Hormonal contraception alters vaginal microbiota and cytokines in South African adolescents in a randomized trial

Christina Balle, Iyaloo N Konstantinus, Shameem Z Jaumdally, Enock Havyarimana, Katie Lennard, Rachel Esra, Shaun L Barnabas, Anna-Ursula Happel, Zoe Moodie, Katherine Gill, Tanya Pidwell, Ulas Karaoz, Eoin Brodie, Venessa Maseko, Hoyam Gamieldien, Steven E Bosinger, Landon Myer, Linda-Gail Bekker, Jo-Ann S Passmore, Heather B Jaspan, Christina Balle, Iyaloo N Konstantinus, Shameem Z Jaumdally, Enock Havyarimana, Katie Lennard, Rachel Esra, Shaun L Barnabas, Anna-Ursula Happel, Zoe Moodie, Katherine Gill, Tanya Pidwell, Ulas Karaoz, Eoin Brodie, Venessa Maseko, Hoyam Gamieldien, Steven E Bosinger, Landon Myer, Linda-Gail Bekker, Jo-Ann S Passmore, Heather B Jaspan

Abstract

Young women in sub-Saharan Africa are disproportionally affected by HIV infection and unintended pregnancies. However, hormonal contraceptive (HC) use may influence HIV risk through changes in genital tract microbiota and inflammatory cytokines. To investigate this, 130 HIV negative adolescent females aged 15-19 years were enrolled into a substudy of UChoose, an open-label randomized crossover study (NCT02404038), comparing acceptability and contraceptive product preference as a proxy for HIV prevention delivery methods. Participants were randomized to injectable norethisterone enanthate (Net-En), combined oral contraceptives (COC) or etonorgesterol/ethinyl estradiol combined contraceptive vaginal ring (CCVR) for 16 weeks, then crossed over to another HC for 16 weeks. Cervicovaginal samples were collected at baseline, crossover and exit for characterization of the microbiota and measurement of cytokine levels; primary endpoints were cervical T cell activation, vaginal microbial diversity and cytokine concentrations. Adolescents randomized to COCs had lower vaginal microbial diversity and relative abundance of HIV risk-associated taxa compared to Net-En or CCVR. Cervicovaginal inflammatory cytokine concentrations were significantly higher in adolescents randomized to CCVR compared to COC and Net-En. This suggests that COC use may induce an optimal vaginal ecosystem by decreasing bacterial diversity and inflammatory taxa, while CCVR use is associated with genital inflammation.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. Study overview and randomization.
Fig. 1. Study overview and randomization.
CONSORT diagram of the number of participants that completed each study visit and provided genital samples, and the distribution of participants on each of the hormonal contraceptive methods analyzed: combined oral contraceptives (COC), the Net-En injection, and the vaginally inserted combined contraceptive ring (CCVR) at crossover and exit. Note that at crossover, some participants could choose their next contraceptive method if assigned to CCVR as their first method. LTFU loss-to-follow-up.
Fig. 2. Vaginal community clusters in a…
Fig. 2. Vaginal community clusters in a South African adolescent cohort.
a Barplot depicting the relative abundance of the most abundant bacteria in the baseline samples identified by 16S rRNA microbiome profiling. Samples (n = 126) are grouped by community-state type (CST) established using soft k-means clustering with weighted UniFrac distances and ordered by the most abundant species in each CST (CST-I: L. crispatus, CST-III: L. iners, and CST-IV: G. vaginalis). Alpha diversity for each sample (Shannon Index) is depicted below the barplot. bd Change in CST from baseline to crossover within study arms. Alluvial plot showing the change in CST distribution from baseline to crossover for matched participants randomized to one of the three study arms: b COC (n = 36), c Net-En (n = 32), and d CCVR (n = 34) in an intention-to-treat (ITT) analysis. Each line represents one adolescent and CST changes over time. The color of the line is based on the CST assigned at baseline. eg Alpha diversity at baseline and crossover within study arms. Boxplot representing in alpha diversity (Shannon Index) of vaginal microbiota for matched participants at baseline and crossover randomized to one of the three study arms: e COC (n = 36), f Net-En (n = 32), and g CCVR (n = 34). hj Alpha diversity within participants changing between hormonal contraceptive methods. Boxplots showing the alpha diverisity (Shannon Index) in h 62 vaginal samples from 29 participants changing from COC to Net-En or vice versa, i 52 vaginal samples from 26 participants changing from COC to CCVR or vice versa, and j 130 vaginal samples from 50 participants changing from Net-En to CCVR or vice versa. P values were generated using two-sided paired Wilcoxon signed-rank tests. Y axes are log10-transformed. Significance codes: *P < 0.05. Bounds of boxes show interquartile range (IQR) with the lower and upper hinges corresponding to the 25th and 75th percentiles, respectively, lines in the middle of the box indicate median, and top and bottom whiskers demonstrate value ranges within 1.5 × IQR from the hinge. Points beyond that are plotted individually. CCVR: combined contraceptive vaginal ring, COC: combined oral contraceptives. Source data are provided as a Source Data file.
