Access to effective affordable contraception is critical for individual and public health. A wide range of hormonal contraceptives (HCs), which differ in composition, concentration of the progestin component, frequency of dosage, and method of administration, is currently available globally. However, the options are rather limited in settings with restricted economic resources that frequently overlap with areas of high HIV-1 prevalence. The predominant contraceptive used in sub-Saharan Africa is the progestin-only three-monthly injectable depot medroxyprogesterone acetate. Determination of whether HCs affect HIV-1 acquisition has been hampered by behavioral differences potentially confounding clinical observational data. Meta-analysis of these studies shows a significant association between depot medroxyprogesterone acetate use and increased risk of HIV-1 acquisition, raising important concerns. No association was found for combined oral contraceptives containing levonorgestrel, nor for the two-monthly injectable contraceptive norethisterone enanthate, although data for norethisterone enanthate are limited. Susceptibility to HIV-1 and other sexually transmitted infections may, however, be dependent on the type of progestin present in the formulation. Several underlying biological mechanisms that may mediate the effect of HCs on HIV-1 and other sexually transmitted infection acquisition have been identified in clinical, animal, and ex vivo studies. A substantial gap exists in the translation of basic research into clinical practice and public health policy. To bridge this gap, we review the current knowledge of underlying mechanisms and biological effects of commonly used progestins. The review sheds light on issues critical for an informed choice of progestins for the identification of safe, effective, acceptable, and affordable contraceptive methods.
Collapse
Affiliation(s)
- Janet P Hapgood
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Charu Kaushic
- Department of Pathology and Molecular Medicine, McMaster University, Ontario, Canada.,McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Zdenek Hel
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama.,Center for AIDS Research, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
19
|
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: 2.0] [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
|
20
|
Riley HEM, Steyn PS, Achilles SL, Bass E, Gray AL, Polis CB, Kiarie JN. Hormonal contraceptive methods and HIV: research gaps and programmatic priorities. Contraception 2017; 96:67-71. [PMID: 28601676 PMCID: PMC6298743 DOI: 10.1016/j.contraception.2017.05.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 05/29/2017] [Accepted: 05/30/2017] [Indexed: 10/19/2022]
Affiliation(s)
- Halley E M Riley
- Emory University, Department of Behavioral Sciences and Health Education, 1518 Clifton Road, Atlanta, GA, 30322, USA.
| | - Petrus S Steyn
- World Health Organization, Human Reproduction Team (HRX), Department of Reproductive Health and Research, Geneva, Switzerland.
| | - Sharon L Achilles
- University of Pittsburgh, Department of Obstetrics, Gynecology and Reproductive Sciences, Halket Street, Pittsburgh, PA 15213, USA; Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
| | - Emily Bass
- AVAC, 423 West 127th Street, New York, NY 10027, USA
| | - Andrew L Gray
- Division of Pharmacology, Discipline of Pharmaceutical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Chelsea B Polis
- Guttmacher Institute, 125 Maiden Lane, 7th Floor, New York, NY 10038, USA
| | - James N Kiarie
- World Health Organization, Human Reproduction Team (HRX), Department of Reproductive Health and Research, Geneva, Switzerland
| |
Collapse
|
21
|
Sex and gender differences in HIV-1 infection. Clin Sci (Lond) 2017; 130:1435-51. [PMID: 27389589 DOI: 10.1042/cs20160112] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 05/06/2016] [Indexed: 01/03/2023]
Abstract
The major burden of the human immunodeficiency (HIV) type 1 pandemic is nowadays carried by women from sub-Saharan Africa. Differences in the manifestations of HIV-1 infection between women and men have been long reported, and might be due to both socio-economic (gender) and biological (sex) factors. Several studies have shown that women are more susceptible to HIV-1 acquisition than men. Following HIV-1 infection, women have lower viral loads during acute infection and exhibit stronger antiviral responses than men, which may contribute to differences in the size of viral reservoirs. Oestrogen receptor signalling could represent an important mediator of sex differences in HIV-1 reservoir size and may represent a potential therapeutic target. Furthermore, immune activation, a hallmark of HIV-1 infection, is generally higher in women than in men and could be a central mechanism in the sex difference observed in the speed of HIV-1 disease progression. Here, we review the literature regarding sex-based differences in HIV-1 infection and discuss how a better understanding of the underlying mechanisms could improve preventive and therapeutic strategies.
Collapse
|
22
|
Scarsi KK, Darin KM, Chappell CA, Nitz SM, Lamorde M. Drug-Drug Interactions, Effectiveness, and Safety of Hormonal Contraceptives in Women Living with HIV. Drug Saf 2016; 39:1053-1072. [PMID: 27562873 PMCID: PMC5048570 DOI: 10.1007/s40264-016-0452-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Family planning options, including hormonal contraceptives, are essential for improving reproductive health among the more than 17 million women living with HIV worldwide. For these women, prevention of unintended pregnancy decreases maternal and child mortality, as well as reduces the risk of perinatal HIV transmission. Similarly, treatment of HIV with antiretroviral therapy (ART) is essential for reducing morbidity and mortality among HIV-positive individuals, as well as preventing HIV transmission between sexual partners or from mother to child. Importantly, despite the benefits of hormonal contraceptives, barriers to effective family planning methods exist for HIV-positive women. Specifically, drug-drug interactions can occur between some antiretroviral medications and some hormonal contraceptives, which may influence both contraceptive efficacy and tolerability. In addition, safety concerns have been raised about the impact of hormonal contraceptives on HIV disease progression, tolerability, and the risk of female-to-male HIV transmission. This review article summarizes the potential for drug-drug interactions, tolerability, and contraceptive effectiveness when hormonal contraceptives are combined with ART. In addition, the evidence surrounding the influence of hormonal contraceptives on HIV transmission and HIV disease progression in women living with HIV are summarized.
