Zim CHIC: A cohort study of immune changes in the female genital tract associated with initiation and use of contraceptives

Sharon L Achilles, Leslie A Meyn, Felix G Mhlanga, Allen T Matubu, Kevin A Stoner, May A Beamer, Zvavahera M Chirenje, Sharon L Hillier, Sharon L Achilles, Leslie A Meyn, Felix G Mhlanga, Allen T Matubu, Kevin A Stoner, May A Beamer, Zvavahera M Chirenje, Sharon L Hillier

Abstract

Problem: Contraceptive hormones are systemically active, potent, and likely to invoke biological responses other than known fertility regulation impacts. We hypothesized that initiation of depot medroxyprogesterone acetate (DMPA) would increase genital HIV-target-cells and soluble immune mediators compared with baseline and initiation of other contraceptive methods.

Method of study: We collected cervical cytobrushes and cervicovaginal fluid from healthy Zimbabwean women aged 18-34 to assess immune cell populations, cytokines, and innate anti-HIV activity at baseline and after 30, 90, and 180 days use of DMPA (n = 38), norethisterone enanthate (n = 41), medroxyprogesterone acetate/estradiol cypionate (n = 36), levonorgestrel implant (n = 43), etonogestrel implant (n = 47), or copper intrauterine device (Cu-IUD) (n = 45). Cells were quantified by flow cytometry, cytokines were detected by multiplex assays, and innate anti-HIV activity was assessed by in vitro HIV challenge.

Results: Compared to baseline, the number of cervical HIV target cells (#CD4 cells P < .04 and #CD11c cells P < .04), the concentration of the inflammatory cytokine IL-1β (P < .01), and the innate in vitro anti-HIV activity (P < .001) significantly decreased following DMPA initiation. In Cu-IUD users, genital HIV target cells increased (#CD4 cells P < .001, #CD4CCR5 cells P = .02, #CD4CD69 cells P < .001, #CD8CD69 P = .01, and #CD11c cells P = .003) at day 30 and resolved by day 180. IFN-γ (P < .001), IL-1β (P < .001), IL-6 (P < .001), IL-8 (P < .001), IL-10 (P < .01), and RANTES (P < .001) were also significantly increased at day 30. Minimal alterations were observed following initiation of subdermal implantable contraceptives.

Conclusions: This head-to-head study compared six contraceptives and found increased HIV target cells and cervical inflammation temporally associated with Cu-IUD initiation. Use of hormonal contraception, including DMPA, did not increase cervical HIV target cells or inflammation. Clinical Trial Number: NCT02038335.

Keywords: CCR5; biomarkers; contraception; copper; medroxyprogesterone acetate; receptors.

Conflict of interest statement

The funder had no role in study design, data collection, data analysis, data interpretation, or manuscript writing. The corresponding author had full access to all study data and final responsibility for the decision to submit for publication. SLA has served as a consultant to Merck Sharp & Dohme Corp and has received research grants from Mithra and Evofem. SLH is a consultant for Merck, Pfizer, Lupin, Hologic, Pfizer, and Daré Bioscience, and receives research funding from Cepheid, Becton‐Dickinson, and Curatek. All other authors declare no competing interests.

© 2020 The Authors. American Journal of Reproductive Immunology published by John Wiley & Sons Ltd.