Fig. 3. Cytokine concentrations from baseline to…
Fig. 3. Cytokine concentrations from baseline to crossover according to hormonal contraceptive arm in an intent-to-treat analysis.
Boxplot showing the change in cytokine concentrations from baseline to crossover for matched participants within each of the three contraceptive arms: a adolescents randomized to COC (n = 36), b adolescents randomized to Net-En (n = 35), and c adolescents randomized to CCVR (n = 34). P values were generated using two-sided paired Wilcoxon signed-rank tests adjusted for multiple comparisons using the Benjamini–Hochberg (BH) method. Y axis log10-transformed. Significance codes: *P < 0.05, **P < 0.01, ***P < 0.001. Bounds of boxes show interquartile range (IQR) with the lower and upper hinges corresponding to the 25th and 75th percentiles, respectively, lines in the middle of the box indicate median, and top and bottom whiskers demonstrate the largest and lowest value within 1.5 × IQR from the hinge. Points beyond that are plotted individually. CCVR: combined contraceptive vaginal ring, COC: combined oral contraceptives. Source data are provided as a Source Data file.
Fig. 4. Effect of hormonal contraception on…
Fig. 4. Effect of hormonal contraception on bacterial taxa and cervicovaginal cytokines implicated in HIV risk.
a A multilevel sparse partial least-squares discriminant analysis (sPLSDA) of cytokines and bacterial taxa associated with HIV grouped before (baseline) and after (crossover) use of COC, Net-En, or CCVR, respectively. b Barplot showing the loadings of components 1 and 2 from the sPLSDA analysis. The absolute value is an indication of the importance of the bacteria or cytokine, while the sign indicates positive/negative correlations between the variables. CCVR: combined contraceptive vaginal ring, COC: combined oral contraceptives. Source data are provided as a Source Data file.

References

    1. Cowan F, Pettifor A. HIV in adolescents in sub-Saharan Africa. Curr. Opin. HIV AIDS. 2009;4:288–293. doi: 10.1097/COH.0b013e32832c7d10.
    1. UNAIDS. Ending AIDS: Progress Towards 90-90-90 Targets (2017).
    1. Sedgh G, et al. Abortion incidence between 1990 and 2014: global, regional, and subregional levels and trends. Lancet. 2016;388:258–267. doi: 10.1016/S0140-6736(16)30380-4.
    1. Shah IH, Ahman E. Unsafe abortion differentials in 2008 by age and developing country region: high burden among young women. Reprod. Health Matters. 2012;20:169–173. doi: 10.1016/S0968-8080(12)39598-0.
    1. Aitken RJ, et al. As the world grows: contraception in the 21st century. J. Clin. Investig. 2008;118:1330–1343. doi: 10.1172/JCI33873.
    1. Morrison CS, et al. Hormonal contraception and the risk of HIV acquisition: an individual participant data meta-analysis. PLoS Med. 2015;12:e1001778. doi: 10.1371/journal.pmed.1001778.
    1. Polis CB, et al. An updated systematic review of epidemiological evidence on hormonal contraceptive methods and HIV acquisition in women. Aids. 2016;30:2665–2683. doi: 10.1097/QAD.0000000000001228.
    1. Myer L, et al. Bacterial vaginosis and susceptibility to HIV infection in South African women: a nested case-control study. J. Infect. Dis. 2005;192:1372–1380. doi: 10.1086/462427.
    1. Gosmann C, et al. Lactobacillus-deficient cervicovaginal bacterial communities are associated with increased HIV acquisition in young South African women. Immunity. 2017;46:29–37. doi: 10.1016/j.immuni.2016.12.013.