Collapse
Affiliation(s)
- Kimberly K Scarsi
- Department of Pharmacy Practice, College of Pharmacy, University of Nebraska Medical Center, 986145 Nebraska Medical Center, Omaha, NE, 68198-6145, USA.
| | - Kristin M Darin
- Center for Global Health, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Catherine A Chappell
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Hospital, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Stephanie M Nitz
- Department of Pharmacy Practice, College of Pharmacy, University of Nebraska Medical Center, 986145 Nebraska Medical Center, Omaha, NE, 68198-6145, USA
| | - Mohammed Lamorde
- Infectious Diseases Institute, Makerere University College of Health Sciences, Kampala, Uganda
| |
Collapse
|
23
|
Calla NEQ, Miguel RDV, Boyaka PN, Hall-Stoodley L, Kaur B, Trout W, Pavelko SD, Cherpes TL. Medroxyprogesterone acetate and levonorgestrel increase genital mucosal permeability and enhance susceptibility to genital herpes simplex virus type 2 infection. Mucosal Immunol 2016; 9:1571-1583. [PMID: 27007679 PMCID: PMC5035233 DOI: 10.1038/mi.2016.22] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 02/19/2016] [Indexed: 02/07/2023]
Abstract
Depot-medroxyprogesterone acetate (DMPA) is a hormonal contraceptive especially popular in areas with high prevalence of HIV and other sexually transmitted infections (STI). Although observational studies identify DMPA as an important STI risk factor, mechanisms underlying this connection are undefined. Levonorgestrel (LNG) is another progestin used for hormonal contraception, but its effect on STI susceptibility is much less explored. Using a mouse model of genital herpes simplex virus type 2 (HSV-2) infection, we herein found that DMPA and LNG similarly reduced genital expression of the desmosomal cadherin desmoglein-1α (DSG1α), enhanced access of inflammatory cells to genital tissue by increasing mucosal epithelial permeability, and increased susceptibility to viral infection. Additional studies with uninfected mice revealed that DMPA-mediated increases in mucosal permeability promoted tissue inflammation by facilitating endogenous vaginal microbiota invasion. Conversely, concomitant treatment of mice with DMPA and intravaginal estrogen restored mucosal barrier function and prevented HSV-2 infection. Evaluating ectocervical biopsy tissue from women before and 1 month after initiating DMPA remarkably revealed that inflammation and barrier protection were altered by treatment identically to changes seen in progestin-treated mice. Together, our work reveals DMPA and LNG diminish the genital mucosal barrier; a first-line defense against all STI, but may offer foundation for new contraceptive strategies less compromising of barrier protection.
Collapse
Affiliation(s)
- Nirk E Quispe Calla
- Department of Microbial infection & Immunity, The Ohio State University College of Medicine, Columbus, Ohio, USA,Corresponding authors: Thomas L. Cherpes, DVM, MD, Biomedical Research Tower, Room 712, 460 West 12th Ave., Columbus, OH 43210, USA, Telephone: 614.688.1897 Fax: 614.292.9616. Rodolfo D. Vicetti Miguel, MD, Biomedical Research Tower, Room 731, 460 West 12th Ave., Columbus, OH 43210, USA, Telephone: 614.688.2165 Fax: 614.292.9616. Nirk E. Quispe Calla, MD, Biomedical Research Tower, Room 740,460 West 12th Ave., Columbus, OH 43210, USA, Telephone: 614.688.2165 Fax: 614.292.9616
| | - Rodolfo D Vicetti Miguel
- Department of Microbial infection & Immunity, The Ohio State University College of Medicine, Columbus, Ohio, USA,Corresponding authors: Thomas L. Cherpes, DVM, MD, Biomedical Research Tower, Room 712, 460 West 12th Ave., Columbus, OH 43210, USA, Telephone: 614.688.1897 Fax: 614.292.9616. Rodolfo D. Vicetti Miguel, MD, Biomedical Research Tower, Room 731, 460 West 12th Ave., Columbus, OH 43210, USA, Telephone: 614.688.2165 Fax: 614.292.9616. Nirk E. Quispe Calla, MD, Biomedical Research Tower, Room 740,460 West 12th Ave., Columbus, OH 43210, USA, Telephone: 614.688.2165 Fax: 614.292.9616
| | - Prosper N Boyaka
- Department of Veterinary Biosciences, The Ohio State University College of Veterinary Medicine, Columbus, Ohio, USA
| | - Luanne Hall-Stoodley
- Department of Microbial infection & Immunity, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Balveen Kaur
- Department of Neurological Surgery, James Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, Ohio, USA
| | - Wayne Trout
- Department of Obstetrics & Gynecology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Stephen D Pavelko
- Department of Microbial infection & Immunity, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Thomas L Cherpes
- Department of Microbial infection & Immunity, The Ohio State University College of Medicine, Columbus, Ohio, USA,Department of Obstetrics & Gynecology, The Ohio State University College of Medicine, Columbus, Ohio, USA,Corresponding authors: Thomas L. Cherpes, DVM, MD, Biomedical Research Tower, Room 712, 460 West 12th Ave., Columbus, OH 43210, USA, Telephone: 614.688.1897 Fax: 614.292.9616. Rodolfo D. Vicetti Miguel, MD, Biomedical Research Tower, Room 731, 460 West 12th Ave., Columbus, OH 43210, USA, Telephone: 614.688.2165 Fax: 614.292.9616. Nirk E. Quispe Calla, MD, Biomedical Research Tower, Room 740,460 West 12th Ave., Columbus, OH 43210, USA, Telephone: 614.688.2165 Fax: 614.292.9616
| |
Collapse
|
24
|
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: 3.0] [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
|
25
|
Kersh EN, Ritter J, Butler K, Ostergaard SD, Hanson D, Ellis S, Zaki S, McNicholl JM. Relationship of Estimated SHIV Acquisition Time Points During the Menstrual Cycle and Thinning of Vaginal Epithelial Layers in Pigtail Macaques. Sex Transm Dis 2016; 42:694-701. [PMID: 26562699 DOI: 10.1097/olq.0000000000000367] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND HIV acquisition in the female genital tract remains incompletely understood. Quantitative data on biological HIV risk factors, the influence of reproductive hormones, and infection risk are lacking. We evaluated vaginal epithelial thickness during the menstrual cycle in pigtail macaques (Macaca nemestrina). This model previously revealed increased susceptibility to vaginal infection during and after progesterone-dominated periods in the menstrual cycle. METHODS Nucleated and nonnucleated (superficial) epithelial layers were quantitated throughout the menstrual cycle of 16 macaques. We examined the relationship with previously estimated vaginal SHIVSF162P3 acquisition time points in the cycle of 43 different animals repeatedly exposed to low virus doses. RESULTS In the luteal phase (days 17 to cycle end), the mean vaginal epithelium thinned to 66% of mean follicular thickness (days 1-16; P = 0.007, Mann-Whitney test). Analyzing 4-day segments, the epithelium was thickest on days 9 to 12 and thinned to 31% thereof on days 29 to 32, with reductions of nucleated and nonnucleated layers to 36% and 15% of their previous thickness, respectively. The proportion of animals with estimated SHIV acquisition in each cycle segment correlated with nonnucleated layer thinning (Pearson r = 0.7, P < 0.05, linear regression analysis), but not nucleated layer thinning (Pearson r = 0.6, P = 0.15). CONCLUSIONS These data provide a detailed picture of dynamic cycle-related changes in the vaginal epithelium of pigtail macaques. Substantial thinning occurred in the superficial, nonnucleated layer, which maintains the vaginal microbiome. The findings support vaginal tissue architecture as susceptibility factor for infection and contribute to our understanding of innate resistance to SHIV infection.