Figures

Figure 1
Figure 1
Diagram of participant flow from eligibility assessment to final categorization
Figure 2
Figure 2
Systemic progestin concentrations after initiation and use of injectable and implantable contraceptives. Serum progestin concentrations for all evaluable participants at day 30, 90, and 180 following initiation and continuous use of (A) DMPA, (B) Net‐En, (C) MPA/estradiol cypionate, (D) levonorgestrel implant, and (E) etonogestrel implant. Mean serum concentrations with SD are indicated with red bars and black brackets, respectively. DMPA, depot medroxyprogesterone acetate; Net‐En, norethisterone enanthate; MPA, medroxyprogesterone acetate; LNG, levonorgestrel; ENG, etonogestrel
Figure 3
Figure 3
Flow cytometric gating strategy and baseline blood and cervical CD4 cells expressing CCR5 and CD69. Recovery of CD3+CD4+CCR5+ and CD3+CD4+CD69+ cells from (A) cervical cytobrush, (B) peripheral blood mononuclear cells (PBMC), (C) CD3+CD4+CCR5CD69+ double positives from cervical cytobrushes and PBMC (D) the proportion of CD3+CD4+CCR5+ and CD3+CD4+CD69+ from PBMC and cervical cytobrush demonstrating gating. Two senior laboratory scientists trained in advanced flow cytometry and masked to contraceptive group, independently reviewed and agreed upon gating parameters for each sample. The percent (D) and number (E) of CD3+CD4+CCR5+ and CD3+CD4+CD69+ cells in PBMC and cervical cytobrush samples collected from all evaluable participants at baseline, prior to initiation of any contraception. Mean values with SD are indicated with red bars and brackets, respectively
Figure 4
Figure 4
Impact of injectable contraceptives on genital immune cells and soluble mediators. Cervical immune cells (A, C, and E) collected by endocervical cytobrush and quantified by flow cytometry are expressed as % change in cell number from baseline to follow‐up at days 30 and 180 after initiation of depot medroxyprogesterone acetate (DMPA), norethisterone enanthate (Net‐En), or medroxyprogesterone acetate and estradiol cypionate (MPA/EC) as indicated. Soluble mediators and innate anti‐HIV activity (B, D, and F) measured in cervicovaginal lavage expressed as the % change from baseline to follow‐up at days 30 and 180 after injectable contraceptive initiation. P‐values from Wilcoxon signed‐rank test comparing baseline (prior to contraceptive initiation) to follow‐up 180 d after initiation and continuous use of contraceptive as indicated; P‐values adjusted using the Holm‐Bonferroni multiple test procedure
Figure 5
Figure 5
Impact of implantable contraceptives on genital immune cells and soluble mediators. Cervical immune cells (A and C) collected by endocervical cytobrush and quantified by flow cytometry are expressed as % change in cell number from baseline to follow‐up at days 30 and 180 after initiation of contraception with levonorgestrel implant (LNG‐I) or etonogestrel implant (ENG‐I) as indicated. Soluble mediators and innate anti‐HIV activity (B and D) measured in cervicovaginal lavage expressed as the % change from baseline to follow‐up at days 30 and 180 after implantable contraceptive initiation. P‐values from Wilcoxon signed‐rank test comparing baseline (prior to contraceptive initiation) to follow‐up 180 d after initiation and continuous use of contraceptive as indicated; P‐values adjusted using the Holm‐Bonferroni multiple test procedure
Figure 6
Figure 6
Impact of copper intrauterine device on genital immune cells and soluble mediators. Cervical immune cells (A) collected by endocervical cytobrush and quantified by flow cytometry are expressed as % change in cell number from baseline to follow‐up at days 30 and 180 after initiation of contraception with copper intrauterine device (Cu‐IUD). Soluble mediators and innate anti‐HIV activity (B) measured in cervicovaginal lavage expressed as the % change from baseline to follow‐up at days 30 and 180 after intrauterine contraceptive initiation. P‐values from Wilcoxon signed‐rank test comparing baseline (prior to contraceptive initiation) to follow‐up 180 d after initiation and continuous use of contraceptive as indicated; P‐values adjusted using the Holm‐Bonferroni multiple test procedure