    1. McClelland RS, 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;18:554–564. doi: 10.1016/S1473-3099(18)30058-6.
    1. Borgdorff H, et al. Lactobacillus-dominated cervicovaginal microbiota associated with reduced HIV/STI prevalence and genital HIV viral load in African women. ISME J. 2014;8:1781–1793. doi: 10.1038/ismej.2014.26.
    1. Brooks JP, et al. Effects of combined oral contraceptives, depot medroxyprogesterone acetate and the levonorgestrel-releasing intrauterine system on the vaginal microbiome. Contraception. 2017;95:405–413. doi: 10.1016/j.contraception.2016.11.006.
    1. van de Wijgert JHHM, Verwijs MC, Turner AN, Morrison CS. Hormonal contraception decreases bacterial vaginosis but oral contraception may increase candidiasis: implications for HIV transmission. AIDS. 2013;27:2141–2153. doi: 10.1097/QAD.0b013e32836290b6.
    1. Achilles, S. L. et al. Impact of contraceptive initiation on vaginal microbiota. Am. J. Obstet. Gynecol. 218, 622-e1 (2018).
    1. Vodstrcil LA, et al. Hormonal contraception is associated with a reduced risk of bacterial vaginosis: a systematic review and meta-analysis. PLoS ONE. 2013;8:e73055. doi: 10.1371/journal.pone.0073055.
    1. Morrison C, et al. Cervical inflammation and immunity associated with hormonal contraception, pregnancy, and HIV-1 seroconversion. J. Acquir. Immune Defic. Syndr. 2014;66:109–117. doi: 10.1097/QAI.0000000000000103.
    1. Deese J, et al. Injectable progestin-only contraception is associated with increased levels of pro-inflammatory cytokines in the female genital tract. Am. J. Reprod. Immunol. 2015;74:357–367. doi: 10.1111/aji.12415.
    1. Roxby AC, et al. Changes in vaginal microbiota and immune mediators in HIV-1-seronegative Kenyan women initiating depot medroxyprogesterone acetate. J. Acquir. Immune Defic. Syndr. 2016;71:359–366. doi: 10.1097/QAI.0000000000000866.
    1. Masson L, et al. Genital inflammation and the risk of HIV acquisition in women. Clin. Infect. Dis. 2015;61:260–269. doi: 10.1093/cid/civ298.
    1. Anahtar MN, et al. Cervicovaginal bacteria are a major modulator of host inflammatory responses in the female genital tract. Immunity. 2015;42:965–976. doi: 10.1016/j.immuni.2015.04.019.
    1. Lennard, K. et al. Microbial composition predicts genital tract inflammation and persistent bacterial vaginosis in South African adolescent females. Infect. Immun. 86, e00410–17 (2018).
    1. Farage M, Maibach H. Lifetime changes in the vulva and vagina. Arch. Gynecol. Obstet. 2006;273:195–202. doi: 10.1007/s00404-005-0079-x.
    1. Hillier SL. The vaginal microbial ecosystem and resistance to HIV. AIDS Res. Hum. Retroviruses. 1998;14(Suppl 1):S17–S21.
    1. Miller L, et al. Depomedroxyprogesterone-induced hypoestrogenism and changes in vaginal flora and epithelium. Obstet. Gynecol. 2000;96:431–439.
    1. Gill, K. et al. An open-label, randomized crossover study to evaluate the acceptability and preference for contraceptive options in female adolescents, 15–19 years of age in Cape Town, as a proxy for HIV prevention methods (UChoose). J. Int.AIDSSoc. 10.1002/jia2.25626 (2020).
    1. Maechler, M., Rousseeuw, P., Struyf, A., Hubert, M. & Hornik, K. Cluster analysis basics and extensions v2.0.6. (R Foundation for Statistical Computing, Vienna, Austria, 2017).
    1. Ravel J, et al. Vaginal microbiome of reproductive-age women. Proc. Natl Acad. Sci. USA. 2011;108(Suppl):4680–4687. doi: 10.1073/pnas.1002611107.
    1. Gajer P, et al. Temporal dynamics of the human vaginal microbiota. Sci. Transl. Med. 2012;4:132ra52. doi: 10.1126/scitranslmed.3003605.
    1. McKinnon LR, et al. Characterization of a human cervical CD4+ T cell subset coexpressing multiple markers of HIV susceptibility. J. Immunol. 2011;187:6032–6042. doi: 10.4049/jimmunol.1101836.