Collapse
Affiliation(s)
- Ellen N Kersh
- From the *National Center for HIV/AIDS, Viral Hepatitis, STD and TB Prevention and †National Center for Emerging & Zoonotic Infectious Diseases, CDC, Atlanta, GA; and ‡Total Solutions Inc, Atlanta, GA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Combination Pod-Intravaginal Ring Delivers Antiretroviral Agents for HIV Prophylaxis: Pharmacokinetic Evaluation in an Ovine Model. Antimicrob Agents Chemother 2016; 60:3759-66. [PMID: 27067321 DOI: 10.1128/aac.00391-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 04/03/2016] [Indexed: 12/24/2022] Open
Abstract
Preexposure prophylaxis (PrEP) against HIV using oral regimens based on the nucleoside reverse transcriptase inhibitor tenofovir disoproxil fumarate (TDF) has been effective to various degrees in multiple clinical trials, and the CCR5 receptor antagonist maraviroc (MVC) holds potential for complementary efficacy. The effectiveness of HIV PrEP is highly dependent on adherence. Incorporation of the TDF-MVC combination into intravaginal rings (IVRs) for sustained mucosal delivery could increase product adherence and efficacy compared with oral and vaginal gel formulations. A novel pod-IVR technology capable of delivering multiple drugs is described. The pharmacokinetics and preliminary local safety characteristics of a novel pod-IVR delivering a combination of TDF and MVC were evaluated in the ovine model. The device exhibited sustained release at controlled rates over the 28-day study and maintained steady-state drug levels in cervicovaginal fluids (CVFs). Dilution of CVFs during lavage sample collection was measured by ion chromatography using an inert tracer, allowing corrected drug concentrations to be measured for the first time. Median, steady-state drug levels in vaginal tissue homogenate were as follows: for tenofovir (TFV; in vivo hydrolysis product of TDF), 7.3 × 10(2) ng g(-1) (interquartile range [IQR], 3.0 × 10(2), 4.0 × 10(3)); for TFV diphosphate (TFV-DP; active metabolite of TFV), 1.8 × 10(4) fmol g(-1) (IQR, 1.5 × 10(4), 4.8 × 10(4)); and for MVC, 8.2 × 10(2) ng g(-1) (IQR, 4.7 × 10(2), 2.0 × 10(3)). No adverse events were observed. These findings, together with previous pod-IVR studies, have allowed several lead candidates to advance into clinical evaluation.
Collapse
|
27
|
Hormonal Contraceptive Effects on the Vaginal Milieu: Microbiota and Immunity. CURRENT OBSTETRICS AND GYNECOLOGY REPORTS 2016. [DOI: 10.1007/s13669-016-0142-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
28
|
Butler K, Ritter J, Ellis S, Henning TR, Montague J, Zaki S, Garber D, McNicholl JM, Kersh EN. Analysis of putative mucosal SHIV susceptibility factors during repeated DMPA treatments in pigtail macaques. J Med Primatol 2015; 44:286-95. [PMID: 26238265 DOI: 10.1111/jmp.12188] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2015] [Indexed: 01/18/2023]
Abstract
BACKGROUND Depot-medroxyprogesterone acetate (DMPA) has been associated in some studies with increased HIV susceptibility in women. We used a pigtail macaque model to document the effects of repeated DMPA treatments and their potential contribution to increased SHIV susceptibility. METHODS Nine pigtails were administered 2.5, 1.5, or 0.5 mg/kg DMPA in study weeks one and four. Menstrual cycling, vaginal epithelial thickness, and other SHIV susceptibility factors were monitored for a mean of 24 study weeks. RESULTS All DMPA treatments suppressed menstrual cycling and increased vaginal pH. The vaginal epithelium thinned naturally during baseline menstrual cycles (from mean of 351 to 161 μm in late-luteal phase). Following DMPA, the non-nucleated layer was temporarily absent. Two weeks post-second DMPA injection, mean epithelial thickness was 53, 45, and 167 μm for the descending doses, respectively. CONCLUSIONS All animals showed temporal vaginal epithelial thinning with loss of the non-nucleated layer, and vaginal pH changes post-DMPA injections.
Collapse
Affiliation(s)
- Katherine Butler
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, Sexually Transmitted Disease and Tuberculosis Prevention, CDC, Atlanta, GA, USA
| | - Jana Ritter
- Infectious Diseases Pathology Branch, Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, CDC, Atlanta, GA, USA
| | | | - Tara R Henning
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, Sexually Transmitted Disease and Tuberculosis Prevention, CDC, Atlanta, GA, USA
| | - Jeltley Montague
- Infectious Diseases Pathology Branch, Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, CDC, Atlanta, GA, USA
| | - Sherif Zaki
- Infectious Diseases Pathology Branch, Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, CDC, Atlanta, GA, USA
| | - David Garber
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, Sexually Transmitted Disease and Tuberculosis Prevention, CDC, Atlanta, GA, USA
| | - Janet M McNicholl
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, Sexually Transmitted Disease and Tuberculosis Prevention, CDC, Atlanta, GA, USA
| | - Ellen N Kersh
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, Sexually Transmitted Disease and Tuberculosis Prevention, CDC, Atlanta, GA, USA
| |
Collapse
|
29
|
|
30
|
Holt JDS, Cameron D, Dias N, Holding J, Muntendam A, Oostebring F, Dreier P, Rohan L, Nuttall J. The sheep as a model of preclinical safety and pharmacokinetic evaluations of candidate microbicides. Antimicrob Agents Chemother 2015; 59:3761-70. [PMID: 25845860 PMCID: PMC4468677 DOI: 10.1128/aac.04954-14] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 03/29/2015] [Indexed: 12/11/2022] Open
Abstract
When developing novel microbicide products for the prevention of HIV infection, the preclinical safety program must evaluate not only the active pharmaceutical ingredient but also the product itself. To that end, we applied several relatively standard toxicology study methodologies to female sheep, incorporating an assessment of the pharmacokinetics, safety, tolerability, and local toxicity of a dapivirine-containing human vaginal ring formulation (Dapivirine Vaginal Ring-004). We performed a 3-month general toxicology study, a preliminary pharmacokinetic study using drug-loaded vaginal gel, and a detailed assessment of the kinetics of dapivirine delivery to plasma, vaginal, and rectal fluid and rectal, vaginal, and cervical tissue over 28 days of exposure and 3 and 7 days after removal of the ring. The findings of the general toxicology study supported the existing data from both preclinical and clinical studies in that there were no signs of toxicity related to dapivirine. In addition, the presence of the physical dapivirine ring did not alter local or systemic toxicity or the pharmacokinetics of dapivirine. Pharmacokinetic studies indicated that the dapivirine ring produced significant vaginal tissue levels of dapivirine. However, no dapivirine was detected in cervical tissue samples using the methods described here. Plasma and vaginal fluid levels were lower than those in previous clinical studies, while there were detectable dapivirine levels in the rectal tissue and fluid. All tissue and fluid levels tailed off rapidly to undetectable levels following removal of the ring. The sheep represents a very useful model for the assessment of the safety and pharmacokinetics of microbicide drug delivery devices, such as the vaginal ring.