References

    1. United Nations. Department of Economic and Social Affairs, Population Division . Trends in Contraceptive Use Worldwide 2015 (ST/ESA/SER.A/349). 2015.
    1. Birth Control Around the World: Mapping Methods of Contraception | Superdrug™ . . Accessed February 15, 2019
    1. Polis CB, Curtis KM, Hannaford PC, et al. An updated systematic review of epidemiological evidence on hormonal contraceptive methods and HIV acquisition in women. AIDS. 2016;30(17):2665‐2683.
    1. Evidence for Contraceptive Options and HIV Outcomes (ECHO) Trial Consortium . HIV incidence among women using intramuscular depot medroxyprogesterone acetate, a copper intrauterine device, or a levonorgestrel implant for contraception: a randomised, multicentre, open‐label trial. Lancet. 2019;394(10195):303‐313.
    1. Gosmann C, Anahtar MN, Handley SA, et al. Lactobacillus‐deficient cervicovaginal bacterial communities are associated with increased HIV acquisition in young South African Women. Immunity. 2017;46(1):29‐37.
    1. McClelland RS, Lingappa JR, Srinivasan S, et al. Evaluation of the association between the concentrations of key vaginal bacteria and the increased risk of HIV acquisition in African women from five cohorts: a nested case‐control study. Lancet Infect Dis. 2018;18(5):554‐564.
    1. Eastment MC, McClelland RS. Vaginal microbiota and susceptibility to HIV. AIDS. 2018;32(6):687‐698.
    1. Cohen CR, Lingappa JR, Baeten JM, et al. Bacterial vaginosis associated with increased risk of female‐to‐male HIV‐1 transmission: a prospective cohort analysis among African couples. PLoS Medicine. 2012;9(6):e1001251.
    1. Farcasanu M, Kwon DS. The influence of cervicovaginal microbiota on mucosal immunity and prophylaxis in the battle against HIV. Curr HIV/AIDS Rep. 2018;15(1):30‐38.
    1. Cohen CR, Moscicki A‐B, Scott ME, et al. Increased levels of immune activation in the genital tract of healthy young women from sub‐Saharan Africa. AIDS. 2010;24(13):2069‐2074.
    1. Masson L, Passmore J‐A, Liebenberg LJ, et al. Genital inflammation and the risk of HIV acquisition in women. Clin Infect Dis. 2015;61(2):260‐269.
    1. Kaul R, Prodger J, Joag V, et al. Inflammation and HIV transmission in sub‐saharan Africa. Curr HIV/AIDS Rep. 2015;12(2):216‐222.
    1. Schacker T, Little S, Connick E, et al. Productive infection of T cells in lymphoid tissues during primary and early human immunodeficiency virus infection. J Infect Dis. 2001;183(4):555‐562.
    1. Loré K, Smed‐Sörensen A, Vasudevan J, Mascola JR, Koup RA. Myeloid and plasmacytoid dendritic cells transfer HIV‐1 preferentially to antigen‐specific CD4+ T cells. J Exp Med. 2005;201(12):2023‐2033.
    1. Moore JP, Trkola A, Dragic T. Co‐receptors for HIV‐1 entry. Curr Opin Immunol. 1997;9(4):551‐562.
    1. Achilles SL, Mhlanga FG, Musara P, Poloyac SM, Chirenje ZM, Hillier SL. Misreporting of contraceptive hormone use in clinical research participants. Contraception. 2018;97(4):346‐353.
    1. Zhang J, Tang C, Oberly PJ, Minnigh MB, Achilles SL, Poloyac SM. A sensitive and robust UPLC–MS/MS method for quantitation of estrogens and progestogens in human serum. Contraception. 2019;99(4):244‐250.
    1. Achilles SL, Creinin MD, Stoner KA, Chen BA, Meyn L, Hillier SL. Changes in genital tract immune cell populations after initiation of intrauterine contraception. Am J Obstet Gynecol. 2014;211(5):489.e1‐489.e9.
    1. Study of Immune Cell Changes in the Genital Tract 2 Months After Initiation of an IUD for Contraception ‐ Full Text View ‐ . . Accessed September 12, 2018
    1. Achilles SL, Austin MN, Meyn LA, Mhlanga F, Chirenje ZM, Hillier SL. Impact of contraceptive initiation on vaginal microbiota. Am J Obstet Gynecol. 2018;218(6):622.e1‐622.e10.
    1. Strober W. Trypan blue exclusion test of cell viability. Curr Protoc Immunol. 2001;Appendix 3:Appendix 3B 10.1002/0471142735.ima03bs21
    1. Nugent RP, Krohn MA, Hillier SL. Reliability of diagnosing bacterial vaginosis is improved by a standardized method of gram stain interpretation. J Clin Microbiol. 1991;29(2):297‐301.
    1. Dezzutti CS, Hendrix CW, Marrazzo JM, et al. Performance of swabs, lavage, and diluents to quantify biomarkers of female genital tract soluble mucosal mediators. PLoS One. 2011;6(8):e23136.
    1. Holm S. A simple sequentially rejective multiple test procedure. Scand J Stat. 1979;6(2):65‐70.
    1. Li L, Zhou J, Wang W, et al. Effects of three long‐acting reversible contraceptive methods on HIV target cells in the human uterine cervix and peripheral blood. Reprod Biol Endocrinol. 2019;17.
    1. Lajoie J, Tjernlund A, Omollo K, et al. Increased cervical CD4+CCR5+ T cells among kenyan sex working women using depot medroxyprogesterone acetate. AIDS Res Hum Retroviruses. 2019;35(3):236‐246.
    1. Byrne EH, Anahtar MN, Cohen KE, et al. Association between injectable progestin‐only contraceptives and HIV acquisition and HIV target cell frequency in the female genital tract in South African women: a prospective cohort study. Lancet Infect Dis. 2016;16(4):441‐448.
    1. Smith‐McCune KK, Hilton JF, Shanmugasundaram U, et al. Effects of depot‐medroxyprogesterone acetate on the immune microenvironment of the human cervix and endometrium: implications for HIV susceptibility. Mucosal Immunol. 2017;10(5):1270‐1278.
    1. Mitchell CM, McLemore L, Westerberg K, et al. Long‐term effect of depot medroxyprogesterone acetate on vaginal microbiota, epithelial thickness and HIV target cells. J Infect Dis. 2014;210(4):651‐655.
    1. Sciaranghella G, Wang C, Hu H, et al. CCR5 expression levels in HIV‐uninfected women receiving hormonal contraception. J Infect Dis. 2015;212(9):1397‐1401.
    1. Levinson P, Kaul R, Kimani J, et al. Levels of innate immune factors in genital fluids: association of alpha defensins and LL‐37 with genital infections and increased HIV acquisition. AIDS. 2009;23(3):309‐317.
    1. Polis CB, Achilles SL, Hel Z, Hapgood JP. Is a lower‐dose, subcutaneous contraceptive injectable containing depot medroxyprogesterone acetate likely to impact women’s risk of HIV? Contraception. 2018;97(3):191‐197.
    1. Heffron R, Achilles SL, Dorflinger LJ, et al. Pharmacokinetic, biologic and epidemiologic differences in MPA‐ and NET‐based progestin‐only injectable contraceptives relative to the potential impact on HIV acquisition in women. Contraception. 2019;99(4):199‐204.

Source: PubMed

3
Subskrybuj