    1. Stieh DJ, et al. Th17 cells are preferentially infected very early after vaginal transmission of SIV in macaques. Cell Host Microbe. 2016;19:529–540. doi: 10.1016/j.chom.2016.03.005.
    1. Verstraelen H, et al. Longitudinal analysis of the vaginal microflora in pregnancy suggests that L. crispatus promotes the stability of the normal vaginal microflora and that L. gasseri and/or L. iners are more conducive to the occurrence of abnormal vaginal microflora. BMC Microbiol. 2009;9:116. doi: 10.1186/1471-2180-9-116.
    1. Santiago GL, dos S, et al. Longitudinal qPCR study of the dynamics of L. crispatus, L. iners, A. vaginae, (sialidase positive) G. vaginalis, and P. bivia in the vagina. PLoS ONE. 2012;7:e45281. doi: 10.1371/journal.pone.0045281.
    1. Borgdorff H, et al. The impact of hormonal contraception and pregnancy on sexually transmitted infections and on cervicovaginal microbiota in african sex workers. Sex. Transm. Dis. 2015;42:143–152. doi: 10.1097/OLQ.0000000000000245.
    1. Ross JA, Agwanda AT. Increased use of injectable contraception in sub-Saharan. Afr. Afr. J. Reprod. Health. 2012;16:68–80.
    1. Sibeko S, Baxter C, Yende N, Karim QA, Karim SSA. Contraceptive choices, pregnancy rates, and outcomes in a microbicide trial. Obstet. Gynecol. 2011;118:895–904. doi: 10.1097/AOG.0b013e31822be512.
    1. Ellen JM, et al. Impact of sexual networks on risk for gonorrhea and chlamydia among low-income urban African American adolescents. J. Pediatr. 2005;146:518–522. doi: 10.1016/j.jpeds.2004.11.023.
    1. Barnabas, S. L. et al. Converging epidemics of sexually transmitted infections and bacterial vaginosis in Southern African female adolescents at risk of HIV. Int. J.STD. AIDS 10.1177/0956462417740487 (2017).
    1. Noguchi LM, et al. Risk of HIV-1 acquisition among women who use different types of injectable progestin contraception in South Africa: a prospective cohort study. Lancet HIV. 2015;2:e279–e287. doi: 10.1016/S2352-3018(15)00058-2.
    1. Kuhl H. Pharmacology of estrogens and progestogens: influence of different routes of administration. Climacteric. 2005;8(Suppl 1):3–63. doi: 10.1080/13697130500148875.
    1. Stanczyk FZ, Hapgood JP, Winer S, Mishell DRJ. Progestogens used in postmenopausal hormone therapy: differences in their pharmacological properties, intracellular actions, and clinical effects. Endocr. Rev. 2013;34:171–208. doi: 10.1210/er.2012-1008.
    1. Africander D, Verhoog N, Hapgood JP. Molecular mechanisms of steroid receptor-mediated actions by synthetic progestins used in HRT and contraception. Steroids. 2011;76:636–652. doi: 10.1016/j.steroids.2011.03.001.
    1. Ahmed K, et al. 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;394:303–313. doi: 10.1016/S0140-6736(19)31288-7.
    1. Huang Y, et al. Effects of a one year reusable contraceptive vaginal ring on vaginal microflora and the risk of vaginal infection: an open-label prospective evaluation. PLoS ONE. 2015;10:e0134460. doi: 10.1371/journal.pone.0134460.
    1. Crucitti T, et al. Contraceptive rings promote vaginal lactobacilli in a high bacterial vaginosis prevalence population: a randomised, open-label longitudinal study in Rwandan women. PLoS ONE. 2018;13:e0201003. doi: 10.1371/journal.pone.0201003.
    1. Hardy L, et al. Association of vaginal dysbiosis and biofilm with contraceptive vaginal ring biomass in African women. PLoS ONE. 2017;12:e0178324. doi: 10.1371/journal.pone.0178324.
    1. Kestelyn E, et al. A randomised trial of a contraceptive vaginal ring in women at risk of HIV infection in Rwanda: safety of intermittent and continuous use. PLoS ONE. 2018;13:e0197572. doi: 10.1371/journal.pone.0197572.
    1. Camacho DP, et al. Vaginal yeast adherence to the combined contraceptive vaginal ring (CCVR) Contraception. 2007;76:439–443. doi: 10.1016/j.contraception.2007.07.012.
    1. Marrazzo J, et al. P369 Incidence of vaginal infections in kenyan women randomized to continuous or cyclic contraceptive ring (CVR) Sex. Transm. Infect. 2019;95:A186 LP–A186187.