Collapse
Affiliation(s)
- Jonathon D S Holt
- International Partnership for Microbicides, Silver Spring, Maryland, USA
| | - David Cameron
- Huntingdon Life Sciences, Huntingdon Research Centre, Huntingdon, Cambridgeshire, United Kingdom
| | - Nicola Dias
- Huntingdon Life Sciences, Huntingdon Research Centre, Huntingdon, Cambridgeshire, United Kingdom
| | - Jeremy Holding
- Huntingdon Life Sciences, Huntingdon Research Centre, Huntingdon, Cambridgeshire, United Kingdom
| | | | | | | | - Lisa Rohan
- University of Pittsburgh, Magee Women's Research Institute, Pittsburgh, Pennsylvania, USA
| | - Jeremy Nuttall
- International Partnership for Microbicides, Silver Spring, Maryland, USA
| |
Collapse
|
31
|
Grabowski MK, Gray RH, Makumbi F, Kagaayi J, Redd AD, Kigozi G, Reynolds SJ, Nalugoda F, Lutalo T, Wawer MJ, Serwadda D, Quinn TC, Tobian AAR. Use of injectable hormonal contraception and women's risk of herpes simplex virus type 2 acquisition: a prospective study of couples in Rakai, Uganda. LANCET GLOBAL HEALTH 2015; 3:e478-e486. [PMID: 26094162 DOI: 10.1016/s2214-109x(15)00086-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 02/03/2015] [Accepted: 02/11/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND The injectable hormonal contraceptive depo-medroxyprogesterone acetate (DMPA) has been associated with increased risk of HIV acquisition, but findings are inconsistent. Whether DMPA increases the risk of other sexually transmitted viral infections is unknown. We assessed the association between DMPA use and incident herpes simplex virus type 2 (HSV2) infection in women. METHODS In this prospective study, we enrolled HIV-negative and HSV2-negative women aged 15-49 years whose HIV-negative male partners were concurrently enrolled in a randomised trial of male circumcision in Rakai, Uganda. We excluded women if either they or their male partners HIV seroconverted. The primary outcome was HSV2 seroconversion, assessed annually. The male circumcision trial was registered with ClinicalTrials.gov, number NCT00425984. FINDINGS Between Aug 11, 2003, and July 6, 2006, we enrolled 682 women in this study. We noted HSV2 seroconversions in 70 (10%) women. Incidence was 13·5 per 100 person-years in women consistently using DMPA (nine incident infections per 66·5 person-years), 4·3 per 100 person-years in pregnant women who were not using hormonal contraception (18 incident infections per 423·5 person-years), and 6·6 per 100 person-years in women who were neither pregnant nor using hormonal contraception (35 incident infections per 529·5 person-years). Women consistently using DMPA had an adjusted hazard ratio for HSV2 seroconversion of 2·26 (95% CI 1·09-4·69; p=0·029) compared with women who were neither pregnant nor using hormonal contraception. Of 132 women with HSV2-seropositive partners, seroconversion was 36·4 per 100 person-years in consistent DMPA users (four incident infections per 11 person-years) and 10·7 per 100 person-years in women who were neither pregnant nor using hormonal contraception (11 incident infections per 103 person-years; adjusted hazard ratio 6·23, 95% CI 1·49-26·3; p=0·012). INTERPRETATION Consistent DMPA use might increase risk of HSV2 seroconversion; however, study power was low. These findings should be assessed in larger populations with more frequent follow-up than in this study, and other contraceptive methods should also be assessed. Access to a wide range of highly effective contraceptive methods is needed for women, particularly in sub-Saharan Africa. FUNDING Bill and Melinda Gates Foundation, Doris Duke Charitable Foundation, US National Institutes of Health, and Fogarty International Center.
Collapse
Affiliation(s)
- Mary K Grabowski
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Ronald H Gray
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; Rakai Health Sciences Program, Entebbe, Uganda
| | | | | | - Andrew D Redd
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, MD, USA
| | | | - Steven J Reynolds
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA; Rakai Health Sciences Program, Entebbe, Uganda; Division of Intramural Research, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, MD, USA
| | | | - Tom Lutalo
- Rakai Health Sciences Program, Entebbe, Uganda
| | - Maria J Wawer
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; Rakai Health Sciences Program, Entebbe, Uganda
| | - David Serwadda
- Rakai Health Sciences Program, Entebbe, Uganda; School of Public Health, Makerere University, Kampala, Uganda
| | - Thomas C Quinn
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA; Division of Intramural Research, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, MD, USA
| | - Aaron A R Tobian
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA; Rakai Health Sciences Program, Entebbe, Uganda.
| |
Collapse
|
32
|
Michel KG, Huijbregts RP, Gleason JL, Richter HE, Hel Z. Effect of hormonal contraception on the function of plasmacytoid dendritic cells and distribution of immune cell populations in the female reproductive tract. J Acquir Immune Defic Syndr 2015; 68:511-8. [PMID: 25763784 PMCID: PMC4874780 DOI: 10.1097/qai.0000000000000531] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Epidemiological evidence suggests an association between the use of hormonal contraception and an increased risk of acquiring sexually transmitted diseases including HIV-1. We sought to elucidate the biological mechanisms underlying the effect of hormonal contraception on the immune system. DESIGN Cross-sectional study. METHODS To delineate the biological mechanisms underlying the effect of hormonal contraceptives on the immune system, we analyzed the functional capacity of circulating plasmacytoid dendritic cells (pDCs), the distribution of vaginal immune cell populations, and the systemic and genital levels of immune mediators in women using depot medroxyprogesterone acetate (DMPA), NuvaRing, or combined oral contraceptives (COC). RESULTS The use of DMPA or NuvaRing was associated with reduced capacity of circulating pDCs to produce interferon (IFN)-α and tumor necrosis (TNF-α) in response to TLR-9 stimulation. Systemic levels of IFN-α and cervicovaginal fluid levels of IFN-α, CXCL10, monocyte chemotactic protein-1, and granulocyte-colony stimulating factor were significantly lower in DMPA users compared to control volunteers not using hormonal contraception. The density of CD207 Langerhans cells in the vaginal epithelium was reduced in NuvaRing and combined oral contraceptive users but not in DMPA users. CONCLUSIONS The presented evidence suggests that the use of some types of hormonal contraception is associated with reduced functional capacity of circulating pDCs and altered immune environment in the female reproductive tract.
Collapse
Affiliation(s)
- Katherine G. Michel
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Jonathan L. Gleason
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Holly E. Richter
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Zdenek Hel
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Center for AIDS Research, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
33
|
Irvin SC, Herold BC. Molecular mechanisms linking high dose medroxyprogesterone with HIV-1 risk. PLoS One 2015; 10:e0121135. [PMID: 25798593 PMCID: PMC4370479 DOI: 10.1371/journal.pone.0121135] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 02/11/2015] [Indexed: 11/24/2022] Open
Abstract
Background Epidemiological studies suggest that medroxyprogesterone acetate (MPA) may increase the risk of HIV-1. The current studies were designed to identify potential underlying biological mechanisms. Methods Human vaginal epithelial (VK2/E6E7), peripheral blood mononuclear (PBMC), and polarized endometrial (HEC-1-A) cells were treated with a range of concentrations of MPA (0.015-150 μg/ml) and the impact on gene expression, protein secretion, and HIV infection was evaluated. Results Treatment of VK2/E6E7 cells with high doses (>15μg/ml] of MPA significantly upregulated proinflammatory cytokines, which resulted in a significant increase in HIV p24 levels secreted by latently infected U1 cells following exposure to culture supernatants harvested from MPA compared to mock-treated cells. MPA also increased syndecan expression by VK2/E6E7 cells and cells treated with 15 μg/ml of MPA bound and transferred more HIV-1 to T cells compared to mock-treated cells. Moreover, MPA treatment of epithelial cells and PBMC significantly decreased cell proliferation resulting in disruption of the epithelial barrier and decreased cytokine responses to phytohaemagglutinin, respectively. Conclusion We identified several molecular mechanisms that could contribute to an association between DMPA and HIV including proinflammatory cytokine and chemokine responses that could activate the HIV promoter and recruit immune targets, increased expression of syndecans to facilitate the transfer of virus from epithelial to immune cells and decreased cell proliferation. The latter could impede the ability to maintain an effective epithelial barrier and adversely impact immune cell function. However, these responses were observed primarily following exposure to high (15-150 μg/ml) MPA concentrations. Clinical correlation is needed to determine whether the prolonged MPA exposure associated with contraception activates these mechanisms in vivo.
Collapse
Affiliation(s)
- Susan C. Irvin
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, 10461, United States of America and Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, United States of America
| | - Betsy C. Herold
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, 10461, United States of America and Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, United States of America
- * E-mail:
| |
Collapse
|
34
|
Depot-medroxyprogesterone acetate does not reduce the prophylactic efficacy of emtricitabine and tenofovir disoproxil fumarate in macaques. J Acquir Immune Defic Syndr 2015; 67:365-9. [PMID: 25202923 DOI: 10.1097/qai.0000000000000340] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Concerns that the injectable contraceptive depot-medroxyprogesterone acetate (DMPA) may increase the risk of HIV acquisition in women led to questions on whether DMPA could reduce efficacy of pre-exposure prophylaxis (PrEP) for HIV prevention. We used a macaque model to investigate the impact of prolonged DMPA exposure on PrEP with emtricitabine/tenofovir disoproxil fumarate. Twelve pigtail macaques treated with DMPA were exposed vaginally to simian HIV once a week for up to 5 months and received either placebo (n = 6) or emtricitabine/tenofovir disoproxil fumarate (n = 6). All control macaques were infected, whereas the PrEP-treated animals remained protected (P = 0.0007). This model suggests that women using DMPA will fully benefit from PrEP.
Collapse
|
35
|
Tjernlund A, Carias AM, Andersson S, Gustafsson-Sanchez S, Röhl M, Petersson P, Introini A, Hope TJ, Broliden K. Progesterone-based intrauterine device use is associated with a thinner apical layer of the human ectocervical epithelium and a lower ZO-1 mRNA expression. Biol Reprod 2015; 92:68. [PMID: 25588510 DOI: 10.1095/biolreprod.114.122887] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Currently, whether hormonal contraceptives affect male to female human immunodeficiency virus (HIV) transmission is being debated. In this study, we investigated whether the use of progesterone-based intrauterine devices (pIUDs) is associated with a thinning effect on the ectocervical squamous epithelium, down-regulation of epithelial junction proteins, and/or alteration of HIV target cell distribution in the human ectocervix. Ectocervical tissue biopsies from healthy premenopausal volunteers using pIUDs were collected and compared to biopsies obtained from two control groups, namely women using combined oral contraceptives (COCs) or who do not use hormonal contraceptives. In situ staining and image analysis were used to measure epithelial thickness and the presence of HIV receptors in tissue biopsies. Messenger RNA levels of epithelial junction markers were measured by quantitative PCR. The epithelial thickness displayed by women in the pIUD group was similar to those in the COC group, but significantly thinner as compared to women in the no hormonal contraceptive group. The thinner epithelial layer of the pIUD group was specific to the apical layer of the ectocervix. Furthermore, the pIUD group expressed significantly lower levels of the tight junction marker ZO-1 within the epithelium as compared to the COC group. Similar expression levels of HIV receptors and coreceptors CD4, CCR5, DC-SIGN, and Langerin were observed in the three study groups. Thus, women using pIUD displayed a thinner apical layer of the ectocervical epithelium and reduced ZO-1 expression as compared to control groups. These data suggest that pIUD use may weaken the ectocervical epithelial barrier against invading pathogens, including HIV.