    1. Baeten JM, et al. Use of a vaginal ring containing dapivirine for HIV-1 prevention in women. N. Engl. J. Med. 2016;375:2121–2132. doi: 10.1056/NEJMoa1506110.
    1. Zalanyi S, Landgren BM, Johannisson E. Pharmacokinetics, pharmacodynamic and endometrial effects of a single dose of 200 mg norethisterone enanthate. Contraception. 1984;30:225–237. doi: 10.1016/0010-7824(84)90086-6.
    1. Colwell HH, et al. The ORTHO BC-SAT—a satisfaction questionnaire for women using hormonal contraceptives. Qual. Life Res. 2006;15:1621–1631. doi: 10.1007/s11136-006-0026-8.
    1. Konstantinus, I. N. et al. Impact of hormonal contraceptives on cervical Th17 phenotype and function in adolescents: results from a randomized cross-over study comparing long-acting injectable norethisterone oenanthate (NET-EN), combined oral contraceptive pills, and combined cont. Clin. Infect. Dis.10.1093/cid/ciz1063 (2019).
    1. Jaspan HB, et al. Immune activation in the female genital tract during HIV infection predicts mucosal CD4 depletion and HIV shedding. J. Infect. Dis. 2011;204:1550–1556. doi: 10.1093/infdis/jir591.
    1. Dabee S, et al. Defining characteristics of genital health in South African adolescent girls and young women at high risk for HIV infection. PLoS ONE. 2019;14:e0213975. doi: 10.1371/journal.pone.0213975.
    1. Lewis DA, et al. Prevalence and associations of genital ulcer and urethral pathogens in men presenting with genital ulcer syndrome to primary health care clinics in South Africa. Sex. Transm. Dis. 2012;39:880–885. doi: 10.1097/OLQ.0b013e318269cf90.
    1. Pearce MM, et al. The female urinary microbiome: a comparison of women with and without urgency urinary incontinence. mBio. 2014;5:e01283–14. doi: 10.1128/mBio.01283-14.
    1. Edgar RC. Search and clustering orders of magnitude faster than BLAST. Bioinformatics. 2010;26:2460–2461. doi: 10.1093/bioinformatics/btq461.
    1. Edgar RC, Haas BJ, Clemente JC, Quince C, Knight R. UCHIME improves sensitivity and speed of chimera detection. Bioinformatics. 2011;27:2194–2200. doi: 10.1093/bioinformatics/btr381.
    1. Caporaso JG, et al. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods. 2010;7:335–336. doi: 10.1038/nmeth.f.303.
    1. Rognes T, Flouri T, Nichols B, Quince C, Mahe F. VSEARCH: a versatile open source tool for metagenomics. PeerJ. 2016;4:e2584. doi: 10.7717/peerj.2584.
    1. Fettweis JM, et al. Species-level classification of the vaginal microbiome. BMC Genomics. 2012;13(Suppl 8):S17. doi: 10.1186/1471-2164-13-S8-S17.
    1. R Core Team. R: A Language and Environment for Statistical Computing (R Foundation for Statistical Computing, Vienna, Austria, 2016).
    1. McMurdie PJ, Holmes S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS ONE. 2013;8:e61217. doi: 10.1371/journal.pone.0061217.
    1. Oksanen, J. et al. vegan: community ecology package (R Foundation for Statistical Computing, Vienna, Austria, 2016).
    1. Tingley D, Yamamoto T, Hirose K, Keele L, Imai K. Mediation: R package for causal mediation analysis. J. Stat. Softw. 2014;59:1–38. doi: 10.18637/jss.v059.i05.
    1. Mangiafico, S. rcompanion: Functions to support extension education program evaluation (R Foundation for Statistical Computing, Vienna, Austria, 2015).
    1. Love MI, et al. Moderated estimation of fold change and dispersion for RNA-Seq data with DESeq2 Moderated estimation of fold change and dispersion for RNA-Seq data with DESeq2. Genome Biol. 2014;15:521–550. doi: 10.1186/s13059-014-0550-8.
    1. Rohart F, Gautier B, Singh A, Le Cao K-A. mixOmics: an R package for’omics feature selection and multiple data integration. PLoS Comput. Biol. 2017;13:e1005752. doi: 10.1371/journal.pcbi.1005752.

Source: PubMed

Подписаться