Collapse
Affiliation(s)
- Annelie Tjernlund
- Department of Medicine Solna, Unit of Infectious Diseases, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ann M Carias
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Sonia Andersson
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Susanna Gustafsson-Sanchez
- Department of Medicine Solna, Unit of Infectious Diseases, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Röhl
- Department of Medicine Solna, Unit of Infectious Diseases, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Pernilla Petersson
- Department of Medicine Solna, Unit of Infectious Diseases, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Andrea Introini
- Department of Medicine Solna, Unit of Infectious Diseases, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas J Hope
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Kristina Broliden
- Department of Medicine Solna, Unit of Infectious Diseases, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
36
|
Xu H, Wang X, Veazey RS. Simian Immunodeficiency Virus Infection and Mucosal Immunity. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00076-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
37
|
Bahamondes MV, Castro S, Marchi NM, Marcovici M, Andrade LA, Fernandes A, Bahamondes L. Human vaginal histology in long-term users of the injectable contraceptive depot-medroxyprogesterone acetate. Contraception 2014; 90:117-22. [DOI: 10.1016/j.contraception.2014.01.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 01/28/2014] [Accepted: 01/29/2014] [Indexed: 10/25/2022]
|
38
|
Physiologic doses of depot-medroxyprogesterone acetate do not increase acute plasma simian HIV viremia or mucosal virus shedding in pigtail macaques. AIDS 2014; 28:1431-9. [PMID: 24759208 DOI: 10.1097/qad.0000000000000294] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Epidemiologic studies remain inconclusive on whether the injectable contraceptive depot-medroxyprogesterone acetate (DMPA) increases mucosal HIV shedding and transmissibility. Nonhuman primate models may help to determine the effects of DMPA on acute HIV replication. DESIGN We defined a physiologic dose of DMPA in macaques and assessed the impact of DMPA on acute simian HIV (SHIV) replication. METHODS Pigtail macaques received 1-30 mg of DMPA intramuscularly followed by measurements of progesterone and medroxyprogesterone acetate (MPA). Vaginal epithelial thickness, number of cell layers and density of intraepithelial CD3 cells were measured. The effect of DMPA on SHIV viremia and genital virus shedding was investigated in six pigtail macaques infected during monthly treatment cycles with 3 mg DMPA. Six DMPA-untreated macaques were controls. RESULTS Plasma MPA concentrations directly correlated with changes in epithelial thickness (correlation = 0.84; P < 0.001) and density of intraepithelial CD3 cells (correlation = 0.41; P = 0.02). A 3 mg DMPA dose recapitulated plasma MPA concentrations and changes in vaginal epithelial thickness seen in women. DMPA-treated and untreated macaques showed similar peak plasma viremia and RNA area under the curve values over 12 weeks (P = 0.94), although treated macaques had higher odds of having virus being detected in plasma (odds ratio 6.6, P = 0.02). Rectal and vaginal virus shedding was similar between treated and untreated macaques (P = 0.72 and P = 0.53, respectively). CONCLUSION In this pigtail macaque model of DMPA and vaginal SHIV infection, we found little or no effect of DMPA on plasma viremia and mucosal virus shedding during acute infection. These results do not support a role of DMPA in increasing mucosal HIV shedding.
Collapse
|
39
|
Abstract
BACKGROUND Concern about a possible association of hormonal contraception with HIV acquisition has been raised by three types of evidence. Firstly, high-dose progestogen treatment greatly increases HIV acquisition in female non-human primates. Secondly, biological plausibility for a link between hormonal contraception anf HIV acquisition is provided by evidence of a hypo-oestrogenic state induced by progestogen contraception with vaginal mucosal thinning, and evidence of effects on the humoral and cellular immune systems. Thirdly, some but not other large observational studies have found an increase in HIV acquisition among women using hormonal contraception. OBJECTIVES To determine, from the best available evidence, the effect of hormonal contraception on HIV acquisition. SEARCH METHODS We used the Cochrane Fertility Regulation Group trials search strategy. SELECTION CRITERIA Published, unpublished and ongoing trials with random allocation, comparing hormonal with non-hormonal methods, other hormonal methods or no contraception in women at risk of HIV acquisition. DATA COLLECTION AND ANALYSIS Data will be extracted from eligible trials onto a data extraction sheet and analysed using routine Cochrane Collaboration methodology, MAIN RESULTS One ongoing randomised trial was identified, with no data available to date. AUTHORS' CONCLUSIONS There is currently no robust evidence from randomized trials on the possible effect of hormonal contraception on HIV acquisition. High quality trials in this area are needed to inform counselling of individual woman and public health policy.
Collapse
Affiliation(s)
- G Justus Hofmeyr
- University of the Witwatersrand, University of Fort Hare, Eastern Cape Department of HealthDepartment of Obstetrics and Gynaecology, East London Hospital ComplexFrere and Cecilia Makiwane HospitalsPrivate Bag X 9047East LondonEastern CapeSouth Africa5200
| | - Mandisa Singata
- University of the Witwatersrand/University of Fort Hare/East London Hospital complexEffective Care Research UnitEast LondonSouth Africa
| | - Jennifer Sneden
- University of CaliforniaGlobal Health Sciences50 Beale StreetSan FranciscoCaliforniaUSA94105
| | | |
Collapse
|
40
|
Venkatesh KK, Cu-Uvin S. Anatomic and Hormonal Changes in the Female Reproductive Tract Immune Environment during the Life Cycle: Implications for HIV/STI Prevention Research. Am J Reprod Immunol 2014; 71:495-504. [DOI: 10.1111/aji.12247] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 03/06/2014] [Indexed: 12/30/2022] Open
Affiliation(s)
- Kartik K. Venkatesh
- Department of Obstetrics and Gynecology; Brigham and Women's Hospital and Massachusetts General Hospital; Harvard Medical School; Boston MA USA
| | - Susan Cu-Uvin
- Department of Obstetrics and Gynecology; Alpert Medical School; Brown University; Providence RI USA
- Division of Infectious Diseases; Department of Medicine; Alpert Medical School; Brown University; Providence RI USA
| |
Collapse
|
41
|
McNicholl JM, Henning TC, Vishwanathan SA, Kersh EN. Non-human primate models of hormonal contraception and HIV. Am J Reprod Immunol 2014; 71:513-22. [PMID: 24716832 DOI: 10.1111/aji.12246] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 03/03/2014] [Indexed: 12/17/2022] Open
Abstract
PROBLEM Recent concerns that hormonal contraception (HC) may increase risk of HIV acquisition has led to keen interest in using non-human primates (NHP) to understand the underlying mechanism and the magnitude of the risk. This is, in part, because some experiments which would be difficult or logistically impossible in women are more easily conducted in NHP. METHOD OF STUDY NHP models of HIV can inform HIV acquisition and pathogenesis research and identify and evaluate biomedical preventions and treatments for HIV/AIDS. Widely used species include rhesus, pigtail, and cynomolgous macaques. RESULTS This paper reviews past, current and proposed NHP research around the intersection of HIV and HC. CONCLUSION NHP research may lead to the identification of hormonally regulated biomarkers that correlate with HIV-acquisition risk, to a ranking of existing or next-generation HC along an HIV-acquisition risk profile, and inform research around new biomedical preventions for HIV.
Collapse
Affiliation(s)
- Janet M McNicholl
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | | | |
Collapse
|
42
|
Mitchell CM, McLemore L, Westerberg K, Astronomo R, Smythe K, Gardella C, Mack M, Magaret A, Patton D, Agnew K, McElrath MJ, Hladik F, Eschenbach D. Long-term effect of depot medroxyprogesterone acetate on vaginal microbiota, epithelial thickness and HIV target cells. J Infect Dis 2014; 210:651-5. [PMID: 24652495 DOI: 10.1093/infdis/jiu176] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Depot medroxyprogesterone acetate (DMPA) has been linked to human immunodeficiency virus type 1 (HIV-1) acquisition. METHODS Vaginal microbiota of women using DMPA for up to 2 years were cultured. Mucosal immune cell populations were measured by immunohistological staining. RESULTS Over 12 months, the proportion with H2O2-positive lactobacilli decreased (n = 32; 53% vs 27%; P = .03). Median vaginal CD3(+) cells also decreased (n = 15; 355 vs 237 cells/mm(2); P = .03), as did CD3(+)CCR5(+) cells (195 vs 128 cells/mm(2); P = .04), HLA-DR(+) cells (130 vs 96 cells/mm(2); P = .27), and HLA-DR(+)CCR5(+) cells (18 vs 10 cells/mm(2); P = .33). CONCLUSIONS DMPA contraception does not increase vaginal mucosal CCR5(+) HIV target cells but does decrease CD3(+) T lymphocytes and vaginal H2O2-producing lactobacilli.
Collapse
Affiliation(s)
- Caroline M Mitchell
- Department of Obstetrics and Gynecology Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington
| | | | | | - Rena Astronomo
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington
| | - Kimberly Smythe
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington
| | | | - Matthias Mack
- Department of Internal Medicine II, University Hospital, Regensburg, Germany
| | - Amalia Magaret
- Department of Laboratory Medicine Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington
| | | | | | - M Juliana McElrath
- Department of Laboratory Medicine Department of Medicine, Division of Allergy and Infectious Diseases Department of Global Health, University of Washington Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington
| | - Florian Hladik
- Department of Obstetrics and Gynecology Department of Medicine, Division of Allergy and Infectious Diseases Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington
| | | |
Collapse
|
43
|
Murphy K, Irvin SC, Herold BC. Research gaps in defining the biological link between HIV risk and hormonal contraception. Am J Reprod Immunol 2014; 72:228-35. [PMID: 24548147 DOI: 10.1111/aji.12209] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 01/07/2014] [Indexed: 01/10/2023] Open
Abstract
Epidemiologic data suggest an association between depot medroxyprogesterone acetate (DMPA), a progesterone-based hormonal contraceptive, and increased risk of HIV acquisition and transmission. DMPA is highly effective and is among the most commonly used form of hormonal contraception in areas of high HIV prevalence. Thus, defining the biological mechanisms that contribute to the potential negative synergy between DMPA and HIV is key and may facilitate the identification of alternative contraceptive strategies. Proposed mechanisms include thinning or disruption of the cervicovaginal epithelial barrier, induction of mucosal inflammation, interference with innate and adaptive soluble and cellular immune responses, and/or alterations in the vaginal microbiome. DMPA may also indirectly increase the risk of HIV by promoting genital herpes or other sexually transmitted infections. However, there is a paucity of rigorous in vitro, animal model and clinical data to support these potential mechanisms highlighting the need for future research.
Collapse
Affiliation(s)
- Kerry Murphy
- Albert Einstein College of Medicine, Bronx, NY, USA
| | | | | |
Collapse
|
44
|
Hadzic SV, Wang X, Dufour J, Doyle L, Marx PA, Lackner AA, Paulsen DB, Veazey RS. Comparison of the vaginal environment of Macaca mulatta and Macaca nemestrina throughout the menstrual cycle. Am J Reprod Immunol 2014; 71:322-9. [PMID: 24521395 DOI: 10.1111/aji.12201] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 12/17/2013] [Indexed: 01/16/2023] Open
Abstract
PROBLEM Pigtail macaques, Macaca nemestrina (PT), are more susceptible to vaginal transmission of simian immunodeficiency virus (SIV) and other sexually transmitted diseases (STD) than rhesus macaques (RM). However, comparative studies to explore the reasons for these differences are lacking. METHOD OF STUDY Here, we compared differences in hormone levels and vaginal mucosal anatomy and thickness of RM and PT through different stages of the menstrual cycle. Concentrations of plasma estradiol (E2) and progesterone (P4) were determined weekly, and vaginal biopsies examined at days 0 and 14 of the menstrual cycle. RESULTS Consistent changes in vaginal epithelial thickness occurred at different stages of the menstrual cycle. In both species, the vaginal epithelium was significantly thicker in the follicular than in luteal phase. Keratinized epithelium was strikingly much more prominent in RM, especially during the luteal phase. Further, the vaginal epithelium was significantly thinner, and the P4:E2 ratio was higher in PT during luteal phase than RM. CONCLUSIONS Striking anatomic differences in the vaginal epithelium between rhesus and pigtail macaques combined with differences in P4:E2 ratio support the hypothesis that thinning and less keratinization of the vaginal epithelium may be involved in the greater susceptibility of pigtail macaques to vaginal transmission of SIV or other STD.
Collapse
Affiliation(s)
- Sarah V Hadzic
- Tulane National Primate Research Center, Covington, LA, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Recent advances in the immunology, pathogenesis, and prevention of human immunodeficiency virus (HIV) infection continue to reveal clues to the mechanisms involved in the progressive immunodeficiency attributed to infection, but more importantly have shed light on the correlates of immunity to infection and disease progression. HIV selectively infects, eliminates, and/or dysregulates several key cells of the human immune system, thwarting multiple arms of the host immune response, and inflicting severe damage to mucosal barriers, resulting in tissue infiltration of 'symbiotic' intestinal bacteria and viruses that essentially become opportunistic infections promoting systemic immune activation. This leads to activation and recruitment or more target cells for perpetuating HIV infection, resulting in persistent, high-level viral replication in lymphoid tissues, rapid evolution of resistant strains, and continued evasion of immune responses. However, vaccine studies and studies of spontaneous controllers are finally providing correlates of immunity from protection and disease progression, including virus-specific CD4(+) T-cell responses, binding anti-bodies, innate immune responses, and generation of antibodies with potent antibody-dependent cell-mediated cytotoxicity activity. Emerging correlates of immunity indicate that prevention of HIV infection may be possible through effective vaccine strategies that protect and stimulate key regulatory cells and immune responses in susceptible hosts. Furthermore, immune therapies specifically directed toward boosting specific aspects of the immune system may eventually lead to a cure for HIV-infected patients.
Collapse
Affiliation(s)
- Huanbin Xu
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433, USA
| | | | | |
Collapse
|
46
|
Thurman AR, Clark MR, Hurlburt JA, Doncel GF. Intravaginal rings as delivery systems for microbicides and multipurpose prevention technologies. Int J Womens Health 2013; 5:695-708. [PMID: 24174884 PMCID: PMC3808127 DOI: 10.2147/ijwh.s34030] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
There is a renewed interest in delivering pharmaceutical products via intravaginal rings (IVRs). IVRs are flexible torus-shaped drug delivery systems that can be easily inserted and removed by the woman and that provide both sustained and controlled drug release, lasting for several weeks to several months. In terms of women's health care products, it has been established that IVRs effectively deliver contraceptive steroids and steroids for the treatment of postmenopausal vaginal atrophy. A novel application for IVRs is the delivery of antiretroviral drugs for the prevention of human immunodeficiency virus (HIV) genital infection. Microbicides are antiviral drugs delivered topically for HIV prevention. Recent reviews of microbicide IVRs have focused on technologies in development and optimizing ring design. IVRs have several advantages, including the ability to deliver sustained drug doses for long periods of time while bypassing first pass metabolism in the gut. IVRs are discreet, woman-controlled, and do not require a trained provider for placement or fitting. Previous data support that women and their male sexual partners find IVRs highly acceptable. Multipurpose prevention technology (MPT) products provide protection against unintended/mistimed pregnancy and reproductive tract infections, including HIV. Several MPT IVRs are currently in development. Early clinical testing of new microbicide and MPT IVRs will require a focus on safety, pharmacokinetics and pharmacodynamics. Specifically, IVRs will have to deliver tissue concentrations of drugs that are pharmacodynamically active, do not cause mucosal alterations or inflammation, and do not change the resident microbiota. The emergence of resistance to antiretrovirals will need to be investigated. IVRs should not disrupt intercourse or have high rates of expulsion. Herein, we reviewed the microbicide and MPT IVRs currently in development, with a focus on the clinical aspects of IVR assessment and the challenges facing microbicide and MPT IVR product development, clinical testing, and implementation. The information in this review was drawn from PubMed searches and a recent microbicide/MPT product development workshop organized by CONRAD.
Collapse
Affiliation(s)
- Andrea Ries Thurman
- CONRAD, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Meredith R Clark
- CONRAD, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Jennifer A Hurlburt
- CONRAD, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Gustavo F Doncel
- CONRAD, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA, USA
| |
Collapse
|
47
|
The complexity of contraceptives: understanding their impact on genital immune cells and vaginal microbiota. AIDS 2013; 27 Suppl 1:S5-15. [PMID: 24088684 DOI: 10.1097/qad.0000000000000058] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
48
|
Hafner LM, Cunningham K, Beagley KW. Ovarian steroid hormones: effects on immune responses and Chlamydia trachomatis infections of the female genital tract. Mucosal Immunol 2013; 6:859-75. [PMID: 23860476 DOI: 10.1038/mi.2013.46] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2011] [Accepted: 06/04/2013] [Indexed: 02/04/2023]
Abstract
Female sex hormones are known to regulate the adaptive and innate immune functions of the female reproductive tract. This review aims to update our current knowledge of the effects of the sex hormones estradiol and progesterone in the female reproductive tract on innate immunity, antigen presentation, specific immune responses, antibody secretion, genital tract infections caused by Chlamydia trachomatis, and vaccine-induced immunity.
Collapse
Affiliation(s)
- L M Hafner
- Infectious Diseases Program, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Kelvin Grove, Queensland, Australia.
| | | | | |
Collapse
|
49
|
Vincent KL, Vargas G, Wei J, Bourne N, Motamedi M. Monitoring vaginal epithelial thickness changes noninvasively in sheep using optical coherence tomography. Am J Obstet Gynecol 2013; 208:282.e1-7. [PMID: 23333551 DOI: 10.1016/j.ajog.2013.01.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 11/21/2012] [Accepted: 01/14/2013] [Indexed: 11/26/2022]
Abstract
OBJECTIVE High-resolution optical coherence tomography can be used noninvasively to evaluate vaginal morphologic features, including epithelial thickness, to assess this protective barrier in transmission of sexually transmitted infections and to monitor tissue response to topical medications and hormonal fluctuations. We examined the use of optical coherence tomography to measure epithelial thickness noninvasively before and after topical treatment with a drug that causes epithelial thinning. STUDY DESIGN Twelve female sheep were treated with intravaginal placebo (n = 4) or nonoxynol-9 (n = 8). Vaginal optical coherence tomography images were obtained before and 24 hours after treatment. Four sheep in the nonoxynol-9 group were also examined on days 3 and 7. Vaginal biopsies were obtained on the last examination day. Epithelial thickness was measured in optical coherence tomography images and in hematoxylin and eosin-stained histologic sections from biopsies. Statistical analysis was performed using analyses of variance (significance P < .05). RESULTS Baseline optical coherence tomography epithelial thickness measurements were similar (85 ± 19 μm placebo, 78 ± 20 μm nonoxynol-9; P = .52). Epithelial thinning was significant after nonoxynol-9 (32 ± 22 μm) compared with placebo (80 ± 15 μm) 24 hours after treatment (P < .0001). In the 4 nonoxynol-9-treated sheep followed for 7 days, epithelial thickness returned to baseline by day 3, and increased significantly on day 7. Epithelial thickness measurements from histology were not significantly different than optical coherence tomography (P = .98 nonoxynol-9, P = .93 hydroxyethyl cellulose). CONCLUSION Drug-induced changes in the epithelium were clearly detectable using optical coherence tomography imaging. Optical coherence tomography and histology epithelial thickness measurements were similar, validating optical coherence tomography as a noninvasive method for epithelial thickness measurement, providing an important tool for quantitative and longitudinal monitoring of vaginal epithelial changes.
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Oral and topical pre-exposure prophylaxis (PrEP) with antiretroviral drugs are novel biomedical interventions recently found to prevent HIV transmission among high-risk populations. In this review, we outline lessons learned from animal studies and discuss next steps in preclinical PrEP research including the study of new PrEP modalities, pharmacologic correlates of protection, and biological factors that may modulate PrEP efficacy. RECENT FINDINGS Studies using macaque or humanized mice models of mucosal simian immunodeficiency virus (SIV), HIV, or simian/human immunodeficiency virus (SHIV) transmission have provided efficacy data against rectal and vaginal infection. A multitude of oral and topical PrEP regimens including drugs such as tenofovir (TFV), tenofovir disoproxil fumarate (TDF) and emtricitabine (FTC) were tested against either wild-type or drug-resistant viruses. These models have also helped define prophylactic windows of protection of nondaily dosing and are being used increasingly to study pharmacokinetic and pharmacodynamic relationships. SUMMARY As human data from PrEP trials validate animal models or help fine tune them, it is expected that these models will play increasingly important roles in PrEP development as the field extends into new drug classes and combinations, episodic dosing, and novel long-acting drug formulations. By providing both efficacy and pharmacologic information these models can define correlates and mechanisms of protection, inform dose selection, and advance the most promising PrEP candidates and dosing modalities.
Collapse
|